serotonin and neuroplasticity – investigated in vivo by

137
1 Serotonin and Neuroplasticity – investigated in vivo by Positron Emission Tomography and structural Magnetic Resonance Imaging Doctoral thesis at the Medical University of Vienna in the program Clinical Neurosciences for obtaining the academic degree Doctor of Medical Science submitted by Christoph Kraus, MD Supervision by Rupert Lanzenberger, Assoc. Prof. PD MD NEUROIMAGING LABs (NIL) - PET & MRI & EEG & Chemical Lab Department of Psychiatry and Psychotherapy Medical University of Vienna Waehringer Guertel 18-20, 1090 Vienna, Austria http://www.meduniwien.ac.at/neuroimaging/ Vienna, July 2015

Upload: others

Post on 01-Dec-2021

0 views

Category:

Documents


0 download

TRANSCRIPT

1

Serotonin and Neuroplasticity – investigated in vivo by

Positron Emission Tomography and structural

Magnetic Resonance Imaging

Doctoral thesis at the Medical University of Vienna

in the program Clinical Neurosciences

for obtaining the academic degree

Doctor of Medical Science

submitted by

Christoph Kraus, MD

Supervision by

Rupert Lanzenberger, Assoc. Prof. PD MD

NEUROIMAGING LABs (NIL) - PET & MRI & EEG & Chemical Lab

Department of Psychiatry and Psychotherapy

Medical University of Vienna

Waehringer Guertel 18-20, 1090 Vienna, Austria

http://www.meduniwien.ac.at/neuroimaging/

Vienna,

July 2015

2

I. Table of Contents

II. Declaration ................................................................................................................ 3

III. Abstract ..................................................................................................................... 4

IV. Kurzfassung .............................................................................................................. 5

V. List of Publicaitons .................................................................................................... 6

VI. Abbreviations ............................................................................................................. 7

VII. Acknowledgements and Project Funding ................................................................... 8

I. BACKGROUND ........................................................................................................... 9

1.1 General introduction .................................................................................................. 9

1.2 The role of neuroplasticity in health and disease ..................................................... 10

1.3 Mechanisms regulating neuroplasticity .................................................................... 11

1.4 Crosslinks between Serotonergic Neurotransmission and Neuroplasticity ............... 13

1.5 In Vivo Quantification of Brain Anatomy with MRI .................................................... 16

1.6 MRI quantification of Neuroplasticity: Voxel-based morphometry ............................ 18

1.7 In Vivo Quantification of Neuroplasticity in the Serotonergic System ....................... 20

1.8 PET radioligands for the Serotonin-1A receptor and Serotonin Transporter ............ 21

1.9 PET quantification: binding potential ....................................................................... 22

1.10 Neuroplasticity quantified with PET and MRI – previous results .............................. 25

1.11 Open questions ....................................................................................................... 27

II. AIMS of the THESIS ................................................................................................ 29

III. RESULTS ................................................................................................................. 30

3.1 First publication ....................................................................................................... 30

3.2 Second publication .................................................................................................. 54

3.3 Third publication ...................................................................................................... 82

IV. GENERAL DISCUSSION and RAISED QUESTIONS ............................................ 104

V. CONCLUSION and FUTURE PERSPECTIVES ..................................................... 107

VI. REFERENCES ....................................................................................................... 108

APPENDIX – CURRICULUM VITAE ...................................................................... 124

3

II. Declaration

This thesis was conducted at the NEUROIMAGING LABs (NIL) - PET & MRI & EEG &

Chemical Lab (http://www.meduniwien.ac.at/neuroimaging, head: Assoc. Prof. PD Dr. med.

Rupert Lanzenberger) at the Department of Psychiatry and Psychotherapy (head: O.Univ.

Prof. Dr. h.c.mult. Dr. med. Siegfried Kasper), Medical University of Vienna.

All PET measurements were performed at the Department of Biomedical Imaging und

Image-guided Therapy, Division of Nuclear Medicine, (http://www.radiopharmaceutical-

sciences.net, Leading co-investigator: Assoc.‐Prof. PD. Dr. Wolfgang Wadsak, PD. Dr.

Markus Mitterhauser), Medical University of Vienna.

All MRI measurements were performed at the MR Centre of Excellence in collaboration with

the Center for Medical Physics and Biomedical Engineering (co-investigator: Assoc. Prof. PD.

Dr. Christian Windischberger).

4

III. ABSTRACT

Neuroplasticity is defined as the sum of structural and functional neuronal adaptions in the

adult brain upon changes of external stimuli or the internal milieu. After neuroimaging

demonstrated restructuration in the brain associated to navigational expertise or learning to

juggle neuroplasticity has received significant scientific attention. Moreover, deficits in

neuroplasticity are thought to contribute to the pathogenesis of depression. Meanwhile,

neurobiological studies revealed many new insights into the mechanisms behind the

regulation of neuronal development, synaptogenesis or neurogenesis. Conclusive evidence

indicates a modulating influence of serotonin (5-HT) in triggering measurable morphological

changes of neuronal cells. While this notion emerged mainly from animal studies, there is a

lack of studies in humans on neuroplastic functions of 5-HT.

We therefore investigated the relationship between 5-HT and neuroplasticity in humans with

a multimodal neuroimaging approach using structural and functional high resolution MRI in a

combination with PET and imaging genetics in three consecutive studies. In a first study, we

demonstrate that binding potential of the 5-HT1A receptor, which is the main inhibitory

serotonergic receptor and which has been frequently linked with modulation of neuroplastic

processes, correlates with regional gray matter volumes (GMV) in distinctive brain regions.

Furthermore, we found a correlation between 5-HT1A autoreceptor binding in the dorsal raphe

nuclei (DRN), known to modulate the forebrain’s serotonergic tone, and cortical GMV. These

results indicate that 5-HT1A receptor densities in certain brain regions interrelate with the

volume of gray matter.

In the second study, we report a significant increase of gray matter after 10 days of oral

selective-serotonin reuptake inhibitor (SSRI) administration in the posterior cingulate cortex,

which is accompanied by an increase in functional neuronal connectivity in the same region.

Here, elevated synaptic 5-HT due to oral SSRI intake, which represent the most often

prescribed antidepressants, leads to dynamic alterations of brain structure and function as

measured by MRI in healthy humans.

In the third study we did not substantiate previously reported increases in 5-HT transporter

(SERT) or 5-HT1A bindings upon lifetime changes of brain-derived neurotrophic factor (BDNF)

function, as produced by a common single nucleotide polymorphism (SNP).

The work of this thesis provides ample evidence that many of serotonin’s neuroplastic effects,

which are highly active during the brain’s development, might be partly conserved throughout

lifetime. Under consideration that both deficits in neuroplasticity and serotonergic function are

important hypotheses in the etiopathogenesis of depression, this thesis offers solid

groundwork for approximating these pathomechanisms in future studies. Finally, the

relationship between 5-HT and neuroplasticity detailed in this work, could lead to further

insights in the brain’s ability to adapt itself.

5

IV. KURZFASSUNG

Nachdem Studien der bildgebenden Hirnforschung zeigten, dass die graue Substanz bei

Erwachsenen dynamischen Veränderungen unterliegt wurde der Neuroplastizität viel

wissenschaftliche Aufmerksamkeit zuteil. Außerdem stellt dysfunktionale Neuroplastizität

eine Haupthypothese der Neuropathogenese von Depression dar. Studien in Zellkulturen

oder Tiermodellen identifizierten zahlreiche Mechanismen der Neuroplastizität, welche

neuronale Entwicklung, Synaptogenese oder Neurogenese steuern können. Hier gibt es

deutliche Hinweise für einen Einfluss von Serotonin (5-Hydroxytryptamin, 5-HT) auf

morphologische Veränderungen von Neuronen. Während dies vor allem bei neuronalen

Netzwerken während der Embryonalphase und während der postnatalen Gehirnentwicklung

nachgewiesen werden konnte, fehlen Studien bei Erwachsenen.

Diese Arbeit setzte sich daher zum Ziel, das Verhältnis zwischen 5-HT und Neuroplastizität

in vivo bei Menschen mit struktureller und funktioneller Magnetresonanztomographie sowie

Positronen-Emissions-Tomographie (PET) und Genetik zu untersuchen. In der ersten Studie,

zeigen wir starke Korrelationen zwischen dem 5-HT1A Rezeptor, welcher neuroplastische

Aktivität vermitteln kann, und dem regionalen Volumen der grauen Substanz in bestimmten

Gehirnregionen. Wir fanden außerdem eine Korrelation zwischen 5-HT1A Autorezeptordichte

im dorsalen Raphe Nucleus, welcher die Aktivität des serotonergen Systems regelt, und

grauer Substanz in höheren kortikalen Regionen. Gemeinsam zeigen diese Ergebnisse,

dass die 5-HT1A Rezeptor Dichte mit mehr grauer Substanz einhergeht. In der zweiten Studie

finden wir starke Signalzunahmen grauer Substanz nach Einnahme eines selektiven

Serotonin-Wiederaufnahmehemmers, was wiederum mit einer Zunahme funktioneller

neuronaler Aktivität vergesellschaftet ist. Hierbei setzt eine vermehrte synaptische 5-HT

Konzentration molekulare Prozesse in Gang, die mit einer Umstrukturierung grauer Substanz

einhergehen. In der dritten Studie konnten wir zuvor berichtete Veränderungen des

Serotonintransporters oder 5-HT1A Rezeptors in Zusammenhang mit reduzierter

Verfügbarkeit des „brain-derived neurotrophic factors“ (BDNF) nicht bestätigen.

Diese Arbeit bietet wichtige Belege dafür, dass neuroplastische Effekte von Serotonin,

welche an der Entwicklung neuronaler Netzwerke beteiligt sind, auch in adulten humanen

Gehirnen aktiv sein könnten. Unter Berücksichtigung, dass sowohl Defizite im serotonergen

System als auch dysfunktionale Neuroplastizität zwei Haupthypothesen der

Neuroätiopathogenese der Depression darstellen, generiert diese Arbeit eine wichtige Basis

für eine Verbindung beider Hypothesen. Schließlich könnte der Zusammenhang zwischen

Serotonin und Neuroplastizität zu weiteren Einsichten über die Fähigkeit des Gehirns sich

selbst zu adaptieren führen.

6

V. LIST of PUBLICATIONS

Publications:

Kraus C, Hahn A, Savli M, Kranz GS, Baldinger P, Höflich A, Spindelegger C, Ungersböck J,

Häusler D, Mitterhauser M, Windischberger C, Wadsak W, Kasper S, Lanzenberger R.

Serotonin-1A receptor binding is positively associated gray matter volume – A multimodal

neuroimaging study combining PET and structural MRI. NeuroImage 2012 Nov

15;63(3):1091-1098. Epub 2012 Jul 23 [2014, IF: 6.35]

Kraus C, Ganger S, Losak J, Hahn A, Savli M, Kranz GS, Baldinger P, Windischberger C,

Kasper S, Lanzenberger R, Gray matter and intrinsic network changes in the posterior

cingulate cortex after selective serotonin reuptake inhibitor intake. NeuroImage 2014;

84:236-244. Epub 2013 Aug 26 [2014, IF: 6.35]

Kraus C, Baldinger P, Rami-Mark C, Gryglewsky G, Kranz GS, Haeusler D, Hahn A,

Wadsak W, Mitterhauser M, Rujescu D, Kasper S, Lanzenberger R. Exploring the impact of

BDNF Val66Met genotype on serotonin transporter and serotonin-1A receptor binding,

PLOS-One, 2014 Sep 4;9(9) [2014, IF: 3.23]

Related Publications:

Hahn A, Wadsak W, Windischberger C, Baldinger P, Höflich A, Losak J, Nics L, Philippe C,

Kranz GS, Kraus C, Mitterhauser M, Karanikas G, Kasper S, Lanzenberger R. Differential

modulation of self-referential processing in the default mode network via serotonin-1A

receptors. Proceedings of the National Academy of Sciences (PNAS) 2012 Feb

14;109(7):2619-24. [2014, IF 9.67]

Kranz GS, Hahn A, Baldinger P, Häusler D, Philippe C, Kaufmann U, Wadsak W, Savli M,

Höflich A, Kraus C, Vanicek T, Mitterhauser M, Kasper S, Lanzenberger R. Cerebral

serotonin transporter asymmetry in males and male-to-female transsexuals: a PET study

with [11C]DASB. Brain Structure and Function 2012 [2014, IF: 5.62]

Baldinger P, Hahn A, Mitterhauser M, Kranz G, Friedl M, Wadsak W, Kraus C, Ungersböck

J, Hartmann A, Giegling I, Rujescu D, Kasper S, Lanzenberger R. Impact of COMT genotype

on serotonin-1A receptor binding investigated with PET. Brain Structure and Function 2013.

Epub 2013 Aug 9. [2014, IF: 5.62]

7

VI. Abbreviations

5-HT – 5-hydroxytryptamine, serotonin

5-HT1A – serotonin-1A receptor

AMPA – α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor

BDNF – brain-derived neurotrophic factor

BOLD – blood oxygen level dependent

CNS – central nervous system

CREB – cyclic AMP response element-binding protein

CSF – cerebro spinal fluid

DARTEL – diffeomorphic anatomical registration using exponentiated lie algebra

ERK – extracellular-regulated kinase

FA – fractional anisotropy

GMV – gray matter volume

GRE – gradient echo sequences

NMDA – N-methyl-D-aspartate receptor

MAO – mono amino oxidase

MAPK – mitogen activated protein-kinase

MPRAGE – magnetization-prepared rapid acquisition of gradient echo

MRI – magnetic resonance imaging

PET – positron emission tomography

ROI – region of interest

SERT – serotonin transporter

SNP – single nucleotide polymorphism

SPM – statistical parametric mapping

STDP – spike-timing dependent plasticity

TE – echo time

TR – repetition time

VBM – voxel-based morphometry

8

VII. ACKNOWLEDGEMENTS and PROJECT FUNDING

I would like to express my dear thankfulness to my family and my friends. I could not exist

otherwise.

This work could not have been done without the direct participation and supportive energy of

young, motivated and funky researchers at the Neuroimaging Lab. With big support by it’s

fabulous members, this group was able to grow continuously as demonstrated by an

increasing number of scientific publications during the timespan this thesis was completed.

Therefore, I would like to express my gratitude to the “brain” behind this group, Rupert

Lanzenberger, Prof. MD, for supervision of this thesis. His friendly nature, scientific sharp-

mindedness and creative ideas were in concert a major stimulus for this work. Additionally, I

would like the thank the two members of the thesis committee Prof. Dr. Wolfgang Wadsak,

head of the Radiochemistry and Biomarker Development Unit at the Department of

Biomedical Imaging and Image-guided Therapy and especially Prof. Siegfried Kasper, MD,

head of the Department of Psychiatry and Psychotherapy, who is always eager support for

young scientists with his rich experience gained during his long and outstanding international

career.

Furthermore, I would like to thank the entire team of the Neuroimaging Lab, Dr. Andreas

Hahn for is support with computing, Dr. Marcus Savli for PET-modeling, as well as Dr. Pia

Baldinger, Dr. Anna Höflich, Dr. Georg Kranz and MSc. Sebastian Ganger for their clinical

and scientific support in a great variety of topics.

Regarding the hardships of simultaneous clinical training and scientific work, I would like to

use this chance to thank Prof. Dr. Richard Frey, Prof. Dr. Dietmar Winkler, Dr. Sandra Strnad

and Dr. Eva Resinger for supporting this scientific work and provide a series of releases of

duty to pursue the scientific studies invoked below and to attend international conferences

and scientific meetings.

9

I. BACKGROUND

1.1 General introduction

Neuroplasticity is defined as the sum of structural and functional neuronal changes in the

adult brain as a response to changes of external stimuli or the internal milieu (May, 2011).

Neuroplasticity mainly subsumes terms such as neurogenesis, synaptogenesis or dendritic

sprouting, while circulating synonyms for neuroplasticity, e.g., cytoarchitectural

restructuration or neuronal remodeling are a source of confusion.

During the past 20 years, neurobiological findings revealed remarkable new insights into the

underpinning mechanisms regulating neuroplasticity. This led to a reconsideration of the

traditional notion that the brain’s structural configuration is created during development from

early ages until early adulthood and remains stable during adulthood. Most prominently, it

was demonstrated that serotonin (5-hydroxytryptamine, 5-HT) is involved in memory

formation and learning by structural adaptation via second messenger proteins such as cyclic

AMP (Kandel, 2004; Kandel & Schwartz, 1982). Beyond this, dynamic neuronal

restructuration was recently demonstrated after motor learning, for example after learning to

juggle or musical training, but also upon enhanced experience of navigation, foreign

language learning and olfactory processing (Barkas et al, 2012; Delon-Martin et al, 2013;

Draganski et al, 2006; Hyde et al, 2009; Maguire et al, 2000; Martensson et al, 2012). These

studies were performed in humans with structural and functional neuroimaging and raised

criticism from many authors regarding the validity of these results (Fields, 2013; Thomas &

Baker, 2013). Researchers questioned, whether changes in neuroplastic MRI markers such

as gray matter volume (GMV) are caused by methodological shortcomings such as head

movement during scanning, realignment artifacts, and alterations in tissue water content or

regional cerebral blood flow changes other than neuronal activation. However, good

evidence in animal models shows significant neuronal remodeling by well-established

histological methods such as immunohistochemistry (Sagi et al, 2012; Tanti et al, 2012)

The molecular and cellular machinery behind neuroplasticity has been investigated with large

efforts and comprises numerous extracellular and intracellular proteins, signaling cascades,

transcription factors and genes, of which many are directly linked with 5-HT. But until now, it

remains unclear, which factors produce effects large enough to explain structural and

functional changes observed in human neuroimaging studies. To resolve open questions in

this regard, this thesis aims to further illuminate the cross-links between 5-HT and

10

neuroplasticity in humans. This was achieved by manipulation of 5-HT and measuring the

impact on neuroplasticity parameters obtained by functional and structural neuroimaging.

1.2 The role of neuroplasticity in health and disease

Neuroplasticity is a well-known key mechanism in learning and memory (Dayan & Cohen,

2011; Kandel, 2001), but besides altered neurotransmission, dysfunctional neuroplasticity is

a major pathogenetic factor in psychiatric disorders. In major depression two main etiologic

theories are well supported by a series of evidence, one of which is the so-called monoamine

hypothesis suggesting alterations in serotonergic and dopaminergic neurotransmission. The

second main etiologic theory is the “neuroplasticity hypothesis” indicating deficient neuronal

adaptation to external or internal stimuli as pathophysiologic factor. Both theories are

clinically relevant, because the mechanisms of action of current and potential future

treatments for psychiatric disorders are evaluated in this context.

The brain undergoes fundamental structural and functional changes from early ontology until

early adulthood, which is subsumed with the term developmental plasticity. Thereby, a

multiplicity of neurobiological changes drive physiologic transformations with far reaching

consequences for brain functionality and connectivity. During fetal brain development, early

precursor cells transform to neurons and migrate from the ventricular zone to form the

cerebral cortex. Dendrites and axons spread to establish a complex network of intercellular

communication via synapses (Rapoport & Gogtay, 2008). Interestingly, there is an initial

overproduction of neurons, which is followed by selective apoptosis leading to loss of about

50% of cortical neurons. During postnatal brain development, synaptic density increases

further to numbers above adult levels and further pruning happens during childhood and

adolescence to adult levels with primary sensory and motor regions maturing early in

comparison to more complex functions. Moreover, the brain needs stimulation by internal and

external environmental stimuli, similar to light stimulation for the developing eye (Rapoport &

Gogtay, 2008). Today it is known that the brain reaches ≈90% of its adult size around an age

of six, but changes constantly occur during adolescence (Brans et al, 2010). During the aging

process a consistent loss of brain tissue is observable (Good et al, 2001). While many of the

neurobiological mechanisms in control of neuroplastic changes during the brain’s

development seem to reduce their effects in adulthood, it remains unclear, which factors

remain significant and to what extent.

Malfunction of the molecular machinery regulating neuronal plasticity during developmental

plasticity may lead to malformations of cortical development, which are frequently associated

with mental retardation, epilepsy and congenital neurological deficits. The majority of these

11

disorders are now thought to share a genetic basis (Leventer et al, 2010), while many of the

underling mechanisms remain unclear. Furthermore, neurodevelopmental disorders such as

autism, fragile-X-syndrome, Down syndrome, motor disorders or schizophrenia (Lesch &

Waider, 2012) are associated with a distinctive serotonergic deficiency. Here, deficits in 5-HT

mediated synaptic signaling are thought to contribute to the pathophysiology and long-term

outcome of these patients.

In contrast to developmental plasticity the brain is able to adapt to internal or external

stressors or injury by compensatory neuroplasticity. Brain injury after stroke can result in a

series of well-studied events including intra-hemispheric changes in representational maps,

or inter-hemispheric balance shift whereby the uninjured hemisphere gains functions.

Furthermore, regional injury results in diffuse adaptive changes between functional network

nodes (Cramer et al, 2011). Similar forms of adaptive neuroplasticity have been described

during other forms of neuronal damage such as traumatic brain injury. In consideration of

age-related adaptive plasticity, it has been demonstrated that lesions in early age exhibit a

more efficient repair and children are able to handle trauma to brain areas of vision, motor,

auditory and language function considerably better than the adult CNS (Rapoport & Gogtay,

2008). Noticeably, adaptive neuroplasticity might not always have a positive impact on

clinical functioning. Neuronal reorganization after brain injury might lead to enhanced

disinhibition, a dysbalance between excitation and inhibition, even after months of the injury,

suggesting that the delayed onset is in relationship with slow axonal sprouting and the

formation of new neuronal connections. Maladaptive neuroplasticity is thought to cause

chronic pain and allodynia after amputation (Cramer et al, 2011).

1.3 Mechanisms regulating neuroplasticity

Ramon y Cayal and independently Sigmund Freud already postulated in 1894 that learning

might produce lasting changes in the effectiveness of synaptic connections (Kandel, 1981),

an idea that was not testable until decades later. Meanwhile several interdependent

mechanisms were found that regulate neuronal structure and function in neuroplastic

processes.

Neuroplasticity can be divided into large-scale morphological changes, such as axonal or

dendritic (neurite) sprouting or pruning and small-scale changes, such as synaptic formation

or pruning (Holtmaat et al, 2013). Due to methodological constraints, at present little is

known about longitudinal neurite and synaptic turnover in vivo in humans. From animal

studies it became clear that dendrite length and complexity are rather stable at low

magnification (Chow et al, 2009; Holtmaat & Svoboda, 2009; Kasai et al, 2010)

12

(Trachtenberg et al, 2002), while axonal length changes are more dynamic (De Paola et al,

2006). Most of the brain’s excitatory synapses are located on dendritic spines, which are

highly specialized, rapidly changing cellular substructures and often serve as proxies of

synapses. There are large, rather stable mushroom-like and small, dynamically changing

spines. During cortical development spine turnover is highest, but they remain under

homoeostatic control during lifetime, whereas stable spines stay in their form during lifetime

(Holtmaat et al, 2013). Short-term synaptic plasticity is essential to influence information

processing and high-pass or low-pass filtering of a synapse (Citri & Malenka, 2008)

according to initial transmitter release probability. Long-term spine plasticity is underlying

fundamental neurophysiologic processes such as long-term potentiation (LTP), long-term

depression (LTD) and spike-timing dependent plasticity (STDP), a subform of the first two

(Citri & Malenka, 2008).

Synaptic plasticity during aversive learning e.g. in fear conditioning located in brain areas

such as the hippocampus and the amygdala are thought to underlie LTP. Long-term

potentiation is considered an important factor for increasing synaptic strength. Repetitive,

simultaneous activation of excitatory synapses (Citri & Malenka, 2008; Jeffery & Reid, 1997)

induces a fast influx of Ca2+ into the postsynaptic cells. This is controlled by the excitatory

NMDA receptor of the glutamate neurotransmitter system and further modulated by AMPA

and metabotropic glutamate receptors. Long-term potentiation leads to an increased release

of neurotrophic factors, which enhance dendritic and synaptic strength (Malenka & Nicoll,

1999). Key players thereby are neurotrophins such as the brain derived neurotrophic factor

(BDNF) and it’s TrkB receptor.

The neurotrophin family consists of structurally related proteins, the nerve growth factor

(NGF), BDNF, neurotrophin 3 and neurotrophin 4. Each protein specifically binds at the TrkA,

TrkB, TrkC or the p75 receptors with receptor dimerization and structural modifications

enhancing specificity (Chao, 2003). Of all neurotrophins BDNF has gained most attention, as

it is associated with at least three intracellular signaling cascades not only directing synaptic

plasticity, but also cell survival and neuronal differentiation and neurite outgrowth (Black,

1999; Chao, 2003; Gentry et al, 2004). Due to the abundance of it’s functions BDNF, it’s

associated pathways and genes became one of the mostly investigated target structures in

neuropsychiatric research. Dysfunction of BDNF was linked with regional brain atrophy in

Alzheimer’s disease, neurodegenerative disorders as well as in depression [(Castrén, 2005;

Macqueen & Frodl, 2010)]. A polymorphism of the proBDNF’s promotor region consisting of a

valine to methionine substitution (val66met) in the codon 66 was found to cause significant

reductions in extracellular BDNF concentrations (Chen et al, 2004; Egan et al, 2003). This

13

polymorphism serves as a model of reduction of endogenous BDNF levels and has been

investigated in numerous neuropsychiatric studies.

Most of the above listed pathways are extracellular molecules circulating in the synaptic cleft

or membrane proteins. To fulfill the neuronal modifications upon cellular stimulation,

intracellular signaling cascades activate transcription factors that ultimately change protein

expression (McClung & Nestler, 2008). Many of the above mentioned structures lead to a

well-studied common transcription factor named cAMP response element-binding protein

(CREB), which transcriptional activity is fine-tuned by at least 30 other proteins. Major

second messenger proteins transducing receptor signals to CREB are protein kinases such

as the mitogen activated protein-kinase (MAPK). Further prominent transcription factors

regulating neuronal plasticity are located within the Fos family, including cFos, FosB, ΔFosB,

Fos-related antigen 1 (Fra1) and Fra2, which dimerize with Jun proteins (McClung & Nestler,

2008). Additionally, the rapid acting nuclear factor kappa-light-chain-enhancer of activated B

cells (NF-κB), plays an important role in synaptogenesis (Boersma et al, 2011). For LTP and

long-term neuroplastic changes, it was demonstrated that epigenetic modifications, histone

acetylation and DNA methylation, are required (Borrelli et al, 2008; McClung & Nestler,

2008). Post translational modifications e.g. by mRNA binding proteins such as

polyadenylation element-binding protein (CPEB) or mRNA regulation by micro RNAs

(miRNA) are intracellular mediators of neuroplasticity. Finally, it was demonstrated that a cell

cycle and differentiation regulating proteins of the S100 protein family, p11 is associated with

neurotransmitter transport, BDNF, neuroplasticity and 5-HT signaling (Warner-Schmidt et al,

2010).

1.4 Crosslinks between Serotonergic Neurotransmission

and Neuroplasticity

Besides histamine and the catecholamines adrenaline, dopamine and noradrenaline, 5-HT

belongs to the classical monoaminergic neurotransmitters. These are transmitters that are

built by one amino group, are connected to an aromatic ring by a two-carbon-chain (-CH2-

CH2-) and are synthesized form aromatic amino acids like phenylalanine, tyrosine or

tryptophan. The majority of the body’s serotonin is found in the enterochromaffine cells in the

digestive system where it controls gut movements. Furthermore, 5-HT is involved in bone

metabolism, vasoconstriction and exerts control in organ development. In the adult brain, 5-

HT is synthesized in the raphe nuclei of the midbrain and brainstem, from where serotonergic

neurons project to forebrain regions. Hence, the midbrain’s raphe are thought to possess a

major control function over the functionality of the serotonergic system. High 5-HT

concentrations were observed in the parahippocampus and hippocampus, amygdala,

14

cingulate cortex, temporal cortex as well as in the basal ganglia (Hornung, 2010). Hence, in

the brain 5-HT has multiple functions ranging from basal physiologic processes like appetite,

thermoregulation, sleep regulation, to “higher” functions such as emotion regulation,

impulsivity, reward processing (Akimova et al, 2009; Hoflich et al, 2012; Kranz et al, 2010;

Savli et al, 2012). The 5-HT system consists of at least 16 receptors and a serotonin

transporter (SERT) (Saulin et al, 2012).

A close interactive relationship between 5-HT and neuroplasticity is well established (Daubert

& Condron, 2010; Gaspar et al, 2003). Treatment with SSRIs is associated with changes in

the expression of BDNF (Koponen et al, 2005; Nestler et al, 2002). Increase of BDNF mRNA

in hippocampus and cortical brain regions have been reported following acute and chronic

administration of SSRIs (Kozisek et al, 2008; Nibuya et al, 1995). Research by E. Castren’s

group demonstrated that antidepressants activate BDNF mediated TrKB signaling and

subsequently CREB (Koponen et al, 2005; Rantamaki et al, 2007). Results from animal

(Karpova et al, 2011; Piubelli et al, 2011; Vetencourt et al, 2008; Vetencourt et al, 2011) or

human subjects (Nitsche et al, 2009; Serra-Millàs et al, 2011) conclusively suggest

enhancement of neuronal plasticity as a result of treatment with SSRIs. Furthermore, SSRIs

were demonstrated to improve motor recovery from ischemic stroke, which might arise from

these mechanisms (Chollet et al, 2011; Mead et al, 2012). Chronic administration of the SSRI

fluoxetine reinstates ocular dominance plasticity in adulthood and promotes the recovery of

visual functions in adult amblyopic animals (Vetencourt et al, 2008). These effects were

accompanied by increased expression of BDNF in the visual cortex (Vetencourt et al, 2011).

Serotonin and other monoamines are one the first neurotransmitters to emerge during

neuronal development (Rubenstein, 1998), where they first mediate autoregulatory effects in

growing serotonergic neurons (Whitaker-Azmitia, 1998), then catalyze the maturation of

astroglial cells (Whitaker-Azmitia, 1998) and finally influence target tissue maturation

(Whitaker-Azmitia et al, 1996). While transgenic mice completely lacking serotonergic

neurons exhibit high perinatal mortality rates and severe deficits in respiratory control

(Hodges et al, 2009), reversible inhibition of serotonin synthesis during early embryogenesis

(embryonic days (E) 12-17) results in long lasting alterations of cortical development (Vitalis

et al, 2007). Excess serotonin produces dystrophic serotonergic neurons (Daubert et al,

2010) as well as migration defects in retinal projection neurons (Upton et al, 1999),

thalamocortical axons (Vitalis et al, 2002) and cortical interneurons (Riccio et al, 2009).

Dystrophic serotonergic neurons were also reported in several neurodegenerative diseases

(Azmitia & Nixon, 2008) and serotonergic dysfunction is a characteristic of down syndrome

and autism (Whitaker-Azmitia, 2001).

15

Further investigations into the underlying molecular mechanisms revealed convergent

signaling pathways between serotonin and neuronal growth factors (Cowen, 2007; Polter &

Li, 2010). This evidence suggests that some serotonergic receptors, beyond their traditional

association with G proteins (all 5-HT receptors but the following) or ligand-gated ion channels

(5-HT3 receptor), are able to modulate the activity of signaling pathways involved in neuronal

plasticity such as extracellular-regulated kinase (ERK) and mitogen-activated protein kinase

(MAPK) (Cowen, 2007). Although knocking out SERT or MAO and genetic polymorphisms in

these enzymes impact on neuronal structure (Frodl et al, 2008; Karabeg et al, 2013; Singh et

al, 2013) effects seem to be less pronounced than manipulation of serotonergic receptors

(Benninghoff et al, 2012). The 5-HT1A, 5-HT1B, 5-HT1D, 5-HT2A, 5-HT2B, 5-HT2C, 5-HT4 and the

5-HT7 receptors are associated with ERK or Akt (proteinkinase B) in neuronal cells, which

indicates that intracellular serotonergic signals are involved in long-term cell protective

processes. The mechamism behind 5-HT’s neuroplastic properties are well examplified by

the 5-HT1A receptor. Starting from embryonic day (E) 12 in mouse embryos the 5-HT1A

receptor contributes to craniofacial development (Moiseiwitsch & Lauder, 1995) and peaks

from E14.5—E16.5 in the thalamus, hippocampus and the cortex (Bonnin et al, 2006). There

might be a time dependent expression pattern with peak expressions e.g. in the amygdala

throughout the developmental period and further peaks in regions maturing in postnatal

development (Bonnin et al, 2006; Mehta et al, 2007). Important neuromodulatory actions of

the 5-HT1A receptor are stimulation of neurogenesis and dentritic maturation in the

hippocampus (Yan et al, 1997) where neurogenesis in the dentate gyrus and the

subventricular zone remains life-long active (Gaspar et al, 2003). For example, treating mice

with a 5-HT1A agonist can reverse microencephaly induced by prenatal treatment with

cocaine (Akbari et al, 1994) and can reduce neuronal damage after ischemic stroke

(Mattson, 2008). Furthermore, 5-HT1A receptors on astroglial cells release a neurite

extension factor (S-100ß) and induce maturation of astrocytes (Azmitia, 2001). Astrocytic 5-

HT1A receptors in combination with S-100ß are responsible for maintenance of a mature

state in adult neurons (Azmitia, 1999; Wilson et al, 1998). Withdrawal of S-100ß leads to a

reduction of synaptic connections between neurons (Wilson et al, 1998) and goes along with

findings, that the 5-HT1A receptor is required for behavioral and neurogenic effects of the

selective serotonin reuptake inhibitor fluoxetine (Santarelli et al, 2003).

In summary, 5-HT is a neurotransmitter that exerts distinct neuromodulatory actions beyond

neurophysiological functions. Highly active in shaping the architecture of serotonergic

neurons during embryonic development and early postnatal neuronal maturation, this

neuroplastic role is partially conserved in specific brain regions throughout adulthood. Many

of these effects are mediated by the 5-HT1A receptor, through direct links to neuromodulatory

16

signaling pathways such as ERK and MAPK, neuronal cell maintenance by astrocytes or

linkages to neurotrophins such as BDNF.

1.5 In Vivo Quantification of Brain Anatomy with MRI -

Principles of Structural Magnetic Resonance Imaging

Magnetic resonance imaging (MRI) is a major clinical application of the nuclear magnetic

resonance phenomenon, which was discovered in 1939 by Isidor Rabi who described nuclei

in an electromagnetic field absorbing and re-emitting electromagnetic radiation. Thereby

every atomic isotope can be characterized by a specific resonance frequency depending on

the magnetic field strength. NMR allows identification of isotopes with an odd number of

protons and/or of neutrons by their unique magnetic moment and angular momentum or in

other words by a given, nonzero nuclear spin, whereas nuclides with even numbers of both

have a zero spin. Simply put, MRI is based on the interaction of a nuclear spin with an

external magnetic field (B0) (Haacke, 1999).

In more detail, MRI in humans relies on the manipulation of magnetic fields and the detection

of bulk precession of hydrogen spins in water, fat and organic tissue. While theoretically a

series of nuclei are detectable (e.g.: 2H, 6Li, 10B, 11B, 14N, 15N, 17O, 19F, 23Na, 29Si, 31P, 35Cl,

113Cd, 129Xe, 195Pt), mostly protons of 1H and 11C are used for MRI given their abundance in

organic material. Precession is defined as the change in the orientation of the rotational axis

of a rotating body. Nuclear spins are unique characteristics describing the circular movement

of a proton whereby it exhibits a loop of electric current around the axis about which it is

spinning (Haacke, 1999; Schneider & Fink, 2013). The electromagnetic current loop may

interact with external magnetic fields and is described by the magnetic dipole moment vector

𝜇 representing the spin axis and realigning, similar to a compass needle, along the force of

any external magnetic field, �⃗⃗�0. The process is influenced by the gyromagnetic ratio γ, which

is a constant unique to each particle given by the ratio of the magnetic dipole moment to its

angular momentum. E.g. the gyromagnetic ratio of a 1H proton has a value of 2.68×108

rad/s/Tesla (so that 𝛾 =γ

2π= 42.58 MHz/T). For a 7T magnetic field, the spins precess at a

radiofrequency of 7 × 42.58 = 289.06 MHz. This precession frequency is referred to as the

Larmor frequency and the relationship is expressed in the Larmor equation:

(1) ω0 = γ0B0

where ω0 is the Larmor frequency in megahertz (MHz), γ0 is the gyromagnetic ratio specific

to a particular nucleus and B0 is the strength of the magnetic field in Telsa (T).

17

To obtain a classical macroscopic MR-image, precession has to be generated by tipping the

magnetization vector, away from an external field. The magnetization is rotated away from its

longitudinal direction alignment with a radiofrequency (rf) pulse for a short time. The rf pulse

is a electromagnetic wave of the same frequency as the target nuclei’s’ Larmor frequency,

generated by a transmit coil. This process of energy absorption is known as excitation and

leads to longitudinal magnetization of the spin system. A pulse strong and long enough is

required to tip magnetization by exactly 90° (90° rf pulse) into a transverse plane. Nuclei are

hence rotated away from the z-axis towards the transverse plane (x,y-axis) perpendicular to

the direction of the main magnetic field, which produces an electrical current at the same

frequency as the Larmor frequency in a receiver coil. After the spin system is excited, the

magnetization vector starts to return to the state before excitation due to two independent

phenomena reducing transverse magnetization called spin-lattice interaction and spin-spin

interaction, which cause T1 relaxation and T2 relaxation. Longitudinal relaxation or T1

recovery describes the slow restoration of the spin vector to the z-axis, whereby the nuclei

emit their excess energy to their environment (i.e. lattice, hence spin-lattice). Transverse

relaxation subsumes an energy transfer between spins resulting from local changes in the

magnetic field. Spin-spin interactions are a consequence of differential durations of

dephasing between spins formerly in so-called phase coherence, which describes spins that

precess synchronously about the z-axis after excitation (T2 relaxation occurs at about 100-

300ms, T1 relaxation at about 0.5-5s, yet both processes are thought to occur independently)

(Weishaupt et al, 2006). Tissue contrast of brain MR-images depends on differential tissue

proton densities, whereby the CSF and fat exhibit highest densities followed by pathologies

such as a meningioma and gray matter. For delineation of T1-weighted tissue proton

densities, repetition time (TR) between iterative excitations is essential to MRI contrast, as

the number of newly excitable spins increases if more spins are already rotated back into the

z-plane. A long enough (over 1.5 s) TR ensures that differential relaxation times according to

tissue property are depicted. Tissue contrast in T2-weighted images is set by the echo time

(TE) of T2-weighted images, which is defined by the interval between deliverance of the

excitation pulse and collection of the MRI signal and in the range of only several hundred ms.

The digitized data are stored in a graphic matrix called k-space and afterwards Fourier

transformed. Signal-to-noise-ratio in MRI depends on multiple factors amongst them slice

thickness, field of view, number of acquisitions, scan parameters (TR, TE, flip angle), the

magnetic field strength or the selection of the transmit and receive coil (RF coil). For

structural MRI capable of in-depth neuroanatomical brain investigations mostly T1-weighted

gradient echo sequences (GRE) are applied, given their advantages in short imaging times

and hence less motion artifacts. Thereby, instead of pairs of RF pulses, GRE uses frequency

encoded gradients administered by gradient coils with alternating polarities. In functional MRI

18

(fMRI) typically T2*-weighted images are recorded, which are typically shorter than T2 and

obtain slightly more signal inhomogeneities at tissue borders or local magnetic fields (e.g. by

a high concentration of iron in several brain areas such as nucleus ruber). fMRI is based on

the effect that deoxygenated hemoglobin (Hb) exhibits shorter dephasing times than

oxygenated Hb. This blood oxygen level dependent (BOLD) contrast allows inference on

neuronal activity by a complex relationship expressed in the hemodynamic response

function.

1.6 MRI quantification of Neuroplasticity: Voxel-based

morphometry

Until recently neuroplasticity in humans could only be investigated by histologic samples

either obtained post mortem or during brain surgery. With improvements in neuroimaging

technologies beginning with computerized tomography scans demonstrating enlarged

ventricular size in schizophrenic patients (Weinberger et al, 1980; Weinberger et al, 1979),

early MRI-based manual brain volume measurements (Mathew & Partain, 1985) to

computational morphometry (Ashburner & Friston, 2000; Wright et al, 1995), in vivo brain

plasticity research has gained successively increased attention. High resolution or ultra-high

resolution structural MRI at field strengths of 3 respectively 7 Tesla and computerized

volumetric analyses are the current state-of-the-art techniques to quantify brain

microstructure at a minimal resolution around one mm3 (Lenglet et al, 2012; Lusebrink et al,

2013). This is achieved by a computational analysis of structural MRI data termed voxel-

based morphometry (VBM) (Ashburner & Friston, 2000) (a voxel is the cubic 3D analogue to

a pixel).

Voxel-based morphometry is a whole-brain automated technique for measuring regional

cerebral volume and tissue concentration differences in MRI. Simply put, it is the comparison

of local concentrations or volumes of gray matter between groups or within subjects

(Ashburner & Friston, 2000; Ashburner & Friston, 2009; Good et al, 2001). In more detail

VBM works with so-called mesoscopic anatomical differences in brain tissue density and

volume after discounting macroscopic differences, which are modeled after warping

individual brains to a common reference space (Ashburner & Friston, 2009). Although

criticized by some authors as susceptible to bias introduced by averaging individual brain

shapes to standardized brain templates (Bookstein, 2001) or lack of validity, because factors

such as head movement or cerebral blood flow may confound the MRI signal (Franklin et al,

2013), structural MRI exhibits an excellent test-retest reliability (Wonderlick et al, 2009) and

gray matter alterations observed with MRI were confirmed by post-mortem histologic analysis

(Vernon et al, 2011). Furthermore, VBM offers the advantage of fast calculations of regional

19

brain volumes, independent of anatomical delineation by manual region of interest (ROI)

drawing, which improves comparability of studies between study centers and improved

access to morphometric analysis, because detailed neuroanatomical and radiologic

knowledge is not a prerequisite.

Technically VBM is mostly performed with T1-weighted structural MRI images recorded with

inversion recovery gradient echo (GE) pulse sequences such as magnetization-prepared

rapid acquisition of GE (MPRAGE) (Mugler & Brookeman, 1990) or its advancement

MP2RAGE, which is thought to be more robust against field inhomogeneities at high static

magnetic fields (≥ 3 T) (Marques et al, 2010). Gradient echo images are designed to provide

the best contrast between gray and white matter with short acquisition times of only several

minutes. A good contrast between brain tissues is highly necessary to pre-process MRI data

in a standard VBM pipeline, which comprises segmentation in gray matter, white matter and

cerebrospinal fluid (CFS) images, registration or normalization of each individual’s brain

image to a standard stereotactic space, smoothing with an isotropic Gaussian kernel and

statistical analysis. The entire procedure is embedded in standard neuroimaging software

bundles implementing the general linear model such as statistical parametric mapping

(SPM). One of the currently available most accurate image segmentation and registration

algorithms is named DARTEL (“Diffeomorphic Anatomical Registration using Exponentiated

Lie algebra”) having significant advantages in tissue segmentation and normalization, due to

an increased number of realignment parameters (Ashburner, 2007; Klein et al, 2009). The

DARTEL-algorithm applies “flow fields” for each subject, which encode how the individual

images are warped or deformed to match best the average shape of a study specific,

iteratively improved template, and MRI data are so segmented into gray matter, white matter

and CSF images (Ashburner, 2009). In a further spatial transformation gray matter data,

which are mostly used for plasticity research, are matched to the canonical MNI (Montreal

Neurological Institute) brain, which is an internationally accepted standard template with a

coordinate system generated by averaging 152 subjects. To preserve the volume of tissue

from each voxel, gray matter data are modulated by Jacobian determinants encoding the

relative volumes of tissue before and after warping. This results in the main parameters in

neuroplasticity neuroimaging: GMV, a surrogate of the volume of gray matter in arbitrary units

and gray matter density (GMD) representing the density of tissue in a voxel. Both units are

usually smoothed by a Gaussian kernel between 8-12 mm to ensure normal distribution for

statistical processing. Now the ready pre-processed VBM data can fit a general linear model

(GLM) as implemented in SPM. Statistic analyses, regression models or longitudinal

analyses of variances (ANOVA) can be calculated with gray matter microstructure data at

around 1-3.5 mm3 resolution. This is the currently gold standard for in vivo assessment of

dynamic brain changes attributed to neuroplasticity.

20

1.7 In Vivo Quantification of Neuroplasticity in the

Serotonergic System with PET

Neurotransmitter release in the brain can be indirectly measured by a combination of

neuroreceptor quantification with radiotracers and positron emission tomography (PET) and

pharmacological challenges (Laruelle, 2000). The principles behind this approach are the

detection of changes in the availability of target receptors to radiotracer occupancy under

baseline and challenge conditions (Paterson et al, 2010). While this approach yielded highly

valuable results for the dopamine system (Laruelle, 2000) this did not yet translate to 5-HT

because only few currently available PET tracers (Selvaraj et al, 2012) were found to be

sensitive for 5-HT change, and these are still less potent as compared to measuring

endogenous dopamine release with [11C]raclopride or [11C]-(+)PHNO (Paterson et al, 2010).

Nevertheless, many important components of the 5-HT system in humans can be reliably

measured with PET and radioligands (Saulin et al, 2012).

A radioligand is composed by a radioactive isotope and a molecule binding selectively to a

specific target. The standard radioactive isotopes in use are 18Flour, 11Carbon 13Nitrogen or

15Oxygen with half-lives ranging from 2 minutes (15O) to almost 2 hours (18F). These emit a

positron, which reacts with an electron after travelling the positron range of about 1 mm. In

the process of positron annihilation two photons emerge exhibiting a charge of 511keV

spreading in opposite directions. The PET detector ring detects the two photons

simultaneously (coincidence), and stores the coincidence events as sinogram, from which

PET raw images are reconstructed (Turkheimer et al, 2014). The radioligand’s biochemical

properties determine the target structures at which the isotope should emit it’s signal. While

there are reversible and irreversible radioligands, in neuroreceptor and transporter mapping,

reversible tracer binding is preferred to mathematically model receptor binding (Laruelle et al,

2003). The chemical properties of the biological tracer to bind on brain structures must meet

several, occasionally contradictory demands making radioligand synthesis a quite

complicated task. First, it must be non-toxic, then it must be intravenously injectable in a

stable solution, exhibit a low peripheral metabolism, pass the blood-brain barrier and must

finally bind to the target structure with high selectivity and specificity. To yield proper binding,

the ligand should exhibit a sufficiently high affinity to the target protein depending on the

concentration of sites in the target region, which lies usually between 0.01 and 1 nM. The

lower the target density, the higher the required affinity (Innis et al, 2007; Laruelle et al,

2003), however a too high affinity will prolong scanning time, which impacts on decay

counting statistics for short-lived isotopes and patient-comfort. To pass the blood-brain barrier

lipophilicity is necessary, but a too high lipophilicity increases unspecific binding and

decreases the amount the free radioligand in the blood plasma. As far as metabolism

21

characteristics are concerned, no active metabolites crossing the blood-brain barrier are a

prerequisite. This multitude of necessary characteristics to synthetize a suitable radioligand

is the obvious reason why it is still a demanding task for radiochemists worldwide to provide

tailor made PET tracers.

During the PET neuroreceptor measurements, which vary dependent on the applied

radioligand between several minutes and about 2 hours, the subject’s head is placed parallel

to the orbitomeatal line guided by a laser beam in a polyurethane cushion. A standard PET

ring has a diameter of about 90 cm, where subjects are placed only with the head inside the

bore, hence claustrophobia is seldom an issue. Subjects are instructed not to talk, apart from

emergencies, and to minimize head and body movements. Subjects are intravenously

injected about 5 ml of the radioligand (using the bolus paradigm in most cases) in a stable

saline solution at the beginning of the PET measurement.

1.8 PET radioligands for the Serotonin-1A receptor and

Serotonin Transporter

For in vivo neuroplasticity research, the 5-HT1A receptor and the SERT are relevant

serotonergic structures, which both control neuroplastic processes and can be quantified with

excellent test-retest variability with PET radioligands. Several radioligands are commonly

used for PET and the 5-HT1A receptor: [3H]-WAY100635 (post-mortem), [carbonyl–11C]-

WAY100635, [18F]-FCWAY, [18F]-FPWAY, [18F]-MPPF, [11C]-CUMI-101 and various

derivatives (Billard et al, 2014), each one exhibiting advantages and disadvantages. One of

the most reliable compounds is [carbonyl–11C]-WAY100635, having excellent affinity,

specificity and selectivity (Martel et al, 2007) and was therefore selected for this work. It is

well synthesizable with simplified synthesis techniques (Rami-Mark et al, 2013; Wadsak W.,

2007). There are no relevant radioactive metabolites passing the blood-brain barrier which

leads to an excellent signal-to-noise-ratio (Pike, 2009; Wu et al, 2007).

Quantitative molecular imaging of the SERT was primarily focused on modification of SSRIs

based on their high affinity and specificity. This approach turned out to be not fruitful,

because in vitro measures proofed not to be translatable to in vivo performances (Huang et

al, 2010). In the following [128I]β-CIT was used in a number of SPECT-Studies, yet high non-

specific binding at the dopamine transporter (DAT) resulted in limited validity. Further

developments yielded radioligands with better specificity namely [11C]MADAM,

[11C]HOMADAM, [11C]DASB and [11C]AFM (Huang et al, 2010). Obtaining a high specific

binding, reversible high brain uptake and equilibrium within a short scanning time, [11C]DASB

emerged as most widely used PET ligand in studying the human SERT (Houle et al, 2000)

22

and was available for this work. Meanwhile a rapid automated preparation and purification of

[11C]DASB became available (Haeusler et al, 2009).

1.9 PET quantification: binding potential

To determine estimates of in vivo distribution of PET target proteins, PET raw data are

quantitatively analyzed by applying mathematical models. One of the most commonly used

outcome parameter is termed binding potential (BP) which is defined by the ratio of receptor

availability Bmax to radioligand dissociation constant KD at equilibrium, while KD is the

reciprocal of the affinity. The binding BP potential can be also viewed as the product of Bmax

and affinity (Innis et al, 2007):

(2) 𝐵𝑃 =𝐵max

𝐾D= 𝐵max

1

𝐾D= 𝐵max𝑎𝑓𝑓𝑖𝑛𝑖𝑡𝑦

The BP concept was originally defined by Mintun et al. (Mintun et al, 1984) and originates

from the Michaelis-Menten equation (Michaelis L., 1913),

(3) 𝐵 =𝐵max[𝑆]

𝐾D+[𝑆]

and was quickly incorporated in quantitative radioligand imaging analyses. B represents the

concentration of receptor bound ligand, Bmax the availability of receptors, S the concentration

of free substrate and KD is the dissociation constant. Here KD is equal to the substrate

concentration at which the reaction rate is half its maximal value (Berg et al, 2002). The

maximal rate Bmax is reached when catalytic sites on the enzyme are fully saturated with

substrate [S]. At very low substrate concentration [S] is much less than KD, yielding a rate

that is directly proportional to the substrate concentration:

(4) 𝐵 = 𝐵max

KD[𝑆]

In situations during radioligand imaging when [S] is very low, [S]≪KD, the Michaelis-Menten

equation (2) reduces to Mintun’s original definition of the binding potential (Bmax/KD) (1)

(Mintun et al, 1984) and equals the equilibrium ratio of specifically bound B ligand to its free

concentration [S].

(5) 𝐵𝑃 =𝐵max

𝐾D=

𝐵

[𝑆]

23

Of note, in vitro radioligand studies mostly use homogenized tissue where all receptors are

available to bind. Thereby, only one compartment is assumed and no distinction is made

between free concentrations in plasma or tissue. In contrast, proper modeling of in vivo

radioligand imaging in the brain demands two or three tissue compartment models with

additional compartments for non-specific binding. Additional compartments include the blood

plasma, and three compartments in the brain: a compartment of freely available ligand, a

non-specific and a specific bound compartment (Figure 1). In this model, the radioligand

binds to specific (the target structure) or non-specific targets (non wanted targets such as

other binding structures) in the brain after crossing the blood-brain barrier. The radioligand’s

free and the non-specific bound compartments are often combined, based on the notion that

the exchange between free and non-specific compartments is faster than between free and

specific compartments. This leads to a two-tissue model in the brain, instead of a three-

tissue compartment model, reduces model input and minimizes computation time. The

Figure 1: Compartment model assumptions commonly used for radioligand modeling. The ligand

enters the brain via the blood brain barrier. Here it is either freely available within the tissue,

specifically bound or non-specifically bound to target proteins. A three tissue compartment model is

reduced to a two-tissue compartment model by combining free and non-specific bound

compartments to a non-displaceable compartment model. The rate constants (k1-k6) describe

amount of ligand and time needed for the radioligand to transfer the compartments. Figure adapted

from (Slifstein et al, 2000).

24

assumptions contain a blood-brain coefficient of clearance into the brain (K1), a rate constant

of fractional clearance (k2) from the exchangeable pool of unbound radioligand in the brain

back to the systemic circulation and, most importantly, an association and a dissociation rate

constant towards and away from free ligand to specifically bound radioligands (k3, k4) and

two rate constants towards and away non-specific binding (k5, k6). To model BP either

volumes of distributions as in clinical pharmacology or rate constants as shown in Figure 1

can be estimated. In pharmacology the volume of distribution (V) reflects the relationship

between the amount of drug in the body at steady state and plasma drug concentration

(Hacker et al, 2009). In radioligand imaging V is the ratio of the concentration of radioligand

in a region of tissue to that in plasma (Innis et al, 2007). Depending on the kind of applied

rate constants (K1-k6) and concentrations in different volumes of distribution three BP models

are mostly used.

(6) 𝐵𝑃F = 𝐵avail 𝐾D⁄ = (𝑉T − 𝑉ND) 𝑓P⁄ =𝐾1𝑘3

𝑓p𝑘2𝑘4

BPF refers to the ratio at equilibrium of concentration of the specifically bound radioligand in

tissue to the concentration of free radioligand in plasma. VT and VND represent the volumes of

distribution for total radioligand in tissue and that of nondisplaceable tissue uptake. The free

fraction of plasma protein-bound radioligand 𝑓P needs to be measured by arterial blood

sampling, which itself underlies hardships (Parsey et al, 2000) such as arterial cannulation,

radioactive blood handling and quick high pressure liquid chromatography (HPLC). The in

vivo BPF is the most similar metric to the in vitro measurements of the relation of unoccupied

available receptors (Bavail) and KD, and many researchers argue that this reflects the most

accurate estimate of receptor distribution. But conditions of in vivo radioligand measurements

differ from in vitro measurements as far as temperature, multiple compartments, receptor

trafficking, phosphorylation state and competition with endogenous neurotransmitter are

concerned (Innis et al, 2007).

(7) 𝐵𝑃P = 𝑓P𝐵avail 𝐾D⁄ = 𝑉T − 𝑉ND =𝐾1𝑘3

𝑘2𝑘4

Another version of BP is BPP, which is similar to BPF but not corrected for 𝑓P, which provides

advantages if 𝑓P cannot be measured accurately or has a small difference between groups.

BPP is the ratio at equilibrium of specifically bound radioligand to that of total parent

radioligand in plasma. It equals VT - VND, yet without relation to 𝑓P, as well as the in vitro

analogue of the ratio between Bavail and KD corrected for 𝑓P . Both BPF and BPP can be

pronounced in mL∙cm-3.

(8) 𝐵𝑃ND = 𝑓ND𝐵avail 𝐾D⁄ = (𝑉T − 𝑉ND) 𝑉ND⁄ =𝑘3

𝑘4

25

Finally, the BPND refers to the ratio at equilibrium of specifically bound radioligand to that of

nondisplaceable radioligand in tissue. It uses a reference region with no target proteins for

non-specific binding, is methodically easier to implement and is computed in reference tissue

models (Gunn et al, 2001; Ichise et al, 2001; Logan et al, 1996). Hence, BPND is calculated

by brain data only and does not require the arterial input function, but it depends on the

assumption that nondisplaceable uptake is not influenced by between-group factors. For a

more in-depth mathematical discussion and consensus nomenclature the reader is referred

to an excellent review by Innis et al. (Innis et al, 2007).

1.10 Neuroplasticity quantified with PET and MRI –

previous results

In vivo studies applying structural MRI and VBM detected a wide array of internal and

external stimuli causing the brain to dynamically adapt itself, amongst them physiological

changes (Protopopescu et al, 2008), motoric training (Draganski et al, 2004; Draganski &

May, 2008; May, 2011; Pereira et al, 2007; Zatorre et al, 2012), playing instruments (Schlaug

et al, 2005) and pharmacological (Tardito et al, 2006; Vetencourt et al, 2008) and electro-

physiological interventions (May et al, 2006). Moreover, these techniques identified gray matter

changes intrinsic to a variety of brain pathologies. Beyond diseases where cerebral atrophy

is a known pathognomonic feature such as Alzheimer’s disease and other forms of dementia,

Huntington’s disease and other genetic disorders building-up toxic amounts of proteins in

neurons, inflammatory diseases such as multiple sclerosis or epilepsy, MRI and VBM

identified alterations in regional brain volumes in a number of psychiatric disorders such as

schizophrenia, depression, anxiety disorders, anorexia nervosa (Asami et al, 2012;

Johansen-Berg, 2012; van Tol et al, 2010). For example, patients with depression exhibit

smaller volumes of the basal ganglia, thalamus, hippocampus, frontal lobe, orbitofrontal

cortex and gyrus rectus while a smaller hippocampal volume is detectable in patients during

a depressive episode in comparison to patients during remission (Kempton et al, 2011).

Most importantly, to this work, the neurobiological correlates of changes in brain morphology

measured by MRI and VBM are completely unclear (Scholz et al, 2009; Tost et al, 2010).

Currently, there is a lack of exact models demonstrating which cellular compounds

correspond to the signal strength in one voxel to what extent. Many of these effects could be

mediated by serotonergic structures such as the 5-HT1A receptor, the SERT, neurotrophins

such as BDNF or reciprocal interaction with other neurotransmitters and neurotrophins.

There are a number of unknown variables, for example to what percentage brain vasculature

or cerebrospinal fluid contributes to the gray matter changes observed in VBM. Furthermore,

changes in proton density might alter proton-based MRI analyses. However, neuronal cell

26

bodies and glial cells are assumed to contribute mostly to gray matter visible on T1-weighted

MRI scans. Even at ultra high-resolution MRI and voxel edge lengths below 1 mm, there are

still ten thousands of interconnected neurons packed in one single voxel. Thus, more data

providing information at a molecular level and subsequently linking these with macroscopic

brain changes are necessary to determine what molecular processes gain large enough

effects to be detectable with structural MRI.

In an attempt to combine molecular with structural neuroimaging previous studies mainly

focused on interactions between brain glucose consumption and cerebral atrophy in

Alzheimer’s disease. Hereby, usually PET with the radioligand 18FDG (2-Fluor-2-desoxy-D-

glucose) and VBM are combined, to reveal cerebral hypometabolism and it’s association with

GMV alterations (Chételat et al, 2008; Ishii et al, 2005; Kanda et al, 2008). These studies

identified hypometabolism exceeding atrophy in many altered brain regions in Alzheimer’s

dementia, confirmed frontal and temporal lobe anomalies in frontotemporal dementia. A

similar approach demonstrated that anterior hippocampal formation volume and the posterior

cingulate glucose metabolism are at least altered in normal aging (Kalpouzos et al, 2009).

Several studies investigated associations between brain amyloid content as measured by

PET and the radioligand 11C-PiB (carbon-11-Pittsburgh compound B) and cerebral atrophy

(Jack et al, 2008; Oh et al, 2010; Villemagne et al, 2013). Furthermore, the relationship

between cerebral morphology and dopamine D2/D3 receptor distribution was shown by PET

and [18F]fallypride (Woodward et al, 2009). This body of evidence does not provide hints

towards molecular mechanisms behind dynamic gray matter alterations in healthy subjects or

gray matter atrophy in abovementioned psychiatric disorders. In that regard, two studies

aimed to closer investigate the relationship of molecular makers of neuronal density, GABAA

receptors labeled with 18F-flumazenil, and neuronal density as measured with MRI (Duncan

et al, 2013; la Fougère et al, 2010). The results did not find a linear correlation but rather

indicate a differential relationship between cortex thickness and cortical surface thickness

and neuronal density.

In two pioneering reviews Johansen-Berg and colleagues recently outlined basically four

potential biological mechanisms underlying dynamic gray matter alterations: neurogenesis,

gliogenesis, synaptogenesis and vascular changes (Johansen-Berg, 2012; Zatorre et al,

2012). As potential underlying molecular mechanisms the authors only refer to BDNF related

signaling. The authors finally argue that structural plasticity as measured with MRI should be

given a place in assessment of functional brain changes in learning and recovery and that

more research is urgently needed to identify molecular mechanisms leading to structural

rearrangement of the brain.

27

1.11 Open questions

While the previous sections could not detail all aspects of the entanglement of 5-HT in

neuroplasticity due to limited space, the introducing section provides a solid groundwork for

the work of this thesis. Serotonin beyond physiological functions as regulator of appetite,

thermal control, emotions, impulsivity and reward – just to name a few – is highly involved in

early embryonic and postnatal cytoarchitectural organization of the central nervous system.

The 5-HT1A, 5-HT1B, 5-HT1D, 5-HT2A, 5-HT2B, 5-HT2C, and 5-HT4 receptors are tightly linked to

signaling cascades involved in neuronal restructuration such as ERK, MAPK, and to

neurotrophic systems such as BDNF and their transcriptions factors (e.g., CREB) (Azmitia &

Nixon, 2008; Cowen, 2007; Polter & Li, 2010). An elaborated regulation of 5-HT in

neuroplasticity is further supported by animal models exhibiting dystrophic neurons upon

excessively elevating 5-HT (Daubert et al, 2010; Homberg, 2012). Additionally, treatment with

SSRIs, the most commonly described antidepressants, elevates 5-HT and subsequently

increases dendritic spine numbers (Hajszan et al, 2005), promotes neurogenesis (Mahar et

al, 2014; Pilar-Cuellar et al, 2013), interacts with synapse formation (Getz et al, 2011) and

enhances BDNF signaling (Pittenger & Duman, 2008; Rantamaki et al, 2007; Vetencourt et

al, 2011). The close interplay of 5-HT and neuroplasticity is further demonstrated by

pronounced serotonergic deficits in neurodevelopmental disorders like autism, fragile-X-

syndrome, Down syndrome, motor disorders or schizophrenia (Lesch & Waider, 2012). While

most findings arose from animal models, mostly as a consequence of limited invasiveness

into the human brain in vivo, only little is known about the mechanisms of neuroplasticity in

humans.

Meanwhile a series of studies applying structural magnetic resonance imaging and VBM

demonstrated dynamic in vivo gray matter alterations in adult brains. This was observed due

to a broad variety of internal and external stimuli such as navigation (Maguire et al, 2006),

language learning (Dorsaint-Pierre et al, 2006), musical expertise (Gaser & Schlaug, 2003),

rehabilitation (Sarkamo et al, 2014) but as well in neurological and psychiatric brain

conditions like multiple sclerosis (Eshaghi et al, 2014), depression or anxiety disorders (van

Tol et al, 2010). Explanatory mechanisms for these structural brain alterations are unclear.

Many of the neuroplastic properties of 5-HT might be conserved throughout adulthood and,

linked with in vivo neuroimaging experiments, provide testable hypothesis on the

mechanisms underlying dynamic gray matter changes in the adult brain. Although

information from structural MRI is frequently used for anatomical co-registration

(Henningsson et al, 2009; Lan et al, 2014) or bias-correction (Greve et al, 2014; Matuskey et

al, 2012) of PET data, until now, no study investigated the relationship between the

distribution of serotonergic receptors and the regional volume of gray matter. Additionally,

28

while there are numerous studies investigating the effects of elevated 5-HT related to SSRIs

on brain functionality as investigated with fMRI, there is a lack of data investigating the

influence of 5-HT challenge on gray matter. Finally, the link between 5-HT and BDNF is well

established in preclinical models, but rarely investigated in humans.

29

II. AIMS of the THESIS

Based on the open questions three main aims of this thesis were generated:

The first aim was to detail associations between a serotonergic receptor with neurotrophic

properties such as the 5-HT1A receptor as measured by PET with

[carbonyl−11C]WAY−100635 and regional volumes of gray matter as measured by structural

MRI. This question is treated in the first publication listed below.

A second aim of this thesis was to investigate dynamic alterations of gray matter after 5-HT

challenge with SSRIs in healthy adult subjects, which is the objective of the second

publication.

Finally, the third target was to investigate in more detail the relation between BDNF and

neurotrophic structures of the serotonergic system such as the 5-HT1A receptor and the

SERT. This was the subject of the third publication. The aims can be pointed out in detail as

follows:

To investigate the relationship between 5-HT1A heteroreceptor distribution in the

human brain measured with PET and the radioligand [carbonyl−11C]WAY−100635 and

the regional volume of gray matter measured with MRI and VBM.

To test the association of 5-HT1A autoreceptor binding in the dorsal raphe nuclei and

whole brain regional GMV.

To test the influence of elevated 5-HT as consequence of treatment with a widely

used SSRI (escitalopram) on gray matter.

To elucidate functional neuronal network changes after 5-HT challenge by

escitalopram with fMRI.

To test the impact of a functional polymorphism of BDNF and Val66Met on 5-HT1A

receptor distribution in healthy subjects and SERT binding in patients with major

depression.

30

III. RESULTS:

3.1 First publication: Serotonin-1A receptor binding is

positively associated with gray matter volume – A

multimodal neuroimaging study combining PET and

structural MRI

Christoph Krausa, Andreas Hahna, Markus Savlia, Georg S. Kranza, Pia Baldingera,

Anna Höflicha, Christoph Spindeleggera, Johanna Ungersboeckb, Daniela Haeuslerb,

Markus Mitterhauserb, Christian Windischbergerc, Wolfgang Wadsakb, Siegfried Kaspera,

Rupert Lanzenbergera*

a Department of Psychiatry and Psychotherapy,

b Department of Nuclear Medicine, PET Center,

c MR Center of Excellence, Center for Medical Physics and Biomedical Engineering,

Medical University of Vienna, Austria

Published in

NeuroImage, 2012 63(3):1091-1098,

[2014, IF: 6.35]

*Corresponding author:

A/Prof. Rupert Lanzenberger, MD

Department of Psychiatry and Psychotherapy

Functional, Molecular and Translational Neuroimaging - PET & MRI

Medical University of Vienna

Waehringer Guertel 18-20, 1090 Vienna, Austria

[email protected]

http://www.meduniwien.ac.at/neuroimaging

31

ABSTRACT

Animal models revealed that the serotonin-1A (5-HT1A) receptor modulates gray matter

structure. However, there is a lack of evidence showing the relationship between 5-HT1A

receptor concentration and gray matter in the human brain in vivo. Here, to demonstrate an

association between the 5-HT1A receptor binding potential, an index for receptor

concentration, and the local gray matter volume (GMV), an index for gray matter structure,

we measured 35 healthy subjects with both positron emission tomography (PET) and

structural magnetic resonance imaging (MRI). We found that regional heteroreceptor binding

was positively associated with GMV in distinctive brain regions such as the hippocampi and

the temporal cortices in both hemispheres (R2 values ranged from 0.308 to 0.503, p < 0.05

cluster-level FDR-corrected). Furthermore, autoreceptor binding in the midbrain raphe region

was positively associated with GMV in forebrain projection sites (R2 = 0.656, p = 0.001). We

also observed a broad range between 5-HT1A receptor binding and GMV. Given the

congruence of altered 5-HT1A receptor concentrations and GMV reduction in depression or

Alzheimer’s disease as reported by numerous studies, these results might provide new

insights towards understanding the mechanisms behind GMV alterations observed in these

brain disorders.

Key words: positron emission tomography, structural magnetic resonance imaging, 5-HT1A

receptor

32

INTRODUCTION

Growing evidence shows distinctive neuromodulatory properties of serotonin (5-

hydroxytryptamine, 5-HT) in developing and mature brain networks (Daubert and Condron,

2010; Gaspar et al., 2003). Early alterations in the 5-HT system are associated with life-long

changes in cognitive and behavioral functioning and the neuronal organization in

neuropsychiatric diseases (Gaspar et al., 2003). The 5-HT1A receptor, one of at least 16

receptors in the serotonergic system, is directly linked to signaling cascades mediating

neuroplasticity (Azmitia, 2001). Structural neuroimaging techniques revealed increased

amounts of gray matter volume (GMV) as surrogate for enhanced neuroplasticity in relation

to motoric training, cognitive performance or treatment with the antidepressant fluoxetine, a

selective serotonin reuptake inhibitor (Draganski et al., 2004; Kanai and Rees, 2011;

Vetencourt et al., 2008). On the other side, GMV loss as measured with high-resolution

structural magnetic resonance imaging (MRI), is a key feature of neuropsychiatric brain

disorders, whereby the hippocampal formation was demonstrated to be especially vulnerable

to volumetric alterations (Benninghoff et al., 2010; Geuze et al., 2005).

Serotonin-1A autoreceptors are located presynaptically on serotonergic neurons in the raphe

nuclei where they reduce tonic cell firing, thus autoinhibiting 5-HT release (Hall et al., 1997).

Postsynaptically, 5-HT1A heteroreceptors are expressed on glutamatergic and GABAergic

neurons and mediate an inhibitory serotonergic response (Amargós-Bosch et al., 2004; Hall

et al., 1997; Puig et al., 2005). Neurobiological studies identified a vast number of second

messenger pathways that exert neuroplastic changes (Citri and Malenka, 2008; Pittenger

and Duman, 2008) triggerd by 5-HT via 5-HT1A receptors (Azmitia, 2001; Tardito et al., 2006).

To sum up, 5-HT1A receptors might be involved in altering GMV, thereby offering a possible

explanation for gray matter atrophy observed in several brain disorders.

Dysfunctional neuronal organization is an important contributor to the pathogenesis of

Alzheimer’s disease (Mesulam, 1999), schizophrenia (Lewis and González-Burgos, 2008)

and depressive disorder (Pittenger and Duman, 2008), however the underlying molecular

mechanisms, leading to gray matter loss in these disorders are complex and not fully

understood. Interestingly, positron emission tomography (PET) studies demonstrated

alterations of 5-HT1A receptors in patients suffering from these disorders (Kepe et al., 2006),

(Mamo et al., 2007); (Kasper et al., 2002; Lanzenberger et al., 2007; Savitz et al., 2009). This

congruence and a lack of data in human brains in vivo lead us to investigate the relationship

between 5-HT1A receptor concentration and GMV with a multimodal neuroimaging approach.

33

MATERIAL AND METHODS

Participants

We examined 35 healthy adults, 18 males and 17 females (age range = 21-52, mean = 26.6

±6.8 years, Table 1), with at least general qualification for university entrance as lowest

educational level. All subjects were recruited via advertisement at the Medical University of

Vienna, Austria and underwent a general physical and neurological examination at the

screening visit including medical history, electrocardiogram and routine laboratory tests.

Inclusion criteria were age between 18 and 60, ability to perform study procedures and

absence of any acute or chronic disease. Exclusion criteria compromised any history of

severe disease, any psychiatric or neurologic disorder, previous drug abuse, pregnancy as

assessed by urine pregnancy tests and any continuous medication for three months prior to

the study. All participants provided written informed consent after written and oral

presentation of a general intelligible information form and received reimbursement after

participation. The institutional review board of the Medical University of Vienna, Austria, gave

approval to all study procedures. The pooled study sample consisted of subjects who were

part of positron emission tomography (PET) and magnetic resonance imaging (MRI) studies

previously published by our group (Hahn et al., 2010; Spindelegger et al., 2009).

Magnetic Resonance Imaging and Image Preprocessing

Structural magnetic resonance imaging was performed at the MR Center of Excellence at the

Medical University of Vienna, Austria, with a 3 Tesla whole-body MEDSPEC S300 MR-

scanner (Bruker BioSpin, Ettlingen, Germany) using a magnetization-prepared rapid gradient

echo (MPRAGE, T1-weighted) sequence (128 slices, 256 × 256 matrix, slice thickness 1.56

mm, voxel size 0.78 × 0.86 mm). To optimize image-preprocessing quality we used the

DARTEL (Diffeomorphic Anatomical Registration using Exponentiated Lie algebra) algorithm

(Ashburner, 2007), which ranked top in a comparison of 14 image registration algorithms

(Klein et al., 2009). The major advantage of the DARTEL algorithm is an increase in the

accuracy of inter-subject alignment by a high number of parameters derived from

deformation fields. T1-weighted images of all 35 subjects in our study were manually re-

oriented and segmented using the New Segment option in SPM8 (2009, Wellcome Trust

Centre for Neuroimaging, Institute of Neurology, University College London, London, United

Kingdom, http://www.fil.ion.ucl.ac.uk/spm/software/spm8/) to generate rigid-body aligned

gray matter, white matter and CSF-images. After segmentation all images were visually

checked for major artifacts. The DARTEL algorithm consecutively generates six individual

templates based on deformation fields calculated during segmentation, where the last

34

template produced (number 6) was used for normalization. Each individual’s segmented gray

matter image together with each deformation field and the template was normalized to

standard Montreal Neurological Institute (MNI) space at a voxel size of 1.5 × 1.5 × 1.5mm. To

correct for nonlinear spatial normalization, images were modulated by multiplication with the

Jacobian determinants of the deformation fields in order to preserve the actual amount gray

matter within each structure before normalization. Based on this, the modulated images are

further referred to as gray matter volume (GMV). The resultant values represent a

quantitative measure of gray matter tissue volume per unit volume of the spatially normalized

images (Ashburner and Friston, 2009). Finally, GMV images were smoothed with an 8-mm

full-width at half-maximum Gaussian kernel. Such smoothing is considered sufficient to

increase the stability of segmented images with respect to small registration errors.

Radiochemistry

The 5-HT1A receptor specific radioligand [carbonyl-11C]WAY-100635 was prepared at the

Cyclotron Unit of the PET center at the Department of Nuclear Medicine of the Medical

University of Vienna, Austria according to the optimized synthesis instruction proposed by

Wadsak et al. (2007). [Carbonyl-11C]WAY-100635 was prepared in a multistep radiosynthesis

starting from cyclotron-produced [11C]CO2 and purified by high-performance liquid

chromatography and solid-phase extraction. [carbonyl-11C]WAY-100635 was dissolved in a

phosphate-buffered saline solution and injected at a target dose of 5.4 MBq/kg bodyweight,

further details of radiochemical variables are given in Table 1.

Positron Emission Tomography (PET) Measurements

PET was performed at the Department of Nuclear Medicine of the Medical University of

Vienna, Austria with a GE advance full-ring scanner (General Electric Medical Systems,

Milwaukee, WI). Each subject’s head was placed in the scanner parallel to the orbitomeatal

line guided by a laser beam system to ensure full coverage of the neocortex and the

cerebellum in the field of view (FOV). A polyurethane cushion and head straps kept the head

in position to minimize head movement and to guarantee a soft head rest during the whole

scanning period. Initially, a 5-minute transmission scan in two-dimensional mode was

conducted to correct for tissue attenuation with a retractable 68Ge ring source. Dynamic PET

scans started simultaneously with the intravenous bolus injection of the radioligand

[carbonyl-11C]WAY-100635. PET scans lasted for 90 minutes per subject and were acquired

in a three-dimensional mode. The overall dynamic scan time was divided in 30 successive

35

time frames (15 × 1 minute, 15 × 5 minutes). The emission data were scatter- and

attenuation corrected based on the data from the transmission scans and reconstructed

using an iterative filtered back-projection algorithm (FORE + ITER). The final spatial

resolution of the reconstructed volume was 4.36 mm full-width at half maximum at the center

of the FOV. We did not perform realignment for head movement upon visual inspection of

PET-data quality. All 30 dynamic PET image frames were summed (PETADD) for co-

registration to the MRI.

Quantification of 5-HT1A Receptor Binding Potential

We assessed in vivo receptor density as indexed by 5-HT1A receptor binding potential (BPND),

the ratio at equilibrium of specifically bound radioligand to that of nondisplaceable radioligand

in tissue (Innis et al., 2007). Binding was computed using the voxel-wise modeling tool in the

PMOD software package (v3.1, 2009, for Linux, PMOD Technologies, Zurich, Switzerland,

http://www.pmod.com) and applying the two-parameter linearized reference tissue model

(MRTM2) (Ichise et al., 2003). Compared to other models such as the simplified reference

tissue model (SRTM), MRTM2 leads to lower BPND bias and hence a better signal-to-noise-

ratio, especially for whole-brain voxel-by-voxel analysis. We modeled 5-HT1A BPND as

previously described by our group using the insula and the cerebellum taken from an

automated anatomical labeling-based (AAL) region of interest (ROI) (Tzourio-Mazoyer et al.,

2002) atlas, as receptor-rich and receptor-poor region, respectively. The cerebellum

excluding cerebellar vermis served as reference region. This was done for the voxel-by-voxel

analysis as well as the ROI-based multimodal analysis.

Image Co-Registration for Multimodal Data Analysis

We combined the advantages of PET in quantifying receptors at the molecular level with

structural MRI, which provides data on brain structure such as cortical folding, regional

cortical thickness and volume or gray/white matter contrast. This was achieved by co-

registration of each individual’s PET image to the corresponding structural MRI image. We

used SPM8 to apply the transformation matrix of the structural scans obtained during

normalization to the PETADD images. As the structural scans were already normalized to

standard MNI space, this step also brought the PET data to MNI space resulting in whole-

brain dynamic [carbonyl-11C]WAY-100635 maps co-registered to the structural MRI images.

36

Quantification of 5-HT1A Receptor Binding Potential in Anatomical Regions of Interest

With this post-hoc analysis we aimed to investigate the area-specific relationship between 5-

HT1A and GMV to further confirm our primary voxel-by-voxel results using a different

approach. The ROI-based analysis also served to test, whether there exists a network

association between 5-HT1A receptors and gray matter in the projection areas of the one of

the main raphe nuclei, the dorsal raphe nucleus. For the ROI-based network analysis the

DRN was manually delineated on an averaged PETADD image in PMOD 3.1. Our DRN ROI

consisted of a sphere, 4 mm in diameter, comprising three slices on the averaged PETADD

image. Each individual’s 5-HT1A BPND value in the DRN was then obtained from individual

time-activity curves averaged across subjects (again using cerebellar gray matter as

reference). For further post-hoc analysis we quantified 5-HT1A receptor BPND values and

GMV values in MNI standard space in 48 ROIs, taken from the AAL atlas covering a broad

range of brain regions as previously shown (Stein et al., 2008). A ratio between GMV and

rescaled 5-HT1A BPND values was calculated by dividing the first through the latter.

Statistical analyses

Demographics

Sex differences in biological demographical and radiochemical variables were calculated to

assess study sample characteristics with either independent sample t-tests or Mann-Whitney

U tests where appropriate, using IBM SPSS Statistics (v19.0, 2010, SPSS, Inc., an IBM

Company, Chicago, United States of America) assuming a significance level of α = 0.05.

Multimodal Analysis

Multimodal image analysis was divided into two parts: we calculated regional voxel-by-voxel

associations between 5-HT1A receptor distribution and gray matter in the whole-brain.

Second, we assessed the associations between 5-HT1A receptor binding in a single area, the

DRN, and GMV in projection sites of the DRN. The DRN was chosen because of its central

role in the regulation of serotonergic firing and neurotransmission.

We thus calculated a multiple linear regression model with 5-HT1A receptor BPND values as

independent and GMV values as dependent variable for every voxel in the entire gray matter.

In this regression model age, sex and total GMV served as controlling variables. This was

done to adjust for age related gray matter alteration, varying brain sizes, and sex differences

(as outlined in Table 1) for. In two further models, we also considered the two radiochemical

variables specific activity (SA) and injected dose (ID) as factors. However, given that the

37

number of control variables should not exceed n/10 and the results (data not shown) were

virtually identical with the primary model, we did not include SA and ID in our further

analyses. The voxel-by-voxel regression model was set up in the Biological Parametric

Mapping (BPM) toolbox for SPM8 (Casanova et al., 2007), which is designed to calculate

voxel-by-voxel statistics for multiple imaging modalities. More precisely, multiple regression

was calculated in each voxel (average voxel number across all subjects = 216,741.2) with

one value for GMV and one for 5-HT1A BPND (in arbitrary units). We used 0.1 as absolute

threshold and a level of statistical significance of α = 0.001. Due to multiple comparisons and

the concomitant high chance of false positives the obtained results were corrected with the

cluster-level false discovery rate (FDR) at a significance level of α = 0.05. Correlation

coefficients were calculated with cluster-wise means (in arbitrary units) in Matlab (v. r2010b,

The MathWorks, Inc., Natick, United States of America).

For the analysis of serotonergic projections from the DRN, we calculated a regression model

in SPM8 using 5-HT1A BPND values of the DRN ROI as independent variables and whole

brain GMV as dependent variable. Sex, age and total GMV were control variables for the

reasons mentioned above. Further, GMV of the DRN was added in the regression model to

eliminate potential confounding effects of DRN gray matter and whole brain gray matter

interactions. GMV values were obtained from the DRN ROI overlaid on the MRI images. We

excluded voxels exhibiting BPND or GMV voxel values below 0.1. The level of statistical

significance was set at α = 0.001 and only results with a cluster size over 100 voxels are

reported.

For the analysis of smoking status on BPND a regression analysis was set up in SPM8 using

5-HT1A BPND values as independent variables and smoking status or number of smoked

cigarettes as dependent variables, respectively, controlling for sex, age and GMV. Age effects

on GMV were calculated with a regression analysis using GMV as independent variable and

age as dependent variable controlling for sex and GMV. In both analyses an uncorrected α =

0.001 was accepted as level of significance.

RESULTS

5-HT1A receptor binding positively correlated with gray matter volumes within

distinctive brain regions

In this pooled study sample, male study subjects significantly differed from females in GMV,

weight and total injected radiotracer dose (Table 1). In line with previous results of our group,

5-HT1A BPND, an index for receptor density, peaked in the parahippocampal gyri, the temporal

poles and the insula (Figures 1A, Figure 3, Table S1 and [Stein et al., 2008]).

38

Serotonin-1A BPND strongly correlated with GMV in the hippocampus (the cluster in the right

hippocampus spread from the posterior hippocampus to the parahippocampus), the posterior

medial temporal cortex, the posterior inferior temporal cortex, the medial occipital cortex and

the pericalcarine region in each hemisphere (R2 values ranged from 0.308 to 0.503, p < 0.05

cluster-level false discovery rate [FDR]) corrected, see Figures 1B, 1C and Table 2). In other

words, 5-HT1A heteroreceptor binding strongly correlated with relative volumes of gray matter

in these specific regions. Negative correlations between 5-HT1A BPND and GMV were

restricted to two regions in the cerebellum (Table 2).

Figure 1 Serotonin-1A (5-HT1A) receptor binding is positively associated with regional gray matter.

(A) 5-HT1A receptor distribution in vivo measured with positron emission tomography displayed with

the surface-rendering algorithm used by the visualization program MRIcro

(http://www.cabiatl.com/mricro/mricro/mricro.html) (B) T maps showing that 5-HT1A binding potential

(BPND) strongly correlates with gray matter volume (GMV). Significant positive correlations were

superimposed on MR images (p < 0.05, FDR cluster-level corrected, see Table 2), coordinates

correspond to the standard Montreal Neurological Institute (MNI) stereotactic system. (C) Regression

graphs between GMV and 5-HT1A BPND (multiple regression analysis controlled for sex, age and total

GMV, adjusted values in arbitrary units) correspond to cluster means of each subject (in red circles

(B), N = 35).

39

5-HT1A receptor binding in the raphe region positively correlated with gray

matter volume in the anterior cingulate cortex

Previous data show that presynaptic 5-HT1A autoreceptors in the DRN regulate tonic

serotonergic firing, serotonin release and the postsynaptic density of 5-HT1A

heteroreceptors and 5-HT transporters (Bose et al., 2011). Hypothesizing that the

influence of the DRN autoregulation extends to gray matter, we investigated

associations between 5-HT1A autoreceptor binding in the DRN and whole brain GMV

at projection sites. We observed a positive correlation between the dorsal raphe 5-

HT1A BPND and GMV in the right perigenual anterior cingulate cortex (R2 = 0.656, p =

0.001, uncorrected, Figure 2A, 2B).

Post-hoc ROI analysis revealed regional differences in the relation between 5-HT1A

receptor binding and GMV

An intuitive caveat to the results might be that these associations could be merely

based on primary larger numbers of neuronal or glial cells expressing 5-HT1A

Figure 2 Network analysis. 5-HT1A receptor

binding of the dorsal raphe nucleus (DRN) is

positively associated with gray matter volume of

the anterior cingulate cortex (ACC). (A)

Significant cluster superimposed on a sagittal

MRI slice (regression analysis, R2 = 0.656, p <

0.001, uncorrected, cluster peak: t = 5, MNI: x =

6, y = 35, z = 3). (B) Data points represent cluster

means (adjusted values in arbitrary units) of each

subject (N = 35) as adjusted by regression

analysis controlled for sex, age total GMV and

GMV of the DRN.

40

receptors and thus a priori higher GMV values. Therefore, we investigated if the

resulting clusters were exclusively situated in regions with high regional GMV and

quantified the BPND and GMV values of 48 ROIs covering the whole brain. We found

several regions, such as the cingulate cortex or the amygdala, which despite high

regional GMV and 5-HT1A BPND values did not exhibit significant positive associations

in the voxel-by-voxel analysis (Figure 3). Furthermore, we calculated ratios between

GMV and BPND values to assess regional proportions between 5-HT1A receptor

binding and gray matter in the whole brain. These GMV/BPND ratios ranged from 0.54

in the temporal pole to 4.8 in the caudate region, suggesting high regional variability

within the ratio of regional GMV and 5-HT1A BPND (Figure 3).

Following that, to confirm the associations between 5-HT1A BPND and GMV obtained

by voxel-by-voxel analysis, we repeated the regression analysis within two ROIs. The

ROIs should have similar GMV and 5-HT1A BPND values, one exhibiting and one

lacking the associations as obtained by voxel-by-voxel analysis. Out of the 48

quantified ROIs, the hippocampus and the insula were the only two regions meeting

the selection criteria (GMV/BPND: insula = 0.63, hippocampus = 0.64, GMV: insula

and hippocampus = 0.53, BPND: insula = 0.84, hippocampus = 0.83, see Figure 3

and Table S1). In the voxel-by-voxel analysis, the hippocampus exhibited significant

positive associations between 5-HT1A receptor BPND and GMV, but in the insula,

despite similar values, 5-HT1A BPND did not correlate with GMV. Congruent to the

voxel-by-voxel analysis, in the post-hoc ROI analysis a significant positive correlation

was observed in the hippocampus (r = 0.41, p = 0.02) but not in the insula (r = - 0.03,

p = 0.87).

No effects of age or smoking status

To rule out cortical atrophy due to aging was somehow related to the results, we analyzed

our dataset for age-related effects. Multiple regression analysis in SPM8 revealed a negative

41

Figure 3 Area-specific differences in the relation between 5-HT1A receptor binding and gray matter

volume. Gray matter volume (GMV, red) and 5-HT1A binding potential (BPND, blue), in arbitrary units,

quantified in 48 brain regions of interest (ROI) covering the whole brain. This demonstrates high

variabilities between 5-HT1A receptor densities and regional volumes of gray matter (also see Table

S1).

correlation for GMV and age in the left medial occipital cortex (t = 4.24, p < 0.001,

uncorrected, x = -27, y = -81, z = 26) near the angular gyrus. BPND was negatively correlated

with age in a cluster around the left postcentral gyrus (t = 3.82, p < 0.001, uncorrected; x = -

22, y = -27, z = 62). These results indicate that an effect of aging in our data occurred in

different brain areas than the main results. Smoking status was available for 34 participants,

out of which 14 were smokers (6 female, mean cigarettes per day = 7.1 ± 4.8). Multiple

42

regression analysis revealed that neither smoking status nor number of smoked cigarettes

was associated with 5-HT1A BPND (all p > 0.001).

DISCUSSION

Our results demonstrate positive associations between 5-HT1A receptor binding and gray

matter. In distinctive regions of both hemispheres, as in the hippocampi and in temporal

cortices, 5-HT1A receptor binding was strongly correlated with gray matter. These results

were not just based on a priori higher regional values of gray matter, because we

demonstrated that in regions such as the insula, in contrast to the hippocampus, there were

no significant positive associations, although having comparable gray matter and 5-HT1A

receptor binding. We observed a large variability between 5-HT1A binding and gray matter in

the whole brain. We also found that 5-HT1A autoreceptor binding in the DRN was positively

associated with gray matter in the anterior cingulate cortex. The results were not affected by

cortical atrophy due to aging or smoking status. A large number of previous findings in animal

models (Daubert and Condron, 2010; Gaspar et al., 2003) show direct links between

serotonergic receptors like the 5-HT1A receptor and neuroplasticity. Furthermore, there is

evidence that allows direct inference from MRI-based measurements to changes of the

underlying neuronal structures (la Fougère et al., 2010). Therefore, we propose that the

discovered associations provide valuable insights into the relationship between 5-HT1A

receptor binding and gray matter cytoarchitecture in adult human brains in vivo.

Serotonin is highly active in shaping neurons during embryonic development and early

postnatal neuronal maturation, and this neuroplastic role is partially conserved in specific

brain regions throughout adulthood (Gould, 1999). Downstream cytosolic signaling kinases

from membrane-bound small G proteins (Ye and Carew, 2010), that activate transcription

factors (McClung and Nestler, 2008) and epigenetic mechanisms (Borrelli et al., 2008) were

suggested to effect neuronal reconfigurations. Serotonergic-1A receptors are able to

modulate the activity of these pathways (Cowen, 2007; Polter and Li, 2010). Recently a study

using hippocampal cell cultures could show, that 5-HT1A receptors are essential for normal

synaptogenesis (Mogha et al., 2012). Blockade of astrocytic 5-HT1A receptors leads to a

reduction of synaptic connections between neurons (Wilson et al., 1998) and fits well to

findings demonstrating that the 5-HT1A receptor is required for behavioral and neurogenic

effects of the selective serotonin reuptake inhibitors (Santarelli et al., 2003). From a

neurobiological perspective, we suggest that neuroplastic effects of 5-HT1A receptors might

contribute to the observed association between 5-HT1A binding and alterations of regional

gray matter.

43

Nevertheless, this interpretation must be considered with caution, because one voxel in high-

resolution structural MRI contains too many neuronal cells to reliably link our results with

mechanisms observed in cell cultures or animal models (for review see [(May, 2011)]). To

better pin down how 5-HT1A receptor-mediated neuroplasticity might affect gray matter in the

living brain, further longitudinal investigations are needed. A possible alternative explanation

to our results could simply be, that regional 5-HT1A binding was elevated through primary

higher regional amounts of gray matter. However, considering results from animal models,

we have several arguments against this.

Firstly, we predominately found positive associations. In a similar study using structural MRI,

PET and the D2/D3 receptor ligand [18F]fallypride, Woodward et al. (Woodward et al., 2009)

previously pointed out that negative associations are unexpected. Hypothetically, the density

of 5-HT1A receptors should vary with the amount of gray matter within a region, in other

words the more gray matter, the more receptors it can support and vice versa. A recent study,

however, nicely demonstrates divergent values between 5-HT1A binding and neuronal

densities in humans as measured by stereology and autoradiography (Underwood et al.,

2008). This result is congruent to the broad variability in the range between 5-HT1A binding

and GMV shown in our study. The predominantly positive associations in our study go well

along with the finding, that astrocytic 5-HT1A receptors (via S-100ß) are necessary for

maintaining the neuronal integrity (Whitaker-Azmitia, 2001). In the absence of S-100ß a

mature neuron can regress its major processes and even enter apoptosis (Whitaker-Azmitia,

2001). Secondly, the associations between 5-HT1A binding and gray matter were obtained

symmetrically in both hemispheres, which indicates validity. The distinctive regional pattern

could be explained by varying strengths of 5-HT1A mediated neuroplastic effects (Cowen,

2007). Thirdly, the insula, even with similar 5-HT1A and GMV values as the hippocampus, did

not exhibhit significant associations. This further suggests a region-specific mechanism and

might indicate that the observed associations were not based on higher numbers of regional

neuronal and glial cells, both associated with GMV. According to the current state of

knowledge 5-HT1A mediated neroplasticity is more active in the hippocampus than in the

insula (Santarelli et al., 2003). In the hippocampus 5-HT1A receptors were demonstrated to

stimulate neurogenesis and dendritic maturation (Yan et al., 1997). Finally, we demonstrated

an association between 5-HT1A receptors in one of the major serotonergic nuclei, the DRN,

and gray matter at serotonergic axon terminals in the anterior cingulate cortex.

Hypothetically, interregional correlation between 5-HT1A auto- and heteroreceptors (Hahn et

al., 2010) could foster the observed association between 5-HT1A receptors and GMV in this

study. The autoregulatory influence of the DRN on serotonergic heteroreceptors at axon

terminals in the forebrain, by neuroplastic properties of 5-HT1A receptors, might thus extend

to GMV. Patients suffering from mood disorders exhibit both significantly reduced GMV of the

44

anterior cingulate cortex and altered 5-HT1A receptor density in the raphe nucleus (Salvadore

et al., 2011; Savitz and Drevets, 2009; van Tol et al., 2010). We could speculate here, that a

disturbance in this association might contribute to the reduction of GMV in the anterior

cingulate cortex.

In summary, the associations between 5-HT1A binding and GMV could theoretically result

from a priori higher regional amounts of gray matter or other unknown mechanisms. But

given the high amount of clear evidence, we suggest that neuroplastic actions of 5-HT1A

receptors should be taken into account as explanatory model for this dataset. The 5-HT1A

receptor, in addition, could prove to be an interesting target in clinical studies on altered

neuroplasticity in brain disorders, due to well known behavioral behavioral functions such as

mediating mood (Savitz et al., 2009), anxiety (Akimova et al., 2009) or cognition (Ogren et

al., 2008).

Limitations

This dataset does not imply that the observed associations can be causally attributed to

neuroplastic actions of 5-HT1A receptors. For such a deduction a longitudinal, interventional

and translational approach in a future study would be more favorable, for which this dataset

provides excellent justification.

Furthermore, as previously pointed out (Tost et al., 2010) the neurobiological correlates of

changes in brain morphology measured by structural neuroimaging are not sufficiently

resolved, for an excellent recent review see (Zatorre et al., 2012). Even at high-resolution

MRI, there are still ten thousands of interconnected neuronal and glial cells packed in one

single voxel. Thus, more translational cell studies on neuroplasticity are necessary to exactly

determine what cellular processes are mediated by serotonin and the 5-HT1A receptor that

could gain effects, large enough to be detectable by structural MRI (May, 2011).

Finally, we did not use correction for partial volume effects (PVC) of the PET data. Although

this may be an obvious issue, PVC is typically carried out by using the corresponding

segmented MRI, namely, the gray and white matter probability maps. More precisely, the GM

values represent the denominator of the PVC algorithm (Muller-Gartner et al., 1992). This

implies that the PET activity concentrations are adjusted for individual differences in the GM

volume. However, the current study particularly aims to investigate the association between

individual differences in 5-HT1A binding and GM volume. Hence, MRI-based PVC would

include the effect of interest as nuisance variable, which in turn cancels the association.

Accordingly, no PVC was carried out in the similar investigation of Woodward et al. (2009).

45

Conclusions

Our results demonstrate that 5-HT1A receptor binding is positively associated with gray

matter in specific regions such as the hippocampus and the temporal cortices in both

hemispheres. Furthermore 5-HT1A autoreceptor binding in the midbrain is positively

associated gray matter in the anterior cingulate cortex. Currently, it is hard to pin down the

molecular mechanisms underlying our results, mostly because, there are no exact models

which cellular compounds correspond to the signal strength in a single voxel. To increase the

validity of neuroimaging studies, this issue must be an objective of further studies.

With regard to translational neuroscience, assessments of processes underlying networking

and reorganization of neurons as well as early surrogate markers to predict and monitor

treatment response were demanded (Cramer et al., 2011). With combinations of structural

and molecular neuroimaging, as performed in this multimodal study, dysfunctional

neuroregulatory processes leading to loss of gray matter might be investigated at early

stages in clinical populations. This could lead to a more comprehensive understanding of

neurodegenerative diseases as Alzheimer’s disease, schizophrenia and mood disorders and

ultimately to a better diagnostic assessment and therapeutic evaluation of patients with these

highly life impairing disorders.

ACKNOWLEDGEMENTS

This research was partly supported by grants from the Austrian Science Fund, and the

Austrian National Bank (P 11468) to R. L. A. Hahn is recipient of a DOC-fellowship of the

Austrian Academy of Sciences at the Department of Psychiatry and Psychotherapy. We are

grateful to the technical and medical teams of the PET and High-Field MRI Centre, Medical

University of Vienna, especially to K. Kletter, R. Dudczak, E. Moser, L.-K. Mien, and F. Gerstl.

Furthermore, we would like to thank U. Moser, M. Fink, and P. Stein for medical support and

A. Saulin for help with the manuscript.

46

REFERENCES

Akimova, E., Lanzenberger, R., Kasper, S., 2009. The serotonin-1A receptor in anxiety disorders. Biol Psychiatry 66, 627-635.

Amargós-Bosch, M., Bortolozzi, A., Puig, M.V., Serrats, J., Adell, A., Celada, P., Toth, M., Mengod, G., Artigas, F., 2004. Co-expression and in vivo interaction of serotonin1A and serotonin2A receptors in pyramidal neurons of prefrontal cortex. Cereb Cortex 14, 281-299.

Ashburner, J., 2007. A fast diffeomorphic image registration algorithm. NeuroImage 38, 95-113.

Ashburner, J., Friston, K.J., 2009. Computing average shaped tissue probability templates. NeuroImage 45, 333-341.

Azmitia, E.C., 2001. Modern views on an ancient chemical: serotonin effects on cell proliferation, maturation, and apoptosis. Brain Res Bull 56, 413-424.

Benninghoff, J., Gritti, A., Rizzi, M., Lamorte, G., Schloesser, R.J., Schmitt, A., Robel, S., Genius, J., Moessner, R., Riederer, P., Manji, H.K., Grunze, H., Rujescu, D., Moeller, H.-J., Lesch, K.-P., Vescovi, A.L., 2010. Serotonin depletion hampers survival and proliferation in neurospheres derived from adult neural stem cells. Neuropsychopharmacology 35, 893-903.

Borrelli, E., Nestler, E.J., Allis, C.D., Sassone-Corsi, P., 2008. Decoding the epigenetic language of neuronal plasticity. Neuron 60, 961-974.

Bose, S.K., Mehta, M.A., Selvaraj, S., Howes, O.D., Hinz, R., Rabiner, E.A., Grasby, P.M., Turkheimer, F.E., Murthy, V., 2011. Presynaptic 5-HT1A is Related to 5-HTT Receptor Density in the Human Brain. Neuropsychopharmacology.

Casanova, R., Srikanth, R., Baer, A., Laurienti, P.J., Burdette, J.H., Hayasaka, S., Flowers, L., Wood, F., Maldjian, J.A., 2007. Biological parametric mapping: A statistical toolbox for multimodality brain image analysis. NeuroImage 34, 137-143.

Citri, A., Malenka, R.C., 2008. Synaptic plasticity: multiple forms, functions, and mechanisms. Neuropsychopharmacology 33, 18-41.

Cowen, D.S., 2007. Serotonin and neuronal growth factors - a convergence of signaling pathways. J Neurochem 101, 1161-1171.

Cramer, S.C., Sur, M., Dobkin, B.H., O'Brien, C., Sanger, T.D., Trojanowski, J.Q., Rumsey, J.M., Hicks, R., Cameron, J., Chen, D., Chen, W.G., Cohen, L.G., deCharms, C., Duffy, C.J., Eden, G.F., Fetz, E.E., Filart, R., Freund, M., Grant, S.J., Haber, S., Kalivas, P.W., Kolb, B., Kramer, A.F., Lynch, M., Mayberg, H.S., McQuillen, P.S., Nitkin, R., Pascual-Leone, A., Reuter-Lorenz, P., Schiff, N., Sharma, A., Shekim, L., Stryker, M., Sullivan, E.V., Vinogradov, S., 2011. Harnessing neuroplasticity for clinical applications. Brain 134, 1591-1609.

Daubert, E.A., Condron, B.G., 2010. Serotonin: a regulator of neuronal morphology and circuitry. Trends Neurosci 33, 424-434.

Draganski, B., Gaser, C., Busch, V., Schuierer, G., Bogdahn, U., May, A., 2004. Neuroplasticity: changes in grey matter induced by training. Nature 427, 311-312.

Gaspar, P., Cases, O., Maroteaux, L., 2003. The developmental role of serotonin: news from mouse molecular genetics. Nat Rev Neurosci 4, 1002-1012.

47

Geuze, E., Vermetten, E., Bremner, J.D., 2005. MR-based in vivo hippocampal volumetrics: 2. Findings in neuropsychiatric disorders. Mol Psychiatry 10, 160-184.

Gould, E., 1999. Serotonin and hippocampal neurogenesis. Neuropsychopharmacology 21, 46S-51S.

Hahn, A., Lanzenberger, R., Wadsak, W., Spindelegger, C., Moser, U., Mien, L.-K., Mitterhauser, M., Kasper, S., 2010. Escitalopram enhances the association of serotonin-1A autoreceptors to heteroreceptors in anxiety disorders. J Neurosci 30, 14482-14489.

Hall, H., Lundkvist, C., Halldin, C., Farde, L., Pike, V.W., McCarron, J.A., Fletcher, A., Cliffe, I.A., Barf, T., Wikstrom, H., Sedvall, G., 1997. Autoradiographic localization of 5-HT1A receptors in the post-mortem human brain using [3H]WAY-100635 and [11C]way-100635. Brain Res 745, 96-108.

Ichise, M., Liow, J.-S., Lu, J.-Q., Takano, A., Model, K., Toyama, H., Suhara, T., Suzuki, K., Innis, R.B., Carson, R.E., 2003. Linearized Reference Tissue Parametric Imaging Methods: Application to [11C]DASB Positron Emission Tomography Studies of the Serotonin Transporter in Human Brain. Journal of Cerebral Blood Flow & Metabolism, 1096-1112.

Innis, R.B., Cunningham, V.J., Delforge, J., Fujita, M., Gjedde, A., Gunn, R.N., Holden, J., Houle, S., Huang, S.-C., Ichise, M., Iida, H., Ito, H., Kimura, Y., Koeppe, R.A., Knudsen, G.M., Knuuti, J., Lammertsma, A.A., Laruelle, M., Logan, J., Maguire, R.P., Mintun, M.A., Morris, E.D., Parsey, R., Price, J.C., Slifstein, M., Sossi, V., Suhara, T., Votaw, J.R., Wong, D.F., Carson, R.E., 2007. Consensus nomenclature for in vivo imaging of reversibly binding radioligands. J Cereb Blood Flow Metab 27, 1533-1539.

Kanai, R., Rees, G., 2011. The structural basis of inter-individual differences in human behaviour and cognition. Nature reviews Neuroscience.

Kasper, S., Tauscher, J., Willeit, M., Stamenkovic, M., Neumeister, A., Kufferle, B., Barnas, C., Stastny, J., Praschak-Rieder, N., Pezawas, L., de Zwaan, M., Quiner, S., Pirker, W., Asenbaum, S., Podreka, I., Brucke, T., 2002. Receptor and transporter imaging studies in schizophrenia, depression, bulimia and Tourette's disorder--implications for psychopharmacology. World J Biol Psychiatry 3, 133-146.

Kepe, V., Barrio, J.R., Huang, S.C., Ercoli, L., Siddarth, P., Shoghi-Jadid, K., Cole, G.M., Satyamurthy, N., Cummings, J.L., Small, G.W., Phelps, M.E., 2006. Serotonin 1A receptors in the living brain of Alzheimer's disease patients. Proc Natl Acad Sci U S A 103, 702-707.

Klein, A., Andersson, J., Ardekani, B.A., Ashburner, J., Avants, B., Chiang, M.-C., Christensen, G.E., Collins, D.L., Gee, J., Hellier, P., Song, J.H., Jenkinson, M., Lepage, C., Rueckert, D., Thompson, P., Vercauteren, T., Woods, R.P., Mann, J.J., Parsey, R.V., 2009. Evaluation of 14 nonlinear deformation algorithms applied to human brain MRI registration. NeuroImage 46, 786-802.

la Fougère, C., Grant, S., Kostikov, A., Schirrmacher, R., Gravel, P., Schipper, H., Reader, A., Evans, A., Thiel, A., 2010. Where in-vivo imaging meets cytoarchitectonics: The relationship between cortical thickness and neuronal density measured with high-resolution [(18)F]flumazenil-PET. NeuroImage.

Lanzenberger, R.R., Mitterhauser, M., Spindelegger, C., Wadsak, W., Klein, N., Mien, L.-K., Holik, A., Attarbaschi, T., Mossaheb, N., Sacher, J., Geiss-Granadia, T., Kletter, K., Kasper, S., Tauscher, J., 2007. Reduced serotonin-1A receptor binding in social anxiety disorder. Biol Psychiatry 61, 1081-1089.

48

Lewis, D.A., González-Burgos, G., 2008. Neuroplasticity of Neocortical Circuits in Schizophrenia. Neuropsychopharmacology 33, 141-165.

Mamo, D., Graff, A., Mizrahi, R., Shammi, C.M., Romeyer, F., Kapur, S., 2007. Differential effects of aripiprazole on D(2), 5-HT(2), and 5-HT(1A) receptor occupancy in patients with schizophrenia: a triple tracer PET study. Am J Psychiatry 164, 1411-1417.

May, A., 2011. Experience-dependent structural plasticity in the adult human brain. Trends Cogn Sci 15, 475-482.

McClung, C.A., Nestler, E.J., 2008. Neuroplasticity mediated by altered gene expression. Neuropsychopharmacology 33, 3-17.

Mesulam, M.M., 1999. Neuroplasticity failure in Alzheimer's disease: bridging the gap between plaques and tangles. Neuron 24, 521-529.

Mogha, A., Guariglia, S., Debata, P., Wen, G., Banerjee, P., 2012. Serotonin 1A receptor-mediated signaling through ERK and PKCa is essential for normal synaptogenesis in neonatal mouse hippocampus. Translational Psychiatry 2.

Muller-Gartner, H.W., Links, J.M., Prince, J.L., Bryan, R.N., McVeigh, E., Leal, J.P., Davatzikos, C., Frost, J.J., 1992. Measurement of radiotracer concentration in brain gray matter using positron emission tomography: MRI-based correction for partial volume effects. J Cereb Blood Flow Metab 12, 571-583.

Ogren, S.O., Eriksson, T.M., Elvander-Tottie, E., D'Addario, C., Ekström, J.C., Svenningsson, P., Meister, B., Kehr, J., Stiedl, O., 2008. The role of 5-HT(1A) receptors in learning and memory. Behav Brain Res 195, 54-77.

Pittenger, C., Duman, R.S., 2008. Stress, depression, and neuroplasticity: a convergence of mechanisms. Neuropsychopharmacology 33, 88-109.

Puig, M.V., Artigas, F., Celada, P., 2005. Modulation of the activity of pyramidal neurons in rat prefrontal cortex by raphe stimulation in vivo: involvement of serotonin and GABA. Cereb Cortex 15, 1-14.

Salvadore, G., Nugent, A.C., Lemaitre, H., Luckenbaugh, D.A., Tinsley, R., Cannon, D.M., Neumeister, A., Jr, C.A.Z., Drevets, W.C., 2011. Prefrontal cortical abnormalities in currently depressed versus currently remitted patients with major depressive disorder. NeuroImage 54, 2643-2651.

Santarelli, L., Saxe, M., Gross, C., Surget, A., Battaglia, F., Dulawa, S., Weisstaub, N., Lee, J., Duman, R., Arancio, O., Belzung, C., Hen, R., 2003. Requirement of hippocampal neurogenesis for the behavioral effects of antidepressants. Science 301, 805-809.

Savitz, J., Lucki, I., Drevets, W.C., 2009. 5-HT(1A) receptor function in major depressive disorder. Prog Neurobiol 88, 17-31.

Savitz, J.B., Drevets, W.C., 2009. Imaging phenotypes of major depressive disorder: genetic correlates. Neuroscience 164, 300-330.

Spindelegger, C., Lanzenberger, R., Wadsak, W., Mien, L.K., Stein, P., Mitterhauser, M., Moser, U., Holik, A., Pezawas, L., Kletter, K., Kasper, S., 2009. Influence of escitalopram treatment on 5-HT 1A receptor binding in limbic regions in patients with anxiety disorders. Mol Psychiatry 14, 1040-1050.

49

Stein, P., Savli, M., Wadsak, W., Mitterhauser, M., Fink, M., Spindelegger, C., Mien, L.K., Moser, U., Dudczak, R., Kletter, K., Kasper, S., Lanzenberger, R., 2008. The serotonin-1A receptor distribution in healthy men and women measured by PET and [carbonyl-11C]WAY-100635. Eur J Nucl Med Mol Imaging 35, 2159-2168.

Tardito, D., Perez, J., Tiraboschi, E., Musazzi, L., Racagni, G., Popoli, M., 2006. Signaling pathways regulating gene expression, neuroplasticity, and neurotrophic mechanisms in the action of antidepressants: a critical overview. Pharmacol Rev 58, 115-134.

Tost, H., Braus, D.F., Hakimi, S., Ruf, M., Vollmert, C., Hohn, F., Meyer-Lindenberg, A., 2010. Acute D2 receptor blockade induces rapid, reversible remodeling in human cortical-striatal circuits. Nat Neurosci 13, 920-922.

Tzourio-Mazoyer, N., Landeau, B., Papathanassiou, D., Crivello, F., Etard, O., Delcroix, N., Mazoyer, B., Joliot, M., 2002. Automated Anatomical Labeling of Activations in SPM Using a Macroscopic Anatomical Parcellation of the MNI MRI Single-Subject Brain. NeuroImage 15, 273-289.

Underwood, M.D., Mann, J.J., Huang, Y.-Y., Arango, V., 2008. Family history of alcoholism is associated with lower 5-HT2A receptor binding in the prefrontal cortex. Alcohol Clin Exp Res 32, 593-599.

van Tol, M.-J., van der Wee, N.J.A., van den Heuvel, O.A., Nielen, M.M.A., Demenescu, L.R., Aleman, A., Renken, R., van Buchem, M.A., Zitman, F.G., Veltman, D.J., 2010. Regional brain volume in depression and anxiety disorders. Arch Gen Psychiatry 67, 1002-1011.

Vetencourt, J.F.M., Sale, A., Viegi, A., Baroncelli, L., De Pasquale, R., O'Leary, O.F., Castrén, E., Maffei, L., 2008. The antidepressant fluoxetine restores plasticity in the adult visual cortex. Science 320, 385-388.

Wadsak W., M.L.K., Ettlinger D.E., Lanzenberger R., Haeusler D., Dudczak R., Kletter K., Mitterhauser M., 2007. Simple and fully automated preparation of [carbonyl-11C]WAY-100635. Radiochimica Acta 95, 1-6.

Whitaker-Azmitia, P.M., 2001. Serotonin and brain development: role in human developmental diseases. Brain Res Bull 56, 479-485.

Wilson, C.C., Faber, K.M., Haring, J.H., 1998. Serotonin regulates synaptic connections in the dentate molecular layer of adult rats via 5-HT1a receptors: evidence for a glial mechanism. Brain Res 782, 235-239.

Woodward, N.D., Zald, D.H., Ding, Z., Riccardi, P., Ansari, M.S., Baldwin, R.M., Cowan, R.L., Li, R., Kessler, R.M., 2009. Cerebral morphology and dopamine D2/D3 receptor distribution in humans: a combined [18F]fallypride and voxel-based morphometry study. NeuroImage 46, 31-38.

Yan, W., Wilson, C.C., Haring, J.H., 1997. 5-HT1a receptors mediate the neurotrophic effect of serotonin on developing dentate granule cells. Brain Res Dev Brain Res 98, 185-190.

Ye, X., Carew, T.J., 2010. Small G protein signaling in neuronal plasticity and memory formation: the specific role of ras family proteins. Neuron 68, 340-361.

Zatorre, R.J., Fields, R.D., Johansen-Berg, H., 2012. Plasticity in gray and white: neuroimaging changes in brain structure during learning. Nat Neurosci 15, 528-536.

50

Table 1 Demographic and radiochemical variables of study subjects

all subjects

males

females

p

n 35

17

18

age 26.6 ± 6.8

29.6 ± 8.4

24.4 ± 2.5

0.026+

weight (kg) 71.3 ± 14.6

79.7 ± 11.7

62.5 ± 12.2

< 0.001

GMV (cm3) 731.5 ± 73.8

777.5 ± 53.9

682.8 ± 58.5

< 0.001

injected dose (MBq) 385 ± 36

396.9 ± 45.8

372.3 ± 14.4

0.002+

RCP (%) 97.7 ± 1.4

98 ± 1.4

97.4 ± 1.3

0,320

Data are given as means ± standard deviation. GMV = total grey matter volume, MBq =

Megabecquerel, RCP = radiochemical purity, p compares males and females with

independant sample t-test or Mann-Whitney U test (+) where normal distribution was not

obtained by Levene's test.

51

Table 2

Statistical results as obtained by Statistical Parametric Mapping (SPM8).

region

peak

cluster

x y z t z R2 p−FWE p−FDR voxels

positive correlation

right posterior medial temporal

42 −32 0

6.7 5.2

0.487 < 0.001 < 0.001 952

right hippocampus/ parahippocampus

29 −39 0

6.2 4.9

0.503 < 0.001 < 0.001 850

right medial occipital

38 −92 −5

4.9 4.1

0.308 < 0.001 < 0.001 541

right inferior orbitofrontal

51 20 −8

4.9 4.2

0.403 0.002 0.001 434

right posterior inferior temporal

59 −32 −15

5.4 4.5

0.436 0.002 0.002 416

left posterior medial temporal

−44 −26 −6

6.7 5.2

0.386 0.003 0.002 400

right superior parietal

24 −53 48

6.0 4.8

0.449 0.004 0.002 381

left posterior inferior temporal

−53 −42 −14

5.1 4.3

0.302 0.004 0.002 374

left medial occipital

−21 −101 9

4.9 4.2

0.217 0.006 0.002 349

left pericalcarine

−27 −57 5

6.1 4.9

0.424 0.009 0.003 323

left precentral

−33 5 41

5.7 4.7

0.183 0.031 0.01 253

right pericalcarine

30 −57 8

7.2 5.4

0.489 0.043 0.013 234

right inferior occipital

38 −65 −11

5.5 4.5

0.308 0.089 0.026 195

left hippocampus

−24 −27 −14

5.3 4.4

0.428 0.127 0.035 176

negative correlation

left posterior lobe of cerebellum

−26 −57 −23

−6 −4.83

0.422 0.102 0.176 363

left cerebellar crus

−50 −69 −23

−5.4 −4.48

0.461 0.349 0.35 10

Voxel−wise regression analysis results between whole−brain 5−HT1A binding potential (BPND) and whole−brain grey

matter volume (GMV). Stereotactical coordinates (x. y. z) represent cluster peaks in standard Montreal Institute of

Neurology (MNI) space. FWE = family wise error. FDR = false discovery rate. Note that R2 values were calulated

cluster−wise between 5−HT1A BPND and GMV and therefore do not correspond to peak t − or z − values.

52

Table S1

Binding potential and gray matter volume values for 48 regions of interest as quantified with an AAL based atlas (data sorted by GMV/nBPND)

Region BPND nBPND GMV GMV/nBPND

temporal pole, middle temporal gyrus 4,98 0,87 0,47 0,54

temporal pole, superior temporal gyrus 5,26 0,92 0,50 0,54

parahippocampus 5,72 1,00 0,55 0,55

superior frontal, medial 3,48 0,61 0,34 0,56

superior frontal, orbital 3,61 0,63 0,37 0,59

superior parietal 2,96 0,52 0,31 0,59

rectus 4,53 0,79 0,48 0,61

inferior temporal 4,83 0,84 0,52 0,62

superior frontal 3,21 0,56 0,35 0,62

Heschl's gyrus 3,99 0,70 0,43 0,62

orbitofrontal 4,05 0,71 0,44 0,62

insula 4,82 0,84 0,53 0,63

hippocampus 4,73 0,83 0,53 0,64

superior temporal 3,99 0,70 0,45 0,65

supplemantary motor area 3,04 0,53 0,36 0,67

fusiform 4,90 0,86 0,57 0,67

middle temporal 4,32 0,76 0,51 0,68

olfactory sulcus 4,50 0,79 0,54 0,69

paracentral 2,88 0,50 0,35 0,70

medial orbitofrontal 3,47 0,61 0,43 0,70

precuneus 3,16 0,55 0,39 0,71

postcentral 2,70 0,47 0,35 0,73

medial occipital 3,36 0,59 0,43 0,74

inferior frontal, operculum 3,44 0,60 0,45 0,74

inferior orbitofrontal 3,48 0,61 0,45 0,75

inferior occipital 3,46 0,60 0,45 0,75

superior occipital 2,67 0,47 0,35 0,75

amygdala 4,21 0,74 0,55 0,75

midbrain 1,44 0,25 0,19 0,75

supramarginal 3,75 0,66 0,50 0,76

dorsal raphe nucleus 2,33 0,41 0,31 0,76

inferior parietal 3,37 0,59 0,45 0,76

medial frontal 3,36 0,59 0,45 0,77

angular 3,50 0,61 0,49 0,80

cuneus 2,59 0,45 0,36 0,80

precentral 2,49 0,43 0,35 0,81

inferior frontal, triangular 3,09 0,54 0,44 0,81

anterior cingulate 3,79 0,66 0,56 0,85

subgenual cingulate 3,76 0,66 0,56 0,86

medial cingulum 3,17 0,55 0,53 0,96

lingual 2,92 0,51 0,51 1,00

calcarine 2,17 0,38 0,40 1,06

53

posterior cingulate 2,51 0,44 0,49 1,11

Nucleus accumbens 1,45 0,25 0,54 2,11

thalamus 0,70 0,12 0,30 2,46

striatum 0,73 0,13 0,37 2,91

putamen 0,75 0,13 0,42 3,20

caudatus 0,64 0,11 0,54 4,80

ROI = region of interest, AAL = automatic anatomical labelling, BPND = binding potential, GMV = gray

matter volume. BPND and GMV values represent means across both hemispheres. For ROI

regression analysis values from one hemisphere were chosen. nBPND (normalized Binding Potential)

values were rescaled for visualization in Figure 3 by dividing through the highest BPND value (5.72 in

the parahippocampus). To further demonstrate a positive association (hippocampus) and a lack of it

(insula) the regions in bold were selected for ROI regression analyses based on their similarity of

GMV and BPND values.

54

1.12 Second publication: Gray matter and intrinsic network

changes in the posterior cingulate cortex after

selective serotonin reuptake inhibitor intake

Christoph Kraus1,2, Sebastian Ganger1,2, Jan Losak1,2, Andreas Hahn1,2,

Markus Savli1,2, Georg S. Kranz1,2, Pia Baldinger1,2, Christian Windischberger3,4,

Siegfried Kasper1, Rupert Lanzenberger1,2

1 Department of Psychiatry and Psychotherapy,

2 Functional, Molecular and Translational Neuroimaging Lab – PET & MRI,

3 Center for Medical Physics and Biomedical Engineering, 4 MR Centre of Excellence,

Medical University of Vienna, Austria

Published in

NeuroImage, 2014; 84:236-244

[2014, IF: 6.35]

* Correspondence to: Rupert Lanzenberger, Assoc. Prof., MD Functional, Molecular and Translational Neuroimaging Lab

Department of Psychiatry and Psychotherapy Medical University of Vienna Waehringer Guertel 18-20, 1090 Vienna, Austria

Phone (Fax): +43 1 40400 3825 (3099) [email protected]

Running Title: Gray matter and functional changes after SSRI intak

55

ABSTRACT

Preclinical studies have demonstrated that serotonin (5-HT) challenge changes neuronal

circuitries and microarchitecture. However, evidence in human subjects is missing.

Pharmacologic magnetic resonance imaging (phMRI) applying selective 5-HT reuptake

inhibitors (SSRIs) and high-resolution structural and functional brain assessment is able to

demonstrate the impact of 5-HT challenge on neuronal network morphology and functional

activity. To determine how SSRIs induce changes in gray matter and neuronal activity, we

conducted a longitudinal study using citalopram and escitalopram. Seventeen healthy

subjects completed a structural and functional phMRI study with randomized, cross-over,

placebo-controlled, double-blind design. Significant gray matter increases were observed

(amongst other regions) in the posterior cingulate cortex (PCC) and the ventral precuneus

after SSRI intake of 10 days, while decreases were observed within the pre- and postcentral

gyri (all P<0.05, family wise error [FWE] corrected). Furthermore, enhanced resting functional

connectivity (rFC) within the ventral precuneus and PCC was associated with gray matter

increases in the PCC (all FWE Pcorr<0.05). Corroborating these results, whole-brain

connectivity density, measuring the brain’s functional network hubs, was significantly

increased after SSRI-intake in the ventral precuneus and PCC (all FWE Pcorr<0.05). Short-

term administration of SSRIs changes gray matter structures, consistent with previous work

reporting enhancement of neuroplasticity by serotonergic neurotransmission. Furthermore,

increased gray matter in the PCC is associated with increased functional connectivity in one

of the brain’s metabolically most active regions. Our novel findings provide convergent

evidence for dynamic alterations of brain structure and function associated with SSRI

pharmacotherapy.

56

1. INTRODUCTION

Magnetic resonance imaging (MRI) and voxel-based morphometry (VBM) studies in patients

with depression and obsessive-compulsive disorder (OCD) showed gray matter

enhancements after treatment with selective serotonin reuptake inhibitors (SSRIs) (Hoexter

et al., 2012; Smith et al., 2012). Moreover, depressive patients homozygous for the LA-allele

in the SERT gene (rs25531) seem to be more susceptible to gray matter atrophy (Frodl et al.,

2008), this polymorphism also seems to affect gray matter in healthy subjects (Frodl et al.,

2008). Remarkably, at least 3 of the 16 known 5-HT receptors (5-HT1A, 5-HT1B and 5-HT2A)

(Gaspar et al., 2003) and the 5-HT transporter (SERT) (Benninghoff et al., 2012) are involved

in neuroplasticity processes (Gould, 1999; Mogha et al., 2012; Vitalis et al., 2002). Tight links

between the neurotrophin system and 5-HT have previously been shown (Castrén and

Rantamäki, 2010) demonstrating the role of 5-HT in regulating neuronal morphology and

circuitry (Daubert and Condron, 2010).

Selective 5-HT reuptake inhibitors represent the first line medication for depression (Bauer et

al., 2007), anxiety disorder, OCD, post-traumatic stress disorder and eating disorders (Aigner

et al., 2011; Bandelow et al., 2008). At the serotonergic synapse, SSRIs bind to a binding site

at the SERT and block reuptake of 5-HT (Kasper et al., 2009; Stahl, 1998). Treatment with

SSRIs results in rapid 20-fold increase in 5-HT levels within the midbrain raphe nuclei (Tao et

al., 2000), increases 5-HT binding at 5-HT1A autoreceptors there, which subsequently alters

neuronal firing rates and promotes desensitization of 5-HT1A receptors (Stahl, 1998; Zimmer

et al., 2004). The resulting lack of autoinhibition triggers 5-HT release at axon terminals

(Gibbons et al., 2012).

Most of the existing studies using phMRI applied functional MRI and investigated task related

blood oxygen dependent level (BOLD) responses. Evidence from phMRI and VBM, which

addresses the impact of pharmaceuticals on gray matter structure, is scarce. Yet, this

technique is a powerful tool that is able to detect morphological alterations in vivo at high

resolution. Recent work has confirmed gray matter alterations detected by MRI with ex vivo

57

MRI scans as well as post-mortem volumetric analysis (Vernon et al., 2011), and structural

MRI exhibits an excellent test-retest reliability (Wonderlick et al., 2009). In vivo gray matter

changes found by MRI and VBM were validated by post mortem findings (Hornberger et al.,

2012).

Taken together, a convergent line of evidence demonstrates that 5-HT is involved in

development and regulation of gray matter morphology through a series of mechanisms

associated with neuroplasticity. Treatment with SSRIs might thus trigger gray matter

changes, yet confirmation in healthy subjects is missing and the impact of regionally altered

gray matter on neuronal functionality is hardly known. Hence, the aim of this study was to (1)

investigate the influence of 5-HT on gray matter and (2) to elucidate the associated functional

neuronal network changes. This was accomplished by administration of SSRIs to healthy

subjects followed by structural and functional phMRI with quantification of gray matter

changes through VBM and assessment of neuronal networks through resting functional

connectivity (rFC) analyses.

2. MATERIALS AND METHODS

2.1. Subjects

A longitudinal, crossover, double-blind, placebo-controlled study design was used. The study

sample is part of a previously published fMRI study (Windischberger et al., 2010), yet all

analyses were previously not considered. Twenty-four healthy adult subjects were recruited

by advertisement at community boards at the General Hospital in Vienna, four subjects did

not meet inclusion criteria or refused to participate, 20 subjects were randomized, two

subjects dropped out (not related illness, non-compliance) and structural data from one

subject was not available for all three points. Hence, structural MRI datasets were available

from 17 healthy Caucasian subjects (6 female, 11 male 26.5±6.1 years, mean±SD, see Table

1). All subjects provided written informed consent and received reimbursement after

58

participation. All subjects underwent a medical examination at the screening visit that

included medical history, electrocardiogram and routine blood tests. Exclusion criteria were

history of severe disease, any psychiatric (according to assessment by Structured Clinical

Interview for DSM-IV Axis I+II Disorders, SCID I+II) or neurological disorder, drug abuse

including anabolic steroids, psychiatric medication, use of hormonal contraceptives for the

past 6 months, and a positive urine pregnancy test. All subjects were naïve to SSRIs and

psychotropic medication. No particular menstrual phase for scanning of female subjects was

defined. The interventions ended with a final check-up visit for each participant. All study

related procedures were approved by the Ethics Committee of the Medical University of

Vienna.

2.2. Study design and medication

All study subjects received 10 mg escitalopram (S-citalopram), an equivalent dosage of

20 mg citalopram (the 1:1 racemic mixture of R-citalopram and S-citalopram) and placebo, in

randomized order respectively, for 10 days prior to MRI scanning. This period of medication

intake was chosen to reach the plasma steady-state condition (Kasper et al., 2009; Klein et

al., 2007). Study subjects consecutively underwent three MRI scanning sessions (one after

citalopram, escitalopram and placebo) with an average interval of 21.8±13.0 (mean, SD)

between screening visit and MRT1 (no wash-out period), 33.8±6.5 days between MRT1 and

MRT2 and 33.1±4.9 between MRT2 and MRT3. According to the half-lives of citalopram and

escitalopram (Bezchlibnyk-Butler et al., 2000; Rao, 2007), visit intervals have provided

enough time to ensure previous drug/placebo washout. Treatment adherence was

ascertained by announcing control of medication intake through measurements of plasma-

levels at any given time point during study duration. Color-matched dextrose tablets were

used as placebo. In order to blind all study personnel and participants to medication group

assignment, independent pharmacists prepared the medication in accordance with a

computer generated randomization list and each blister was encoded with a unique number

59

to prevent inferences on treatment type and subject. For quantification of plasma levels,

blood samples were taken from each subject approximately 10 min before each fMRI

session. Plasma was frozen at -20 °C and shipped for analysis (Quintiles Analytical Services,

Sweden).

2.3. MRI Measurements and Image Analyses

Structural MRI measurements were performed on 3 Tesla (T) whole−body MEDSPEC S300

MR−scanner (Bruker BioSpin, Ettlingen, Germany) using a standard quadrature single-loop

transmit/receive birdcage head coil at the MR Center of Excellence at the Medical University

of Vienna, Austria. The imaging protocol comprised a magnetization−prepared rapid gradient

echo (MPRAGE, T1−weighted) sequence yielding 128 slices, a 256×256 matrix, at a slice

thickness of 1.56 mm and a voxel size of 0.78×0.86 mm.

Additionally, all study subjects underwent fMRI with a facial expression task (described

below), of which the fMRI results were published previously (Hahn et al., 2009). In the same

MRI session a single-shot gradient-recalled echo planar imaging (GR-EPI) sequence was

applied, optimized for imaging blood oxygen dependent (BOLD) contrast. This EPI-sequence

was done at a TE=31 ms, TR=1000 ms and a matrix size=128×91, which resulted in a total

slab width of 34.5 mm with 10 axial slices of 3 mm thickness aligned to the AC-PC line (0.5

mm slice gap).

2.4. Voxel-based morphometry

In order to test the main hypothesis of this study, which was to detect alterations of gray

matter after 5-HT reuptake inhibition, we used VBM for structural brain assessment. All

analyses of images were performed with statistical parametric mapping (SPM8, Wellcome

Trust Centre for Neuroimaging, Institute of Neurology, University College London, London,

60

United Kingdom, http://www.fil.ion.ucl.ac.uk/spm/software/spm8/) and MATLAB 7.10 (Math

Works, Natick, MA). An optimized VBM protocol was used, applying the DARTEL

(Diffeomorphic Anatomical Registration using Exponentiated Lie algebra) algorithm

(Ashburner, 2007). The images were segmented into gray matter, white matter, and

cerebrospinal fluid (CSF) compartments and successfully passed visually checking for major

artifacts. Subsequently, the gray matter maps obtained by this procedure were separately

normalized to a gray matter template representing the stereotaxic standardized Montreal

Neurological Institute (MNI) space at a voxel size of 1.5 × 1.5 × 1.5 mm. Based on

deformation fields calculated during segmentation, a template was generated by the

DARTEL algorithm. The Jacobian determinants derived from the spatial normalization were

used during nonlinear spatial transformations, which modulated the regional differences of

partitioned gray matter on images from relative to absolute amounts (volume). Unmodulated

images constitute gray matter density (GMD), while modulated images constitute gray matter

volume (GMV). Gray matter images were smoothed with a Gaussian filter of 8 × 8 × 8 mm

full width at half maximum (FWHM).

2.5. Preprocessing Resting Functional Connectivity

Resting functional connectivity was determined using a method to separate network-specific

low frequency BOLD (LF-BOLD) signals from task-evoked BOLD responses (Fair et al.,

2007). Intrinsic connectivity networks are present during task performance (Fox et al., 2006)

so that the obtained resting activity mimics real-life setting, where the brain is usually

continuously engaged in task processing. So gathered intrinsic network activity at rest offers

a reliable alternative to resting-state fMRI at the advantage of controlling for inter- and

intraindividual variations in thought processes and mentation (Shih et al., 2011).

Functional MRI data preprocessing comprised slice-timing correction, realignment, spatial

normalization and spatial smoothing as implemented in SPM8. An in-house, scanner-specific

EPI template was created from previous data by spatial normalization of each individual scan

61

to the SPM template which was followed by averaging across individuals. The advantage of

using an in-house template is that potential local field inhomogeneities of individual scans

match those of the template, which in turn improves the spatial normalization. This template

was used for normalization to MNI-space and a Gaussian smoothing kernel of 8 × 8 × 8 mm

FWHM for spatial smoothing.

Resting functional connectivity was extracted from a blocked facial fMRI paradigm (Hahn et

al., 2009), consisting of 20s baseline blocks, during which a black screen with a white fixation

cross at the center was presented. Baseline blocks were alternated by 20s presentation of

faces as active task blocks, whereby the facial expression and attractiveness had to be

rated. The entire paradigm consisted of 5 alternating baseline and active paradigm blocks

leading to total paradigm duration of 200 s. In accordance with Fair et al (Fair et al., 2007),

the active task periods were removed by cutting out the five 20 s baseline periods. In

consideration of the delay in the hemodynamic response function, cuts were set 5 s after

active and resting blocks, respectively, leading to a time shift of 5 s. This number was used to

maximize the number of frames within steady-state data.

2.6. Resting Functional Connectivity

Because both SSRIs and brain changes underlying VBM findings have previously been

demonstrated to alter brain connectivity (Reetz et al., 2012; van Marle et al., 2011), we were

particularly interested in differences in brain rFC associated with a principal VBM finding

between the SSRI and placebo group. Hence, seed voxel correlation analysis was performed

with resting data extracted from the FEDT fMRI paradigm as described above. Resting data

were corrected as published in an earlier study (Weissenbacher et al., 2009). Linear

regression was applied to correct for changes in white-matter, ventricular and global signal.

Then, data were band-pass filtered by a 12-term finite impulse response (FIR) filter to 0.009-

0.08 Hz with Matlab. Connectivity maps were calculated by cross-correlation between the

BOLD time course from the seed region obtained by VBM (see results section) and the time

62

course from the remaining voxels of the entire brain. For group comparisons, correlation

maps were converted to z-values using Fisher’s r-to-z transformation.

2.7. Resting Functional Connectivity Density

The brains’ resting activity has recently been shown to be subdivided into functional “hubs”

representing network nodes with high functional connectivity (Bullmore and Sporns, 2009),

which differ between their resting activity and anatomical topology (Tomasi and Volkow,

2011). We hypothesized that the brain’s functional hubs are changed themselves by 5-HT

reuptake inhibition. To investigate this question we applied resting functional connectivity

density (rFCD) mapping. This recently developed functional connectivity analysis to define

functional network hubs is a fast voxel-wise data-driven approach sensitive to the number of

local functional connections in brain regions (Tomasi and Volkow, 2010; Tomasi and Volkow,

2011). Technically, functional hubs represent network nodes with a large number of edges as

defined by graph theory. This approach yields FCD, in other words the node degree, and

resembles the cross-correlation function of every voxel. Functional connectivity density has a

‘‘scale-free’’ distribution in the brain (Tomasi and Volkow, 2010; Tomasi and Volkow, 2011),

with few hubs and numerous weakly connected nodes, consistent with the emergence of

scaling in neural networks (He et al., 2010).

To reduce spatial dimensionality for more efficient computation of the BOLD time series,

rFCD maps were downsampled by spline interpolation to a resolution of 4 × 4 × 4 mm. Only

gray matter voxels were further processed using a gray matter mask. White-matter,

ventricular and global signal were regressed-out and the time series were detrended by a 4th

order polynomial function, which was chosen because of optimal results after visually

inspecting the plots of the time-series. For FCD, a connectivity matrix with Pearson

correlation coefficients was generated and a threshold was set at the 1% strongest

correlations in each graph, the remaining voxels were set to zero (Fornito et al., 2012).

Functional connectivity density maps were generated by summing the number of

63

connections within the connectivity matrix for each voxel separately, corresponding to global

FCD as defined previously (Tomasi and Volkow, 2010).

2.8. Statistical Analyses

To decrease the variance in the verum condition, we combined citalopram and escitalopram

groups by averaging the effects within repeated-measure analysis of variance (rmANOVA)

design (i.e., the average of the two verum conditions was equally weighted as the placebo

condition). Three main analyses were performed: one for VBM (1), rFC (2) and rFCD (3),

respectively.

(1) In the main VBM statistical analysis a contrast for gray matter density (GMD) differences

between structural MRI scans after SSRIs (pooled citalopram and escitalopram scans) and

those after placebo was defined by rmANOVA as implemented in SPM8. In this analysis we

controlled for total GMD (tGMD) and sex as two additional factors. Results were corrected

applying the voxel-level family-wise error (FWE) rate at a significance level of Pcorr<0.05.

Only results reaching a cluster-size above 100 voxels are reported. An identical analysis was

performed with gray matter volume (GMV), stronger results are reported.

(2) Proceeding from the major VBM finding, the cluster coordinates were used as seed in

functional connectivity statistics. Seed voxel rmANOVA with rFC maps was performed in

SPM8 and contrasted between SSRI intake (pooled) and placebo condition. Total GMD and

sex were controlled as two additional factors. Results were corrected for false positives using

FWE-correction at a significance level of Pcorr<0.05.

(3) Finally, group differences in rFCD maps from scans after SSRI intake (pooled) and

placebo intake were calculated with rmANOVA in SPM8 and again controlled for sex and

tGMD. Results were corrected for false positives using FWE-correction at a significance level

of Pcorr<0.05.

64

Follow-up Pearson correlations were calculated between principal voxel-based morphometry

(VBM) finding and selective serotonin reuptake inhibitor (SSRI) plasma levels, to test the

relationship between plasma levels and gray matter changes. Furthermore, to investigate a

more global effect of SSRI plasma levels on VBM-results, we performed an identical

rmANOVA controlling for SSRI plasma levels, only. Sex and gray matter density (GMD) were

not added here, because the number of variables roughly should not exceed N/10 to avoid

overfitting.

To test the validity of the extracted resting data and subsequent rFCD analysis, we aimed to

depict major FCD hubs as previously demonstrated (Tomasi and Volkow, 2011). Therefore, in

an intermediate step before SSRI vs. placebo analysis we calculated a one-sample t-test

over rFCD maps of subjects receiving placebo only. Results were corrected with voxel-level

FWE-correction at a significance level of Pcorr<0.05.

Additional statistical tests were used: to rule out cortical atrophy as source of variance and to

investigate global gray matter differences between SSRIs and placebo. Subject gender

differences were calculated with independent sample T-test or Mann-Whitney U test where

appropriate.

We aimed to exclude cortical atrophy due to aging as a potential source of variance. Since

constant covariates across the scans are not part of the general linear model in SPM and

could therefore not be included in the main rmANOVA analyses, the linear effect was

computed ([citalopram+escitalopram]/2-placebo) between the different scanning sessions

resulting in a separate map for every subject. The subsequent one-sample t-test across

these maps is equal to the contrast in the rmANOVA reported above. Hence, the linear effect

maps could be used in a regression analysis with age as independent variable to investigate

the effects of age on VBM. Age effects were corrected using voxel-level FWE-correction at a

significance level of Pcorr<0.05.

Additionally, we aimed to compare global gray matter across treatment with SSRIs or

placebo. Therefore, we performed a repeated-measure analysis of variance (rmANOVA) in

65

SPSS 19.0 comparing total GMD after SSRIs (tGMD=citalopram+escitalopram/2) or placebo

intake using sex as between-subjects variable and age as covariate. A statistical level of

significance was accepted at Puncorr<0.001.

3. RESULTS

Age, body mass index, total brain gray matter, alcohol and cigarette consumption were not

significantly different between males and females (all P>0.1, Table 1). No significant effect of

age was observed between the linear-effect maps of SSRI vs. placebo condition and age (all

FWE Pcorr>0.05. Comparison of total gray matter revealed no significant effect of SSRI

treatment on total brain GMD (Puncorr=0.471).

3.1. Voxel-based morphometry differences between SSRIs and Placebo

The main VBM analysis revealed significant increases of GMD after oral intake of SSRIs

compared to placebo. Gray mater increases were found in the posterior cingulate cortex

(PCC), the ventral precuneus (peak T-values=16.6), the fusiform gyrus, the insula and the

medial superior frontal cortex (all regions bilaterally, 8.1<T<16.6, all FWE Pcorr <0.05; for all

regions see Figure 1 and Table 2). There was no significant correlation between GMD and

SSRI plasma levels (r=0.27, P=0.278) in the PCC cluster. Topologically similar results were

obtained after correcting for SSRI plasma levels (data not shown). However, in the PCC and

precuneus, T-values were attenuated and cluster size decreased (T=6.3, FWE Pcorr=0.012)

Furthermore, significant decreases of GMD after SSRI intake were observed bilaterally in the

precentral gyrus, the cerebellum and the cingulate cortex (8.4<T<13.4, all FWE Pcorr<0.05,

see Table 3). Gray matter density and GMV results were topologically comparable (data not

shown).

66

Figure 1: Increases of gray matter density in 17 healthy subjects after 10 days of SSRI intake

compared to placebo in the bilateral posterior cingulate cortex and adjacent ventral precuneus as

measured with structural MRI and voxel-based morphometry. Color bar represents T-values displayed

at P<0.05 (FWE-corrected). Numbers represent coordinates in MNI standard space at the location of

the crosshair, warm color tones represent increases.

3.2. Seed-voxel rFC differences between SSRIs and Placebo

The PCC is a known hotspot of neuronal activity and exhibited gray matter increases in the

VBM results. Hence, the PCC cluster was chosen for further analysis and subsequent

discussion. The PCC result from VBM served as seed region (VBM peak MNI: x,y,z = 8, -45,

38; 3261 voxels) for calculating differences in rFC between the SSRI and placebo group. In

the voxel-wise rFC analysis, enhanced connectivity from the PCC seed region was observed

after SSRI intake compared to placebo in the bilateral PCC spreading to the ventral

precuneus (T=5.7, FWE Pcorr<0.05, Figure 2A). The resulting rFC cluster in the PCC/ventral

precuneus was located overlapping and caudally adjacent to the VBM seed region (Figure

2C). No other regions exhibiting significant positive or negative rFC alterations. Applying less

stringent correction, increased rFC was obtained in the cuneus (Puncorr<0.001, T=3.6, 40

voxels).

3.3. FCD connectivity hub changes under SSRI administration

The extraction of resting activity and subsequent rFCD analysis at baseline levels of block

design fMRI data in subjects after intake of placebo yielded typical rFCD hubs as reported

67

Figure 2: Increased resting functional connectivity (rFC) and resting functional connectivity density

(rFCD) associated with increased gray matter (VBM) in the PCC after SSRI intake compared to

placebo. (A) Increased seed-voxel rFC associated with increased gray matter in the PCC (VBM peak

MNI: x,y,z = 8, -45, -19; 3261 voxels) to gray matter within small parts of the PCC and caudally

adjacent in gray matter of the ventral precuneus. (B) Increased rFCD in subjects after SSRI intake

compared to placebo in the PCC and ventral precuneus. (C) Triple overlay indicating close proximity of

increased gray matter, rFC and rFCD in the PCC and ventral precuneus, known to represent one of

the brain’s metabolically most active region (Gusnard and Raichle, 2001). Blue color bars represent

rFC T-values at P<0.05 (FWE-corrected), displayed at Puncorr<0.001. Purple color bars represent rFCD

T-values at P<0.05 (FWE-corrected), displayed at Puncorr<0.001. Spectral color bar represents VBM T-

values displayed at P<0.05 (FWE-corrected). Numbers represent coordinates in MNI standard space

at the location of the crosshair.

68

Figure 3: Resting functional connectivity density (rFCD) hubs in 17 subjects receiving placebo.

Resting functional connectivity was extracted from baseline blocks of a previously published block

design fMRI study (Windischberger et al., 2010) and further analyzed by rFCD mapping. The

topological pattern matches previously published large datasets (Tomasi and Volkow, 2011) and

demonstrates activation of typical resting-state networks in the cuneus/precuneus, cingulate cortex

and medial frontal cortex. Color bar represents T-values displayed at P<0.05 (FWE-corrected) and

numbers represent coordinates in MNI standard space at the location of the crosshair.

previously (Tomasi and Volkow, 2011), specifically, in the brain’s visual- and default mode

network (8.4<T<15.2, all FWE Pcorr<0.05, see Figure 3).

The enhanced VBM and rFC signals in the PCC and adjacent ventral precuneus are located

in and around the significant rFCD hub observed in scans after placebo intake (Figure 3).

Compared to placebo, whole-brain rFCD was significantly increased after SSRI-intake in the

ventral precuneus spreading to the PCC (T=4.3, FWE-Pcorr<0.05, Figure 2B). We observed

no other regions exhibiting significant FCD alterations.

4. DISCUSSION

Our main finding is the localization of dynamic gray matter changes in healthy subjects after

10 days of SSRI intake. These results were independent of VBM gray matter modality (GMD

or GMV), controlled for sex and total gray matter and not associated with age. Increased

69

gray matter did not correlate with SSRI plasma levels, statistical significance was, however

attenuated upon correcting for plasma levels. Total gray matter was not significantly different

between treatment groups. Furthermore, we observed increased functional connectivity after

SSRI intake associated with increased gray matter in the PCC within the PCC and to

adjacent gray matter in the ventral precuneus. Connectivity density, a measure for the brain’s

functional network hubs, was increased after SSRI intake in a topologically identical region in

the PCC/ventral precuneus. Taken together, our results provide consistent evidence that

SSRI intake is associated with changes of gray matter and neuronal functionality as

measured by phMRI.

Our findings are striking for two apparent reasons. First, although differently calculated, both

increased rFCD and rFC spatially overlap in the PCC/ventral precuneus. Enhanced

functional connectivity seeded from increased PCC gray matter was validated by whole brain

connectivity density, which resembles whole brain correlational strength of LF-BOLD signals

without the need of seeds. Second, these brain structures exhibit large neuronal densities

and are amongst the brain’s metabolically most active (Gusnard and Raichle, 2001). Peak

values in blood flow, metabolic activity and oxygen turnover are located there, which all might

impact changes of structural and functional MRI signals.

We observed several regions with significant gray matter increases, such as the fusiform

gyrus, the insula, and the frontal cortex, and the posterior cingulate cortex where

serotonergic neurotransmission might impact on structural plasticity as measured with MRI.

Furthermore, several decreases were found, such as in the pre- and postcentral gyri. The

principal neurobiological mechanisms effecting volumetric changes in the living brain as

reported by numerous MRI studies are currently being investigated with large effort. The

findings revealed by this study appear very interesting because they indicate a role of 5-HT in

changing gray matter and functional neuronal activity. Decreased metabolism was

discovered at one week and reversal towards increased metabolism after 6 weeks of

treatment in the posterior cingulate cortex, hippocampus, insula, putamen as well as in

70

temporal and prefrontal cortices (Mayberg et al., 2000). This was explained by the authors to

be caused by receptor downregulation or changes in second messenger systems and further

undermines the notion that SSRIs impact on energy metabolism (Webhofer et al., 2011).

Furthermore, response dependent differences in regional blood flow were demonstrated in

depressive patients in the PCC (Joe et al., 2006). Beyond this, previous evidence from

animal research enables a brief discussion on 5-HT mediated neuroplasticity. Hypothetically,

SSRI-dependent altered plasticity might occur due to binding at serotonergic receptors,

which are linked to second messenger systems effecting restructuration. Candidate

molecules potentially mediating such remodeling in the PCC and ventral precuneus are 5-

HT1A, 5-HT1B and 5-HT2A receptor (Savli et al., 2012) and cross-links to the neurotrophin

system with prominent targets such as BDNF and CREB. Unfortunately, MRI data at the

current spatial resolution do not allow inference on the kind of neuronal reactivity or the type

of cellular remodeling that cause gray matter signal changes. A number of factors such as

altered neuronal or glial reactivity (Lesch and Waider, 2012), liquor circulation, blood flow and

angiogenesis (Seevinck et al., 2010) might likewise account for brain volume changes and

subsequently altered neuronal functionality.

Our data show that changes of regional gray matter MRI signals are also associated with

altered functional activity. Higher amount of gray matter in the PCC was associated with

increased resting activity in the PCC itself and the adjacent ventral precuneus. This region is

a key node of the default mode network and differentially active during learning, memory,

reward and task engagement (for review see (Pearson et al., 2011)). We demonstrated that

the brain’s functional connectivity in our dataset is compartmentalized in resting network

hubs at locations in line with earlier studies (Tomasi and Volkow, 2010; Tomasi and Volkow,

2011). The anatomical localization and functional activation of these hubs is consistently

reported (Gusnard and Raichle, 2001; Tomasi and Volkow, 2010; Tomasi and Volkow, 2011),

though, according to our study, SSRIs might alter the activity of such hubs. In consideration

of the fact that multiple imaging modalities detected increases in gray matter, altered

functional connectivity and hub activity in the PCC/ventral precuneus, this study provides

71

robust evidence that SSRIs interfere with a broad cascade of mechanisms affecting several

physiological brain systems. Moreover, these data are indicative that functional changes

occur in concert with structural alterations, even after short treatment periods. However, our

study design does not allow inference on the length of time the observed network changes

last. Previous reports indicate that structural modifications usually take more time than

changes in functionality, yet demand further research on the structure-function relationship of

brain networks (Bullmore and Sporns, 2009), which emphasizes the relevance of our

findings.

We are not aware of a similar study in healthy subjects investigating the impacts of SSRIs on

neuronal structure and function by combining structural and functional MRI. Increases of gray

matter have been recently published in studies using phMRI in psychiatric patients

undergoing SSRI therapy. A coinciding longitudinal study using the DARTEL algorithm shows

increases of gray matter in the dorsolateral prefrontal cortex after intake of sertraline (Smith

et al., 2012). Yet herein the authors investigated depressed patients over a time course of 12

weeks. Moreover, gray matter increases were reported for the left putamen after 12 week

intake of fluoxetine in patients with obsessive compulsive disorder (Hoexter et al., 2012).

Another coinciding study investigated patients with posttraumatic stress disorder and found

hippocampal volume increases after 9-12 months continuous treatment with paroxetine

(Vermetten et al., 2003). Gray matter increases after SSRI intake are further supported by a

study in first-episode drug-naïve depressive disorder and panic disorder, detailing moderate

increases in subcortical structures such as the nucleus accumbens, the putamen and the

hippocampus (Lai and Wu, 2011). On the other side, a cross-sectional study with patients

suffering from depression and anxiety disorders did not reveal a protecting effect of stable

SSRI treatment on gray matter reductions observed in these disorders (van Tol et al., 2010).

In consideration of neuroplastic effects of 5-HT, the observed decreases of gray matter in our

study are challenging to interpret, but potentially the same mechanisms mediating gray

matter increases could be involved. Only one study reported gray matter decreases, located

in the superior temporal cortex in patients with social anxiety disorder after a 12 week-long

72

intake of 20 mg escitalopram (Cassimjee et al., 2010). Taken together, different patient

collectives drug types and duration in relation to our study impairs comparability.

Until now, rFC alterations upon SSRI challenge were only scarcely investigated. A study in

healthy subjects identified decreased functional coupling between the amygdala and the

ventral medial prefrontal cortex (McCabe and Mishor, 2011). Enhanced functional coupling

was reported between the amygdala and the anterior insula after duloxetine intake (van

Marle et al., 2011), here 19 healthy subjects were analyzed. VBM-based seed regions were

not considered in either of these studies. Noteably, rFC and rFCD changes after SSRI intake

were regionally restricted to intrinsic areas around increased gray matter in the PCC.

Cognitive load adherent in blocked design task activation was previously located at the PCC

(Newton et al., 2011). Furthermore, transient BOLD responses at block transitions occur at

the PCC and many other regions (Fox et al., 2005), yet inserted intervals accounting for

delay in the hemodynamic response function limit these transitions. Variance from cognitive

load and transitions might theoretically spill into the analyzed baseline blocks, which should

be taken into account upon comparison of our data with traditionally obtained resting-state

data. Hence, it remains intriguing that whole-brain connectivity density analysis, which is not

related to seed-based connectivity, identifies the same region as connectivity from increased

gray matter signal. The mechanisms that SSRIs interfere with in this region are therefore

likely to be associated with several factors altering substrates detectable both in T1-weighted

as well as EPI MRI sequences. The existing gap between underlying molecular mechanisms

and alterations of voxel-intensity values are vigorously debated (for critical review and

comments see (Draganski and Kherif, 2013; Erickson, 2013; Fields, 2013; Thomas and

Baker, 2013), so that strong gray matter changes as shown in this study emphasize the need

for more translational work on molecular players mediating in vivo structural and functional

changes of neuronal networks as measured by MRI.

The following study limitations must be reported. Though we analyzed a rather low subject

number, sample sizes of active groups within previous studies have been even lower than in

73

our study (Anand et al., 2005; McCabe and Mishor, 2011). Therefore, this factor, though

indeed a limitation, remains a common feature in many pharmacologic neuroimaging studies.

In addition, subjects were not balanced according to sex, this issue was however addressed

by including sex as nuisance variable.

In summary, we found that study subjects after SSRI intake exhibited significant gray matter

changes. Moreover, almost identical locations of increased resting functional connectivity and

connectivity density associated with gray matter increases in the PCC provide evidence for

the involvement of SSRIs in multiple mechanisms changing brain structure and functionality.

When taken together, these results point towards plastic changes of brain structure and

function as neuronal substrate of effects associated with SSRI intake and offer a paradigm

for further exploration of these mechanisms in psychiatric patients.

5. ACKNOWLEDGEMENTS

Data have been measured within a project funded by an investigator-initiated and

unrestricted research grant from H. Lundbeck A/S, Denmark to S. Kasper. The sponsors and

funders did not participate in the design and conduct of the study and were not involved in

the preparation, review, or approval of the manuscript. The study protocol has been planned

by the authors who retained full academic control. In the study presented here we applied

new data analysis approaches in structural and functional magnetic resonance imaging

recently available beyond the scope of a study already published (Windischberger et al.,

2010). The work of C. Kraus has been funded by an intramural grant of the research cluster

between the Medical University of Vienna and the University of Vienna (FA103FC001) to R.

Lanzenberger and C. Lamm. A. Hahn was funded by a DOC fellowship of the Austrian

Academy of Sciences at the Department of Psychiatry and Psychotherapy. The authors are

grateful to C. Spindelegger, U. Moser, P. Stein, M. Fink, L. Pezawas, A. Erfurth, and M. Willeit

for their medical support, and to A. Holik, S. Friedreich, F. Gerstl, and E. Moser for technical

74

support. We thank M. Spies for native English editing. The study is part of C. Kraus’ thesis

“Serotonin and Neuroplasticity” supervised by R. Lanzenberger in the Clinical Neurosciences

PhD program at the Medical University of Vienna, Austria. Parts of this study have been or

will be presented by P. Baldinger at the 19th European Congress of Psychiatry (EPA), March

12-15, 2011, Vienna, Austria, by M. Savli at the 24thEuropean College of

Neuropsychopharmacology (ECNP) Congress, September 3-7, 2011, Paris, France, and by

C. Kraus at the 11th World Congress of Biological Psychiatry (WFSBP), June 23-27, 2013,

Kyoto, Japan.

6. CONFLICT OF INTEREST

Without any relevance to this work, S. Kasper declares that he has received grant/research

support from Eli Lilly, Lundbeck A/S, Bristol-Myers Squibb, Servier, Sepracor,

GlaxoSmithKline, Organon, and has served as a consultant or on advisory boards for

AstraZeneca, Austrian Sick Found, Bristol-Myers Squibb, GlaxoSmithKline, Eli Lily, Lundbeck

A/S, Pfizer, Organon, Sepracor, Janssen, and Novartis, and has served on speakers’

bureaus for AstraZeneca, Eli Lilly, Lundbeck A/S, Servier, Sepracor and Janssen. R.

Lanzenberger received travel grants and conference speaker honoraria from AstraZeneca

and Lundbeck A/S.

75

REFERENCES Aigner, M., Treasure, J., Kaye, W., Kasper, S., Disorders, W.T.F.O.E., 2011. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the pharmacological treatment of eating disorders. World J Biol Psychiatry 12, 400-443.

Anand, A., Li, Y., Wang, Y., Wu, J., Gao, S., Bukhari, L., Mathews, V.P., Kalnin, A., Lowe, M.J., 2005. Antidepressant effect on connectivity of the mood-regulating circuit: an FMRI study. Neuropsychopharmacology 30, 1334-1344.

Ashburner, J., 2007. A fast diffeomorphic image registration algorithm. NeuroImage 38, 95-113.

Bandelow, B., Zohar, J., Hollander, E., Kasper, S., Moller, H.J., Wfsbp Task Force on Treatment Guidelines for Anxiety, O.-C., Post-Traumatic Stress, D., Allgulander, C., Ayuso-Gutierrez, J., Baldwin, D.S., Buenvicius, R., Cassano, G., Fineberg, N., Gabriels, L., Hindmarch, I., Kaiya, H., Klein, D.F., Lader, M., Lecrubier, Y., Lepine, J.P., Liebowitz, M.R., Lopez-Ibor, J.J., Marazziti, D., Miguel, E.C., Oh, K.S., Preter, M., Rupprecht, R., Sato, M., Starcevic, V., Stein, D.J., van Ameringen, M., Vega, J., 2008. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the pharmacological treatment of anxiety, obsessive-compulsive and post-traumatic stress disorders - first revision. World J Biol Psychiatry 9, 248-312.

Bauer, M., Bschor, T., Pfennig, A., Whybrow, P.C., Angst, J., Versiani, M., Möller, H.-J., Disorders, W.T.F.o.U.D., 2007. World Federation of Societies of Biological Psychiatry (WFSBP) Guidelines for Biological Treatment of Unipolar Depressive Disorders in Primary Care. World J Biol Psychiatry 8, 67-104.

Benninghoff, J., van der Ven, A., Schloesser, R.J., Moessner, R., Moller, H.J., Rujescu, D., 2012. The complex role of the serotonin transporter in adult neurogenesis and neuroplasticity. A critical review. World J Biol Psychiatry 13, 240-247.

Bezchlibnyk-Butler, K., Aleksic, I., Kennedy, S.H., 2000. Citalopram--a review of pharmacological and clinical effects. J Psychiatry Neurosci 25, 241-254.

Bullmore, E., Sporns, O., 2009. Complex brain networks: graph theoretical analysis of structural and functional systems. Nat Rev Neurosci 10, 186-198.

Cassimjee, N., Fouche, J.-P., Burnett, M., Lochner, C., Warwick, J., Dupont, P., Stein, D.J., Cloete, K.J., Carey, P.D., 2010. Changes in regional brain volumes in social anxiety disorder following 12 weeks of treatment with escitalopram. Metab Brain Dis 25, 369-374.

Castrén, E., Rantamäki, T., 2010. The role of BDNF and its receptors in depression and antidepressant drug action: Reactivation of developmental plasticity. Dev Neurobiol 70, 289-297.

Daubert, E.A., Condron, B.G., 2010. Serotonin: a regulator of neuronal morphology and circuitry. Trends Neurosci 33, 424-434.

Draganski, B., Kherif, F., 2013. In vivo assessment of use-dependent brain plasticity-Beyond the "one trick pony" imaging strategy. NeuroImage 73, 255-259.

Erickson, K.I., 2013. Evidence for structural plasticity in humans: Comment on Thomas and Baker (2012). NeuroImage 73, 237-238.

Fair, D.A., Schlaggar, B.L., Cohen, A.L., Miezin, F.M., Dosenbach, N.U., Wenger, K.K., Fox, M.D., Snyder, A.Z., Raichle, M.E., Petersen, S.E., 2007. A method for using blocked and

76

event-related fMRI data to study "resting state" functional connectivity. NeuroImage 35, 396-405.

Fields, R.D., 2013. Changes in brain structure during learning: Fact or artifact? Reply to Thomas and Baker. NeuroImage 73, 260-264.

Fornito, A., Zalesky, A., Pantelis, C., Bullmore, E.T., 2012. Schizophrenia, neuroimaging and connectomics. NeuroImage 62, 2296-2314.

Fox, M.D., Snyder, A.Z., Barch, D.M., Gusnard, D.A., Raichle, M.E., 2005. Transient BOLD responses at block transitions. NeuroImage 28, 956-966.

Fox, M.D., Snyder, A.Z., Zacks, J.M., Raichle, M.E., 2006. Coherent spontaneous activity accounts for trial-to-trial variability in human evoked brain responses. Nat Neurosci 9, 23-25.

Frodl, T., Koutsouleris, N., Bottlender, R., Born, C., Jäger, M., Mörgenthaler, M., Scheuerecker, J., Zill, P., Baghai, T., Schüle, C., Rupprecht, R., Bondy, B., Reiser, M., Möller, H.-J., Meisenzahl, E.M., 2008. Reduced gray matter brain volumes are associated with variants of the serotonin transporter gene in major depression. Mol Psychiatry 13, 1093-1101.

Gaspar, P., Cases, O., Maroteaux, L., 2003. The developmental role of serotonin: news from mouse molecular genetics. Nat Rev Neurosci 4, 1002-1012.

Gibbons, R.D., Hur, K., Brown, C.H., Davis, J.M., Mann, J.J., 2012. Benefits from antidepressants: synthesis of 6-week patient-level outcomes from double-blind placebo-controlled randomized trials of fluoxetine and venlafaxine. Arch Gen Psychiatry 69, 572-579.

Gould, E., 1999. Serotonin and hippocampal neurogenesis. Neuropsychopharmacology 21, 46S-51S.

Gusnard, D.A., Raichle, M.E., 2001. Searching for a baseline: functional imaging and the resting human brain. Nat Rev Neurosci 2, 685-694.

Hahn, A., Holik, A., Gerstl, F., Savli, M., Stein, P., Akimova, E., Angleitner, P., Windischberger, C., Kasper, S., Lanzenberger, R., 2009. Anxiety Scores are Related to Amygdala Activity Induced by Facial Attractiveness and Emotional Expressions. NeuroImage 47 48S.

He, B.J., Zempel, J.M., Snyder, A.Z., Raichle, M.E., 2010. The temporal structures and functional significance of scale-free brain activity. Neuron 66, 353-369.

Hoexter, M.Q., de Souza Duran, F.L., D'Alcante, C.C., Dougherty, D.D., Shavitt, R.G., Lopes, A.C., Diniz, J.B., Deckersbach, T., Batistuzzo, M.C., Bressan, R.A., Miguel, E.C., Busatto, G.F., 2012. Gray matter volumes in obsessive-compulsive disorder before and after fluoxetine or cognitive-behavior therapy: a randomized clinical trial. Neuropsychopharmacology 37, 734-745.

Hornberger, M., Wong, S., Tan, R., Irish, M., Piguet, O., Kril, J., Hodges, J.R., Halliday, G., 2012. In vivo and post-mortem memory circuit integrity in frontotemporal dementia and Alzheimer's disease. Brain 135, 3015-3025.

Joe, A.Y., Tielmann, T., Bucerius, J., Reinhardt, M.J., Palmedo, H., Maier, W., Biersack, H.J., Zobel, A., 2006. Response-dependent differences in regional cerebral blood flow changes with citalopram in treatment of major depression. J Nucl Med 47, 1319-1325.

Kasper, S., Sacher, J., Klein, N., Mossaheb, N., Attarbaschi-Steiner, T., Lanzenberger, R., Spindelegger, C., Asenbaum, S., Holik, A., Dudczak, R., 2009. Differences in the dynamics of

77

serotonin reuptake transporter occupancy may explain superior clinical efficacy of escitalopram versus citalopram. Int Clin Psychopharmacol 24, 119-125.

Klein, N., Sacher, J., Geiss-Granadia, T., Mossaheb, N., Attarbaschi, T., Lanzenberger, R., Spindelegger, C., Holik, A., Asenbaum, S., Dudczak, R., Tauscher, J., Kasper, S., 2007. Higher serotonin transporter occupancy after multiple dose administration of escitalopram compared to citalopram: an [123I]ADAM SPECT study. Psychopharmacology (Berl) 191, 333-339.

Lai, C.H., Wu, Y.T., 2011. Duloxetine's modest short-term influences in subcortical structures of first episode drug-naive patients with major depressive disorder and panic disorder. Psychiatry Res 194, 157-162.

Lesch, K.P., Waider, J., 2012. Serotonin in the modulation of neural plasticity and networks: implications for neurodevelopmental disorders. Neuron 76, 175-191.

Mayberg, H.S., Brannan, S.K., Tekell, J.L., Silva, J.A., Mahurin, R.K., McGinnis, S., Jerabek, P.A., 2000. Regional metabolic effects of fluoxetine in major depression: serial changes and relationship to clinical response. Biol Psychiatry 48, 830-843.

McCabe, C., Mishor, Z., 2011. Antidepressant medications reduce subcortical-cortical resting-state functional connectivity in healthy volunteers. NeuroImage 57, 1317-1323.

Mogha, A., Guariglia, S.R., Debata, P.R., Wen, G.Y., Banerjee, P., 2012. Serotonin 1A receptor-mediated signaling through ERK and PKCalpha is essential for normal synaptogenesis in neonatal mouse hippocampus. Transl Psychiatry 2, e66.

Newton, A.T., Morgan, V.L., Rogers, B.P., Gore, J.C., 2011. Modulation of steady state functional connectivity in the default mode and working memory networks by cognitive load. Hum Brain Mapp 32, 1649-1659.

Pearson, J.M., Heilbronner, S.R., Barack, D.L., Hayden, B.Y., Platt, M.L., 2011. Posterior cingulate cortex: adapting behavior to a changing world. Trends Cogn Sci 15, 143-151.

Rao, N., 2007. The clinical pharmacokinetics of escitalopram. Clin Pharmacokinet 46, 281-290.

Reetz, K., Dogan, I., Rolfs, A., Binkofski, F., Schulz, J.B., Laird, A.R., Fox, P.T., Eickhoff, S.B., 2012. Investigating function and connectivity of morphometric findings--exemplified on cerebellar atrophy in spinocerebellar ataxia 17 (SCA17). NeuroImage 62, 1354-1366.

Savli, M., Bauer, A., Mitterhauser, M., Ding, Y.S., Hahn, A., Kroll, T., Neumeister, A., Haeusler, D., Ungersboeck, J., Henry, S., Isfahani, S.A., Rattay, F., Wadsak, W., Kasper, S., Lanzenberger, R., 2012. Normative database of the serotonergic system in healthy subjects using multi-tracer PET. NeuroImage 63, 447-459.

Seevinck, P.R., Deddens, L.H., Dijkhuizen, R.M., 2010. Magnetic resonance imaging of brain angiogenesis after stroke. Angiogenesis 13, 101-111.

Shih, P., Keehn, B., Oram, J.K., Leyden, K.M., Keown, C.L., Muller, R.A., 2011. Functional differentiation of posterior superior temporal sulcus in autism: a functional connectivity magnetic resonance imaging study. Biol Psychiatry 70, 270-277.

Smith, R., Chen, K., Baxter, L., Fort, C., Lane, R.D., 2012. Antidepressant effects of sertraline associated with volume increases in dorsolateral prefrontal cortex. J Affect Disord.

78

Stahl, S.M., 1998. Mechanism of action of serotonin selective reuptake inhibitors. Serotonin receptors and pathways mediate therapeutic effects and side effects. J Affect Disord 51, 215-235.

Tao, R., Ma, Z., Auerbach, S.B., 2000. Differential effect of local infusion of serotonin reuptake inhibitors in the raphe versus forebrain and the role of depolarization-induced release in increased extracellular serotonin. J Pharmacol Exp Ther 294, 571-579.

Thomas, C., Baker, C.I., 2013. Teaching an adult brain new tricks: A critical review of evidence for training-dependent structural plasticity in humans. NeuroImage 73, 225-236.

Tomasi, D., Volkow, N.D., 2010. Functional connectivity density mapping. Proc Natl Acad Sci U S A 107, 9885-9890.

Tomasi, D., Volkow, N.D., 2011. Association between Functional Connectivity Hubs and Brain Networks. Cereb Cortex, 1-11.

van Marle, H.J., Tendolkar, I., Urner, M., Verkes, R.J., Fernandez, G., van Wingen, G., 2011. Subchronic duloxetine administration alters the extended amygdala circuitry in healthy individuals. NeuroImage 55, 825-831.

van Tol, M.-J., van der Wee, N.J.A., van den Heuvel, O.A., Nielen, M.M.A., Demenescu, L.R., Aleman, A., Renken, R., van Buchem, M.A., Zitman, F.G., Veltman, D.J., 2010. Regional brain volume in depression and anxiety disorders. Arch Gen Psychiatry 67, 1002-1011.

Vermetten, E., Vythilingam, M., Southwick, S.M., Charney, D.S., Bremner, J.D., 2003. Long-term treatment with paroxetine increases verbal declarative memory and hippocampal volume in posttraumatic stress disorder. Biol Psychiatry 54, 693-702.

Vernon, A.C., Natesan, S., Modo, M., Kapur, S., 2011. Effect of chronic antipsychotic treatment on brain structure: a serial magnetic resonance imaging study with ex vivo and postmortem confirmation. Biol Psychiatry 69, 936-944.

Vitalis, T., Cases, O., Gillies, K., Hanoun, N., Hamon, M., Seif, I., Gaspar, P., Kind, P., Price, D.J., 2002. Interactions between TrkB signaling and serotonin excess in the developing murine somatosensory cortex: a role in tangential and radial organization of thalamocortical axons. J Neurosci 22, 4987-5000.

Weissenbacher, A., Kasess, C., Gerstl, F., Lanzenberger, R., Moser, E., Windischberger, C., 2009. Correlations and anticorrelations in resting-state functional connectivity MRI: a quantitative comparison of preprocessing strategies. NeuroImage 47, 1408-1416.

Windischberger, C., Lanzenberger, R., Holik, A., Spindelegger, C., Stein, P., Moser, U., Gerstl, F., Fink, M., Moser, E., Kasper, S., 2010. Area-specific modulation of neural activation comparing escitalopram and citalopram revealed by pharmaco-fMRI: a randomized cross-over study. NeuroImage 49, 1161-1170.

Wonderlick, J.S., Ziegler, D.A., Hosseini-Varnamkhasti, P., Locascio, J.J., Bakkour, A., van der Kouwe, A., Triantafyllou, C., Corkin, S., Dickerson, B.C., 2009. Reliability of MRI-derived cortical and subcortical morphometric measures: effects of pulse sequence, voxel geometry, and parallel imaging. NeuroImage 44, 1324-1333.

Zimmer, L., Riad, M., Rbah, L., Belkacem-Kahlouli, A., Le Bars, D., Renaud, B., Descarries, L., 2004. Toward brain imaging of serotonin 5-HT1A autoreceptor internalization. NeuroImage 22, 1421-1426.

79

TABLES

all subjects

males

females

P

N 17

11

6

Age (years) 26.5 ± 6.1

28 ± 7.1

23.8 ± 2.1

0.185

BMI (kg2/m) 22.3 ± 2.3

22 ± 2.7

23 ± 1.3

0.382

Cigarettes/day 1.8 ± 4

0.8 ± 2.2

3.6 ± 5.7

0.533+

Alcohol/week 5.7 ± 6.5

3.9 ± 4.9

9 ± 8.2

0.128

tGMD 919.3

918.6 ± 19.9

920.6 ± 14.8

0.746

Table 1: Demographic data of the study sample. Data are given as means ± SD. Alcohol units per

week = alcohol consumption (liter) × alcohol by volume ratio. BMI = body mass index. tGMD = total

gray matter density (placebo condition). P compares males and females with independent sample t-

test or Mann-Whitney U test (+) where normal distribution was not obtained by Levene's test.

80

Table 2: Regions exhibiting increases of gray matter density (GMD) after SSRI intake vs. placebo

intake. Voxel−wise whole brain repeated measurements ANOVA using GMD MRIs with treatment

modality (SSRIs or placebo) as factors controlling for whole brain GMD and sex. Stereotactical

coordinates (x, y, z) represent cluster peaks in standard Montreal Institute of Neurology (MNI) space.

FWE = family wise error. L = left, R = right.

region

peak

x y z

T Z P−FWE voxels

Fusiform gyrus (see Figure 2C, cororal plane)

28 –59 –19

15.3 Inf

< 0.001 7758

Posterior cingulate cortex / ventral precuneus (Fig. 1)

8 –45 38

16.6 Inf

< 0.001 3261

Insula L

–33 –14 5

10.8 6.9

< 0.001 748

Insula R

33 –20 11

11.8 7.2

< 0.001 739

Medial superior frontal cortex L

–10 48 51

9.4 6.4

< 0.001 635

Supramarginal cortex L

–52 –39 26

9.6 6.5

< 0.001 581

Supramarginal cortex R

38 –32 42

11.7 7.2

< 0.001 548

Medial superior frontal cortex R

21 45 54

9.2 6.4

< 0.001 409

Superior occipital cortex L

–21 –69 33

15.0 Inf

< 0.001 235

Medial temporal pole R

34 12 –33

8.3 6.0

< 0.001 223

Cerebellar crus R

50 –74 –37

10.8 6.9

< 0.001 218

Precentral cortex L

–62 1 38

10.4 6.8

< 0.001 191

Postcentral cortex L

–44 –20 38

9.0 6.3

< 0.001 176 Inferior frontal cortex, pars triangularis L

–39 34 12

8.1 5.9

< 0.001 174

Medial temporal cortex L

–45 –54 20

10.1 6.7

< 0.001 155

Precentral cortex R

33 –6 48

10 6.7

< 0.001 155

Posterior hippocampus L

–26 –33 –3

8.8 6.2

< 0.001 119

Cerebellar crus L

–16 –87 –37

8.8 6.2

< 0.001 116

Medial frontal cortex L

–27 –12 51

9.5 6.5

< 0.001 102

81

region

peak

x y z T Z P−FWE voxels

Precentral / postcentral L/R

37 –8 68

13.4 7.6

< 0.001 6259

Cerebellum R

36 –32 –49

11.4 7.1

< 0.001 1086

Cerebellum R

18 –60 –28

10.0 6.6

< 0.001 845

Posterior cingulate R

–4 –35 17

12.4 7.4

< 0.001 807

Cerebellum L

–9 –62 –25

10.3 6.7

< 0.001 806

Medial occipital/ temporal R

50 –83 22

8.7 6.2

< 0.001 582

Inferior frontal trigonum R

60 31 12

11.0 7.0

< 0.001 578

Cerebellum R

–21 –27 –39

10.9 7.0

< 0.001 566

Superior temporal L

–36 –41 6

10.1 6.7

< 0.001 542

Anterior cingulate L

–10 33 6

10.6 6.9

< 0.001 373

Medial cingulate R

15 –8 45

10.1 6.7

< 0.001 334

Cuneus L

–8 –78 18

9.4 6.4

< 0.001 271

Cerebellum L

–27 –99 –21

8.4 6.0

< 0.001 257

Superior temporal R

34 –38 12

9.2 6.4

< 0.001 228

Precentral R

48 –2 27

8.6 6.1

< 0.001 218

Medial temporal R

48 –33 –7

8.5 6.1

< 0.001 146

Rectus R

2 40 –39

14.0 7.8

< 0.001 144

Precuneus L

–10 –60 36

9.8 6.6

< 0.001 136

Medial cingulate R

–12 –3 45

8.2 5.9

< 0.001 122

Calcarine R

12 –78 18

8.4 6.0

< 0.001 119

Table 3: Regions exhibiting decreases of gray matter density (GMD) after SSRI intake vs. placebo

intake. Voxel−wise whole brain repeated measurements ANOVA using GMD MRIs with treatment

modality (SSRIs or placebo) as factors and whole brain GMD and sex as controlling factors.

Stereotactical coordinates (x, y, z) represent cluster peaks in standard Montreal Institute of

Neurology (MNI) space. FWE = family wise error. L = left, R = right.

82

1.13 Third publication: Exploring the impact of BDNF

Val66Met genotype on serotonin transporter and

serotonin-1A receptor binding

Christoph Kraus1, MD, Pia Baldinger1, MD, Christina Rami-Mark2, MSc, Gregor Gryglewski1,

Georg S. Kranz1, PhD, Daniela Haeusler2, PhD, Andreas Hahn1, PhD,

Wolfgang Wadsak2, Assoc. Prof. PD PhD, Markus Mitterhauser2, Assoc. Prof. PD,

Dan Rujescu3, Prof. MD, Siegfried Kasper1, Prof. MD, Rupert Lanzenberger1*, Assoc. Prof. PD MD

1 Department of Psychiatry and Psychotherapy,

2 Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine,

Medical University of Vienna, Austria

3 Department of Psychiatry, Medical University of Halle, Germany

published in:

PLOS-One, 2014 Sep 4;9(9)

[2013: 3.53]

* Correspondence to: Rupert Lanzenberger, Assoc. Prof. PD, MD

Functional, Molecular and Translational Neuroimaging Lab

Department of Psychiatry and Psychotherapy

Medical University of Vienna

Waehringer Guertel 18-20, 1090 Vienna, Austria

Phone (Fax): +43 1 40400 3825 (3099)

[email protected]

http://www.meduniwien.ac.at/neuroimaging

83

ABSTRACT

Background: The brain-derived neurotrophic factor (BDNF) Val66Met polymorphism

(rs6265) may impact on the in-vivo binding of important serotonergic structures such as the

serotonin transporter (5-HTT) and the serotonin-1A (5-HT1A) receptor. Previous positron

emission tomography (PET) studies on the association between Val66Met and 5-HTT and 5-

HT1A binding potential (BPND) have demonstrated equivocal results.

Methods: We conducted an imaging genetics study investigating the effect of Val66Met

genotype on 5-HTT or 5-HT1A BPND in 92 subjects. Forty-one subjects (25 healthy subjects

and 16 depressive patients) underwent genotyping for Val66Met and PET imaging with the

5-HTT specific radioligand [11C]DASB. Additionally, in 51 healthy subjects Val66Met

genotypes and 5-HT1A binding with the radioligand [carbonyl-11C]WAY-100635 were

ascertained. Voxel-wise and region of interest-based analyses of variance were used to

examine the influence of Val66Met on 5-HTT and 5-HT1A BPND.

Results: No significant differences of 5-HTT nor 5-HT1A BPND between BDNF Val66Met

genotype groups (val/val vs. met-carrier) were detected. There was no interaction between

depression and Val66Met genotype status.

Conclusion: In line with previous data, our work confirms an absent effect of BDNF

Val66Met on two major serotonergic structures. These results could suggest that altered

protein expression associated with genetic variants, might be compensated in vivo by

several levels of unknown feed-back mechanisms. In conclusion, Val66Met genotype status

is not associated with changes of in-vivo binding of 5-HTT and 5-HT1A receptors in human

subjects.

84

INTRODUCTION

The brain-derived neurotrophic factor (BDNF) is the most prominent member in the

neurotrophin family and involved in development and activity-dependent regulation of

neuronal structures [1]. Cumulating evidence demonstrated a functional interplay between

BDNF and the neurotransmitter serotonin (5-HT), constituting common intracellular signaling

pathways and transcription factors, BDNF control over the development and function of

serotonergic neurons as well as serotonergic regulation of BDNF gene expression and

signaling [2].

Briefly, BDNF is linked with at least three major intracellular signaling cascades: the

phosphoinositide-3 kinase pathway enabling cell survival, the phospholipase-gamma

pathway effecting synaptic plasticity and the mitogen-activated protein kinase pathway

associated with neuronal differentiation and neurite outgrowth [3]. Beside the p75

neurotrophin receptor, which is activated by proBDNF and all other neurotrophins, BDNF

releases it’s effects by binding to tropomyosin-kinase related receptor B (TrkB) [4-6].

Thereby, BDNF is a major factor in the proper development and plastic regulation of the

central nervous system and highly active in limbic structures such as the hippocampus and

the amygdala, where long-term potentiation, learning and memory are facilitated [7].

However, it should be stated here that most of the evidence of BDNF in this context is based

on rodent data.

The BDNF gene is located at chromosome 11p13-14, including many splice sites and

promoters. All BDNF mRNAs are initially translated into proBDNF and are then cleaved into

mature BDNF [8]. The most investigated polymorphism of the BDNF gene exists in the

codon 66 of proBDNF (Val66Met, rs6265) and consists of a valine to methionine substitution,

which is associated with reduced intracellular proBDNF trafficking, synaptic secretion of

BDNF, and thus a lower extracellular BDNF concentration in met-allele carriers [9]. Thought

85

to trigger deficits in neuronal development and plasticity, the Val66Met polymorphism is of

major interest in neuropsychiatric research [2, 7].

Interestingly, in humans the molecular connections between 5-HT and BDNF, and how

alterations in one system affect the other are hardly known. Due to the lack of current

methods to measure BDNF, TrkB or p75 in the living human brain, in vivo research in

humans mainly focuses on the investigation of alterations of serotonergic structures thought

to be mediated via changes in BDNF. In imaging genetics studies, serotonergic markers are

labeled by radioligands and their binding is measured using PET. As yet, there exist three

studies investigating alterations of BDNF, as represented by the Val66Met polymorphism,

and it’s association with binding of 5-HT1A, 5-HT2A receptors as well as the 5-HTT in the

human brain [10-12]. Two previous studies failed to detect links between Val66Met and

binding of 5-HT1A and 5-HT2A receptors. On the other side, a recently published study

reports lower 5-HT1A binding in healthy subjects carrying the met-allele compared to val-

homozygotes, a difference which was not observed in depressed subjects [12]. As far as 5-

HTT is concerned, in one study, applying the serotonin transporter (5-HTT) specific

radioligand [11C]-MADAM (N=25) with PET and [123I]-ß-CIT (N=18) with single photon

emission tomography (SPECT) in two independent samples, the authors found increased 5-

HTT binding in val-homozygote male subjects and compared to met-allele carriers [10]. On

the other hand applying the radioligand [11C]DASB (N=49), the second study failed to detect

any effect of Val66Met genotype status on 5-HTT binding [11].

To resolve contradictory results we conducted an imaging genetics study investigating the

association between 5-HTT binding using PET with the radioligand [11C]DASB and the

Val66Met genotype status in healthy subjects as well as in depressive patients. We also

measured 5-HT1A receptor binding in healthy subjects genotyped for Val66Met, in order to

resolve two equivocal findings. We hypothesized, that Val66Met impacts on 5-HTT binding in

patients with major depression and healthy subjects. Furthermore, we hypothesized that

86

significant differences are detected between BDNF genotype status and 5-HT1A binding in

healthy subjects.

METHODS

Subjects

In a neuroimaging genetics study with a cross-sectional design in total 92 subjects, aged 18-

65 years were included. The study was divided into two groups, in the first one 51 healthy

adult volunteers (37 female) were included and measured with [carbonyl-11C]WAY-10063. In

the second group 25 healthy subjects (HS) and 16 currently depressed patients with an

Hamilton Depression Rating Scale ≥ 16 (HAMD: 19.7±3.5, mean±SD) were included (for

further details see table 1) and measured with [11C]DASB. None of the subjects received

both radioligands. The study population originates from a pooled sample, which is part of

previously published studies [13-16]. Genotyping data of BNDF were previously not

published. All subjects underwent a psychiatric screening by the help of the complete

Structured Clinical Interview for DSM-IV type disorders (SCID I+II), physical and neurological

examination, clinical history, ECG, routine laboratory analysis, urinary drug and pregnancy

screening. All subjects were at least three months free of any psychotropic medication.

Every study subject was enrolled in study participation after detailed oral information about

all study procedures and subsequent signing of a written informed consent form. The study

and all study related procedures were approved by the Ethics Committee of the Medical

University of Vienna.

BDNF Genotyping

All procedures were performed as previously described [13]. Briefly, DNA was isolated from

peripheral blood mononuclear cells by the QIAamp DNA Mini-Kit (QIAGEN®, Hilden,

87

Germany). Genotyping of BDNF rs6265 single nucleotide polymorphism (SNP) was

conducted with the MassARRAY platform (SEQUENOM®, San Diego, CA) as described

elsewhere [17]. PCR-primers were generated with the Assay Designer 4.0 software

(SEQUENOM®). Multiplex PCR reactions were performed with 12.5ng of genomic DNA,

500μM dNTPs (ABgene®, Hamburg, Germany), 100nM PCR primers, 1.625mM MgCl2 and

0.5U HotStar Taq polymerase (QIAGEN®). Shrimp alkaline phosphatase (SAP) treatment,

an iPLEX reaction cocktail with extension primers (7-14μM), a iPLEX termination mix and an

iPLEX enzyme (SEQUENOM®) were added to the PCR-products. The resulting extension

products were desalted using SpectroCLEAN resin (SEQUENOM®), then spotted on

SpectroCHIPs GenII (SEQUENOM®) and analyzed with the MassARRAY MALDI-TOF mass

spectrometer. Typer 3.4 Software was used to identify allele specific extension products and

resulting genotypes (SEQUENOM®). For genotyping quality assurance CEU HapMap Trios

(Coriell Institute for Medical research, Camden, NJ) were included and compared with the

HapMap-CEU population (www.hapmap.org). For all analyses val/val homozygotes (=GG-

carriers) were compared against met-carriers (AG- and AA-carriers).

Radiochemistry of [11C]DASB and [carbonyl-11C]WAY-100635 and PET Procedures

Radioligand synthesis and all PET measurements were conducted at the Department of

Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine at the Medical

University of Vienna. PET measurements were performed with a GE Advance full ring PET

scanner (General Electric Medical Systems, Waukesha, WI, USA). Subjects were placed

with their head parallel to the orbitomeatal line guided by a laser beam system to ensure full

coverage of the neocortex and the cerebellum in the field of view (FOV). A polyurethane

cushion and head straps were used to minimize head movement and to guarantee a soft

head rest during the whole scanning period.

88

For a complete description of [11C]DASB radioligand synthesis see [18]. Mean injected dose

was 358.97±70.47 MBq, specific activity at time of injection was 49.00±38.10 MBq/nmol and

radiochemical purity was above 95%. After a 5 min transmission scan with retractable 68Ge

rod sources the 3D dynamic emission measurement was initiated simultaneously with the

intravenous bolus injection of the radioligand [11C]DASB. The total acquisition time (35

slices) was 90 min and reconstructed images comprised a spatial resolution of 4.36 mm full-

width at half-maximum (FWHM).

For a complete description of [carbonyl-11C]WAY-100635 please see [19, 20]. Mean injected

dose was 312.04±105.84 MBq, specific activity at time of injection was

285.47±251.22GBq/µmol and radiochemical purity was above 95%. Again, a 5 min

transmission scan (68Ge) was followed by 90 min dynamic scanning per subject at a spatial

resolution of 4.36 mm FWHM.

Data preprocessing and calculation of binding potential

PET preprocessing was done in SPM8 (Wellcome Trust Centre for Neuroimaging, London,

UK, http://www.fil.ion.ucl.ac.uk/spm/) using standard algorithms and parameters unless

stated differently. After realignment to the mean image (quality = 1) scans of the entire time

series were summed up and spatially normalized (affine regularization = average sized

template) to a tracer-specific template within standard MNI-space (Montreal Neurological

Institute). Thereafter, the resulting transformation matrix was applied to each time frame.

We assessed in vivo target structure density as indexed by 5-HT1A receptor and 5-HTT

binding potentials (BPND), which represent the ratio at equilibrium of specifically bound

radioligand to that of nondisplaceable radioligand in tissue [21]. All binding potentials were

computed using the voxel-wise modeling tool in the PMOD 3.3 software package (PMOD

Technologies, Ltd., Zurich, Switzerland) and applying the two-parameter linearized reference

89

tissue model (MRTM2) [22], which provides advantages in signal-to-noise-ratio, especially

for whole-brain voxel-wise analysis.

We modeled 5-HT1A BPND as previously described by our group using the insula as receptor-

rich region and the cerebellum as receptor-poor region [23]. The cerebellar gray matter

excluding cerebellar vermis and venous sinus served as reference region. Serotonin

transporter BPND were modeled using the MRTM2 as previously described [16]. In short, k2’

was estimated from the striatum as 5-HTT-rich region and the cerebellar gray matter (excl.

vermis and venous sinus) as 5-HTT-poor region. The cerebellar gray matter was chosen

because it represents an optimal reference region for the quantification of the serotonin

transporter with [11C]DASB [24, 25]. Regions of interest (ROI) for both radioligands were

taken from an automated anatomical labeling-based (AAL) atlas [26] after normalization of

BPND maps to standard MNI-space. Values were averaged across both hemispheres. Due to

inherent smoothness of PET data of the scanner and temporary smoothing during

normalization we did not smooth during statistical processing.

Statistical Analysis

For normally distributed demographic variables and clinical measures student’s t-tests, for

nominal variables chi-squared tests were performed. Significance was determined as p<0.05

and all tests were two-sided.

Differences of 5-HT1A and 5-HTT BPND between BDNF Val66Met genotype groups were

calculated using a voxel-wise and a ROI-based approach. For the voxel-wise analysis both

in the 5-HTT and the 5-HT1A – groups an ANOVA was performed as implemented in SPM8.

Grouped genotype status (val/val, vs. met-carrier = GG vs. A-carrier) served as factor and

radioligand specific activity, sex and age served as covariates. In the 5-HTT-collective

diagnosis was added as additional factor in a second step analysis. F-tests and group-wise

post-hoc t-tests between genotype groups were calculated and contrasted in SPM8.

90

Additionally, in the 5-HTT-group an interaction between diagnosis and genotype status was

contrasted by weighting contrast vectors in SPM according to group size. An absolute image

threshold was set at 0.1 BPND to remove voxels with low signal-to-noise ratio and a cluster

threshold was set at 50 voxels. A statistical level of p<0.05 corrected for multiple

comparisons by the family-wise error rate (FWE) at voxel-level was considered significant,

for subsequent explorative analysis an uncorrected threshold of p<0.001 was accepted.

In the ROI-based analyses differences between genotypes groups (val/val vs. met carrier)

were calculated with a linear mixed model in SPSS 19 (IBM Corp. Released 2010. IBM

SPSS Statistics for Windows, Version 19.0. Armonk, NY: IBM Corp.). Thereby, subject

served as the random effect and BDNF genotype status, region, sex and age served as

fixed effects. Ten representative regions were chosen due to their a priori known high

distribution of 5-HT1A receptors and 5-HTT and implications in psychiatric disorders (see

tables 1,2 and figures 1,2). Diagnosis was taken as additional factor in the 5-HTT-study

collective. Significance was determined as p<0.05. Post-hoc t-tests were conducted two-

sided in 10 AAL ROIs (see tables 1,2 and figures 1,2).

RESULTS

Out of the 51 HS in the 5-HT1A-group 30 carried GG, 18 carried AG and 3 AA. The 5-HTT-

group had 25 HS with 19 carrying GG, 5 carried AG and 1 AA, whereas in the MDD group

with 16 depressed patients 13 carried GG, 3 carried AG and 0 the AA allele (table 1). Allele

frequencies of the BDNF gene in all study groups were distributed in accordance with the

Hardy-Weinberg equilibrium [5-HT1A-group: 2=0.02, p=0.891, 5-HTT-group HS: 2=0.72

p=0.4, MDD patients 2=0.17 p=0.68). The AA and AG+GG study groups did not differ in

demographical, clinical measures or radiopharmaceutical measures (table 1). The allelic

distribution was not associated with diagnosis in the 5-HTT-group (2=0.157, p=0.692).

91

In the voxel-wise analysis there was no significant association of BDNF genotype (GG vs. A-

carrier) status with 5-HT1A BPND (F-test: all p>0.05 FWE corr. and all p>0.001 uncorr.).

Furthermore, there was no significant association of BDNF genotype (GG vs. A-carrier) with

5-HTT BPND (F-test: all p>0.05 FWE corr. and all p>0.001 uncorr.). There was no interaction

between BDNF genotype status, diagnosis or sex and 5-HTT BPND (t-test: all p>0.05 FWE

corr. and all p>0.001 uncorr.).

The mixed model analyses of ROIs in the 5-HT1A-group, controlling for potential effects of

sex, age and specific radioligand activity, yielded no significant difference of 5-HT1A BPND in

selected ROIs between GG homozygotes and A-allele carriers (F=0.342, df=1,45, p=0.562).

In the 5-HTT-group, the mixed model revealed no significant difference between 5-HTT BPND

in the selected ROIs between GG homozygotes and A-allele carriers (F=0,41, df=1,33,

p=0.526). There was no interaction between diagnosis and allele in the statistical model

(p=0.989). Post-hoc t-tests and average BPND values for both study groups are shown in

Figure 1: Bar chart plotting serotonin-1A binding potential (5-HT1A BPND) according to BNDF

Val66Met genotype status. Values at the y-axis represent 5-HT1A BPND separated for val/val and

met-carrier, respectively, x-axis shows regions of interest. Regions and values correspond to table

2. ACC: anterior cingulate cortex, AMY: amygdala, MCC: medial cingulate cortex, HIPP:

hippocampus, INS: insula, paraHIPP: parahippocampus, PCC: posterior cingulate cortex,

TempPole: temporal pole, DRN: dorsal raphe nucleus.

92

table 2 and table 3, BPND-values of allele groups are displayed in figure 1 and figure 2. Here,

in the 5-HTT-group, a significant difference between GG and A-carriers was observed in HS

in the midbrain (p=0.040, uncorr., table 3) as well as between GG in HS and GG in MDD

patients (p=0.034, uncorr.), with BPND increases in GG-carriers, respectively. All other post-

hoc tests (5-HT1A: GG vs. A-carrier; 5-HTT HS: GG vs. A-carrier, MDD GG vs. A-carrier, HS

vs. MDD GG, HS males GG vs. HS males A-carrier) did not yield significant results (all

p>0.05 uncorr.).

DISCUSSION

In a voxel-wise analysis as well as in a ROI-based approach, we did not observe significant

differences of 5-HT1A-receptor BPND nor of 5-HTT BPND according to BDNF genotype status.

Figure 2: Bar chart plotting serotonin transporter binding potential (5-HTT BPND) according to

BNDF Val66Met genotype status. Values at the y-axis represent 5-HTT BPND in pooled healthy

subjects and depressive patients. Binding potential is separated for val/val and met-carriers,

respectively, x-axis shows regions of interest. Because healthy subjects and depressive patients

were pooled here, regions do, but values do not correspond to table 3. ACC: anterior cingulate

cortex, AMY: amygdala, MCC: medial cingulate cortex, HIPP: hippocampus, CAUD: caudatum,

PUT: putamen, THAL: thalamus, STRIA: striatum, MID: Midbrain, NACC: nucleus accumbens.

93

There was no interaction between MDD diagnosis or sex and 5-HTT BPND. In the midbrain,

weak increases of 5-HTT-BPND in healthy subjects between val-homozygotes and met-

carriers were found. Furthermore, weak increases of 5-HTT BPND were observed in the

midbrain in val-homozygote healthy subjects compared to val-homozygote MDD patients.

There was no association between allelic distribution and major depression. To sum up, all

voxel-wise and ROI-based testing yielded negative results and none of the post-hoc tests

survived correction.

Our results are in concordance with a previous PET study applying [11C]DASB in 49 healthy

subjects, where the authors neither detected differences in 5-HTT binding in relation to

BDNF genotype nor a correlation between blood BDNF levels and central 5-HTT binding

[11]. Additionally, no effect on 5-HT2A binding was shown in this work. Here, the authors

calculated the radiotracer BPND similar to our study by applying a fully automated reference

region model (MRTM2) [22] and an automated ROI-delineation. The only other currently

published human PET-study investigating the impact of BDNF polymorphisms on 5-HTT

binding reports differences in men and shows no effect of genotype status on 5-HT1A binding

[10]. Men homozygous for the val-allele exhibited significantly higher 5-HTT binding in

regions such as the hippocampus, insula or dorsal raphe compared to met-carrier, while this

effect was absent in women. Furthermore, reductions of 5-HTT binding in met-carrier (n=3)

compared to val-homozygotes (n=6) in an independent [123I]-ß-CIT-study with male suicide

attempters were demonstrated, but this reduction was absent when pooled with healthy

controls. The authors also used a reference region model with [11C]-MADAM, a tracer

exhibiting a comparable 5-HTT affinity to [11C]DASB [27], the ROIs were manually delineated

on individual magnetic resonance images (MRI). Notably, our group previously reported

strong correlations of BPND values between automatically and manually delineated ROIs

[23]. The radioligand and the method of ROI generation are on these grounds an unlikely

source of variance leading to alternative results. Importantly, in search of arguments for this

difference, one must mention that the number of male met-carriers in that collective was low

94

(n=4), which makes the analysis vulnerable to outliers and hence may increase type-I errors.

Likewise, our study exhibits a subgroup with a low subject number and indeed we saw an

outlier in the MDD met-carrier group (n=3) when we plotted the individual BPND values (data

not shown). Hence, our results in depressed patients have to be interpreted with caution. But

the fact that both the study by Klein et al., which exhibits a large sample size of healthy

volunteers, as well as our study did not reproduce higher 5-HTT binding in val-homozygote

healthy subjects, rather speaks for an absent effect of BDNF Val66Met on 5-HTT binding.

Apart from this, our study agrees with the data by Henningsson et al., on an absent effect of

Val66Met on 5-HT1A receptor binding in healthy subjects [10]. Both studies apply the same

radioligand, i.e. [carbonyl-11C]WAY-100635, exhibit an almost identical number of subjects

(n=53 in Henningsson et al.), and modeled 5-HT1A binding by a reference region model

(BPND). These results are in contradiction to a recent finding reporting 5-HT1A reductions in

healthy met-allele carriers [12], which is not present in MDD patients. In this study 50 healthy

subjects and 50 MDD patients were measured with the radioligand [carbonyl-11C]WAY-

100635, yet 5-HT1A binding was calculated by an arterial input function (BPF). Most

interestingly, when the authors repeated their analysis with BPND values, the reduction of 5-

HT1A binding in healthy met-carriers was not detectable, suggesting that this finding was

associated with the method of radioligand modeling. Following the discussion of the authors,

one cannot rule out that Val66Met causes differences of radioligand binding in the blood

leading to a bias in the arterial input function. Although, our results are in agreement with all

previous studies on 5-HT1A binding using reference tissue models [10, 12], validation by a

different tracer not susceptible to modeling methodology is further needed. Taken together,

while there are currently contradicting findings on the in vivo effect of BDNF Val66Met

genotypes on 5-HTT binding [10, 11], this study adds data emphasizing the absence of such

an effect. Moreover, this work corroborates previous results by reference tissue models

demonstrating no association between BDNF Val66Met genotype status and 5-HT1A

95

receptor binding [10, 12] and is in contradiction with a study reporting binding values

modeled with arterial blood sampling [12].

Preclinical data report that BDNF promotes development and function of serotonergic

neurons by enhancing survival and differentiation [28], increasing local 5-HT [29] modifying

the firing pattern of serotonergic raphe neurons [28, 30] and altering the function of

serotonergic receptors such as the 5-HT1A and 5-HT2A receptors and the 5-HTT [2, 29, 31].

Vice-versa, raised extracellular 5-HT levels occurring upon administration of SSRIs are

thought to increase local BDNF levels by enhanced phosphorylation of serotonergic receptor

coupled cAMP response element-binding (CREB) protein [32-34], a common target of BDNF

and G protein-coupled serotonergic receptors [2]. Confronted with this evidence, one is

puzzled upon the lack of strong evidence for an association between BDNF and

serotonergic structures in humans in vivo. However, preclinical studies are not consistent

and negative results regarding the expression of 5-HT receptors and transporter are

reported [31, 35]. Although the interaction between the BNDF and 5-HT provides a

promising bridge between structural and functional neuronal activity, and serves as

explanatory hypothesis for neuronal plasticity deficits in neuropsychiatric disorders, exact

mechanisms underlying the regulation of the cross connection between BDNF and 5-HT in

humans remain unresolved [36]. Our data in concert with above referred work speak for a

similar expression of 5-HTT and 5-HT1A receptors upon life-time BDNF reduction, but

unfortunately do not illuminate the mechanisms leading to this observation. Theoretically,

counter-regulatory or compensatory effects may have altered 5-HTT and 5-HT1A expression.

Furthermore, it is possible that not absolute numbers but functional activity of serotonergic

structures is altered by BDNF.

The evidence on connections between depression and BNDF genotype status is

inconsistent as well. Meta-analytical research suggested an association of Val66Met with

major depressive disorder antidepressant treatment response or hippocampal volume and a

role of gender and ethnicity [37-39]. However, recent meta-analyses refuted these

96

associations and detected power deficits in many trials [40-42]. Low serum levels of BDNF

were suggested as potential peripheral marker of depression and increase of serum BDNF

as response to the appropriate first-line treatment with selective 5-HT reuptake inhibitors

(SSRIs). Likewise, this association is weaker than initially thought and there is no

relationship between symptom severity and BDNF serum concentration [43]. Our results

suggest no association between allelic distribution and diagnosis. Our small number of MDD

subjects remain a limiting factor in that regard.

LIMITATIONS

Unfortunately a common problem of human PET studies is weak power resulting from low

subject numbers, owed to the large effort of conducting PET-imaging. This is even more

intrinsic to genetic PET studies reporting results based on genotype subgroups [44] and in

SNP neuroimaging studies where pooling of rare genotype groups is common practice. The

low subject number in the MDD met-carrier group could therefore be a limitation of our study.

One elegant way to circumvent this issue in future studies would be pooling data between

PET centers, which is already common in MRI studies. Second, mean age of genotype

groups is heterogeneous, yet controlled for in all statistical analyses. Finally, we did not

model PET data with an arterial input function [45], because arterial blood data were not

collected. This would have been useful to confirm reported differences according to the

methodology for calculating 5-HT1A binding with [carbonyl-11C]WAY-100635, an issue we are

trying to resolve in future studies [46].

CONCLUSION

Although others have investigated the effects of the BDNF gene on 5-HTT and 5-HT1A

binding with PET, this study adds data to the ongoing discussion about the cross connection

97

between 5-HT and BDNF. While previous work in humans demonstrated contradicting

results, due to this work the conclusion of an absent influence of Val66Met on 5-HTT and 5-

HT1A has gained substantial support.

ACKNOWLEDGEMENTS

The authors are grateful to U. Moser, E. Akimova, P. Stein, M. Fink, C. Spindelegger, A.

Höflich, I. Hofer-Irmler, S. Zgud, S. Pichler, A. Kautzky and D. Winkler for medical and

administrative support, and M. Savli for technical support. We thank the PET team,

especially G. Karanikas, T. Traub-Weidinger, L.-K. Mien, J. Ungersboeck, K. Kletter, L. Nics,

and C. Philippe for technical support. Further, we thank the genetics team of D. Rujescu,

especially M. Friedl, A. Hartmann, I. Giegling.

The study is part of C. Kraus’ thesis “Serotonin and Neuroplasticity” supervised by R.

Lanzenberger in the Clinical Neurosciences PhD program at the Medical University of

Vienna, Austria. Parts of this study have been presented by P. Baldinger at the 19th at the

11th World Congress of Biological Psychiatry (WFSBP), June 23rd – 27th, 2013, Kyoto, Japan.

REFERENCES

1. Johnson JE, Barde YA, Schwab M and Thoenen H (1986) Brain-derived neurotrophic factor supports the survival of cultured rat retinal ganglion cells. J Neurosci 6: 3031-3038.

2. Martinowich K and Lu B (2008) Interaction between BDNF and serotonin: role in mood disorders. Neuropsychopharmacology 33: 73-83.

3. Chao MV (2003) Neurotrophins and their receptors: a convergence point for many signalling pathways. Nat Rev Neurosci 4: 299-309.

4. Gentry JJ, Barker PA and Carter BD (2004) The p75 neurotrophin receptor: multiple interactors and numerous functions. Prog Brain Res 146: 25-39.

5. Gan WB (1999) TrkB works at postsynaptic sites. Neuron 24: 491-492.

6. Black IB (1999) Trophic regulation of synaptic plasticity. J Neurobiol 41: 108-118.

98

7. Castrén E (2005) Is mood chemistry? Nature reviews Neuroscience 6: 241-246.

8. Liu QR, Walther D, Drgon T, Polesskaya O, Lesnick TG, et al. (2005) Human brain derived neurotrophic factor (BDNF) genes, splicing patterns, and assessments of associations with substance abuse and Parkinson's Disease. Am J Med Genet B Neuropsychiatr Genet 134B: 93-103.

9. Chen ZY, Patel PD, Sant G, Meng CX, Teng KK, et al. (2004) Variant brain-derived neurotrophic factor (BDNF) (Met66) alters the intracellular trafficking and activity-dependent secretion of wild-type BDNF in neurosecretory cells and cortical neurons. J Neurosci 24: 4401-4411.

10. Henningsson S, Borg J, Lundberg J, Bah J, Lindstrom M, et al. (2009) Genetic variation in brain-derived neurotrophic factor is associated with serotonin transporter but not serotonin-1A receptor availability in men. Biol Psychiatry 66: 477-485.

11. Klein AB, Trajkovska V, Erritzoe D, Haugbol S, Madsen J, et al. (2010) Cerebral 5-HT2A receptor and serotonin transporter binding in humans are not affected by the val66met BDNF polymorphism status or blood BDNF levels. J Cereb Blood Flow Metab 30: e1-7.

12. Lan MJ, Ogden RT, Huang YY, Oquendo MA, Sullivan GM, et al. (2014) Genetic variation in brain-derived neurotrophic factor val66met allele is associated with altered serotonin-1A receptor binding in human brain. NeuroImage.

13. Baldinger P, Hahn A, Mitterhauser M, Kranz GS, Friedl M, et al. (2013) Impact of COMT genotype on serotonin-1A receptor binding investigated with PET. Brain Struct Funct.

14. Lanzenberger R, Kranz GS, Haeusler D, Akimova E, Savli M, et al. (2012) Prediction of SSRI treatment response in major depression based on serotonin transporter interplay between median raphe nucleus and projection areas. NeuroImage.

15. Lanzenberger R, Wadsak W, Spindelegger C, Mitterhauser M, Akimova E, et al. (2010) Cortisol plasma levels in social anxiety disorder patients correlate with serotonin-1A receptor binding in limbic brain regions. Int J Neuropsychopharm: 1-15.

16. Hahn A, Haeusler D, Kraus C, Hoflich AS, Kranz GS, et al. (2014) Attenuated serotonin transporter association between dorsal raphe and ventral striatum in major depression. Hum Brain Mapp.

17. Oeth P, Beaulieu M, Park C, Kosman D, del Mistro G, et al. (2006) iPLEXTM Assay: Increased Plexing Efficiency and Flexibility for MassARRAY® System Through Single Base Primer Extension with Mass-Modified Terminators. Application Note. Sequenom®.

18. Haeusler D, Mien LK, Nics L, Ungersboeck J, Philippe C, et al. (2009) Simple and rapid preparation of [11C]DASB with high quality and reliability for routine applications. Appl Radiat Isot 67: 1654-1660.

19. Wadsak W. MLK, Ettlinger D.E., Lanzenberger R., Haeusler D., Dudczak R., Kletter K., Mitterhauser M. (2007) Simple and fully automated preparation of [carbonyl-11C]WAY-100635. Radiochimica Acta 95: 1-6.

99

20. Rami-Mark C, Ungersboeck J, Haeusler D, Nics L, Philippe C, et al. (2013) Reliable set-up for in-loop (1)(1)C-carboxylations using Grignard reactions for the preparation of [carbonyl-(1)(1)C]WAY-100635 and [(1)(1)C]-(+)-PHNO. Appl Radiat Isot 82: 75-80.

21. Innis RB, Cunningham VJ, Delforge J, Fujita M, Gjedde A, et al. (2007) Consensus nomenclature for in vivo imaging of reversibly binding radioligands. J Cereb Blood Flow Metab 27: 1533-1539.

22. Ichise M, Liow J-S, Lu J-Q, Takano A, Model K, et al. (2003) Linearized Reference Tissue Parametric Imaging Methods: Application to [11C]DASB Positron Emission Tomography Studies of the Serotonin Transporter in Human Brain. Journal of Cerebral Blood Flow & Metabolism: 1096-1112.

23. Savli M, Bauer A, Mitterhauser M, Ding YS, Hahn A, et al. (2012) Normative database of the serotonergic system in healthy subjects using multi-tracer PET. NeuroImage 63: 447-459.

24. Parsey RV, Kent JM, Oquendo MA, Richards MC, Pratap M, et al. (2006) Acute occupancy of brain serotonin transporter by sertraline as measured by [11C]DASB and positron emission tomography. Biol Psychiatry 59: 821-828.

25. Meyer JH (2007) Imaging the serotonin transporter during major depressive disorder and antidepressant treatment. J Psychiatry Neurosci 32: 86-102.

26. Tzourio-Mazoyer N, Landeau B, Papathanassiou D, Crivello F, Etard O, et al. (2002) Automated Anatomical Labeling of Activations in SPM Using a Macroscopic Anatomical Parcellation of the MNI MRI Single-Subject Brain. NeuroImage 15: 273-289.

27. Laruelle M, Slifstein M and Huang Y (2003) Relationships between radiotracer properties and image quality in molecular imaging of the brain with positron emission tomography. Mol Imaging Biol 5: 363-375.

28. Djalali S, Holtje M, Grosse G, Rothe T, Stroh T, et al. (2005) Effects of brain-derived neurotrophic factor (BDNF) on glial cells and serotonergic neurones during development. J Neurochem 92: 616-627.

29. Guiard BP, David DJP, Deltheil T, Chenu F, Le Maître E, et al. (2008) Brain-derived neurotrophic factor-deficient mice exhibit a hippocampal hyperserotonergic phenotype. Int J Neuropsychopharmacol 11: 79-92.

30. Yu H, Wang DD, Wang Y, Liu T, Lee FS, et al. (2012) Variant brain-derived neurotrophic factor Val66Met polymorphism alters vulnerability to stress and response to antidepressants. J Neurosci 32: 4092-4101.

31. Daws LC, Munn JL, Valdez MF, Frosto-Burke T and Hensler JG (2007) Serotonin transporter function, but not expression, is dependent on brain-derived neurotrophic factor (BDNF): in vivo studies in BDNF-deficient mice. J Neurochem 101: 641-651.

32. Nibuya M, Morinobu S and Duman RS (1995) Regulation of BDNF and trkB mRNA in rat brain by chronic electroconvulsive seizure and antidepressant drug treatments. J Neurosci 15: 7539-7547.

100

33. Khundakar AA and Zetterstrom TS (2006) Biphasic change in BDNF gene expression following antidepressant drug treatment explained by differential transcript regulation. Brain Res 1106: 12-20.

34. Coppell AL, Pei Q and Zetterstrom TS (2003) Bi-phasic change in BDNF gene expression following antidepressant drug treatment. Neuropharmacology 44: 903-910.

35. Szapacs ME, Mathews TA, Tessarollo L, Ernest Lyons W, Mamounas LA, et al. (2004) Exploring the relationship between serotonin and brain-derived neurotrophic factor: analysis of BDNF protein and extraneuronal 5-HT in mice with reduced serotonin transporter or BDNF expression. J Neurosci Methods 140: 81-92.

36. Castren E (2013) Neuronal network plasticity and recovery from depression. JAMA Psychiatry 70: 983-989.

37. Zou YF, Ye DQ, Feng XL, Su H, Pan FM, et al. (2010) Meta-analysis of BDNF Val66Met polymorphism association with treatment response in patients with major depressive disorder. Eur Neuropsychopharmacol 20: 535-544.

38. Verhagen M, van der Meij A, van Deurzen PA, Janzing JG, Arias-Vasquez A, et al. (2010) Meta-analysis of the BDNF Val66Met polymorphism in major depressive disorder: effects of gender and ethnicity. Mol Psychiatry 15: 260-271.

39. Hajek T, Kopecek M and Hoschl C (2012) Reduced hippocampal volumes in healthy carriers of brain-derived neurotrophic factor Val66Met polymorphism: meta-analysis. World J Biol Psychiatry 13: 178-187.

40. Molendijk ML, van Tol MJ, Penninx BW, van der Wee NJ, Aleman A, et al. (2012) BDNF val66met affects hippocampal volume and emotion-related hippocampal memory activity. Transl Psychiatry 2: e74.

41. Gyekis JP, Yu W, Dong S, Wang H, Qian J, et al. (2013) No association of genetic variants in BDNF with major depression: a meta- and gene-based analysis. Am J Med Genet B Neuropsychiatr Genet 162B: 61-70.

42. Cole J, Weinberger DR, Mattay VS, Cheng X, Toga AW, et al. (2011) No effect of 5HTTLPR or BDNF Val66Met polymorphism on hippocampal morphology in major depression. Genes Brain Behav 10: 756-764.

43. Molendijk ML, Spinhoven P, Polak M, Bus BA, Penninx BW, et al. (2013) Serum BDNF concentrations as peripheral manifestations of depression: evidence from a systematic review and meta-analyses on 179 associations (N=9484). Mol Psychiatry.

44. Willeit M and Praschak-Rieder N (2010) Imaging the effects of genetic polymorphisms on radioligand binding in the living human brain: A review on genetic neuroreceptor imaging of monoaminergic systems in psychiatry. NeuroImage: 1-15.

45. Ichise M, Meyer JH and Yonekura Y (2001) An introduction to PET and SPECT neuroreceptor quantification models. J Nucl Med 42: 755-763.

46. Parsey RV, Ogden RT, Miller JM, Tin A, Hesselgrave N, et al. (2010) Higher Serotonin 1A Binding in a Second Major Depression Cohort: Modeling and Reference Region Considerations. Biol Psychiatry 68: 170-178.

101

Table 1. Demographic variables of the entire study sample.

val/val

met-carrier

p

healthy subjects

[carbonyl-11C]WAY-100635

N (=51) 30

21

Age (years) 43.8 ± 13.1

45.1 ± 12.36

0.737

Sex (f/m) 21/9

16/5

0.626*

weight 72.9 ± 17.1

67.1 ± 10.5

0.169

SA 296.9 ± 269.1

285.7 ± 197.3

0.702

[11

C]DASB

N (=25) 19

6

Age (years) 31.0 ± 8.8

33.0 ± 13.2

0.672

Sex (f/m) 8/11

1/5

0.258*

weight 76.7 ± 12.1

80.2 ± 10.8

0.537

SA 44.1 ± 47.7

25.6 ± 25.4

0.378

MDD patients

[11

C]DASB N (=16) 13

3

HAMD 19.4 ± 3.6

21± 3.5

0.495

Age (years) 41.1 ± 8.9

46.7 ± 7.5

0.34

Sex (f/m) 9/4

3/0

0.267*

weight 77.7 ± 21.3

61.3 ± 2.5

0.251

+

SA 63.9 ± 22.6

62.5 ± 16.7

0.925

Data are given as means ± standard deviations (SD). P-values compare pooled

BNDF Val66Met genotype groups with independent sample t-test, chi-square(*) or

Mann-Whitney U test (+) where appropriate.

102

Table 2. Post-hoc t-tests comparing serotonin-1A receptor (5-HT1A) binding

potential (BPND) according to BDNF Val66Met genotype status in 51 healthy

subjects.

healthy subjects [carbonyl-11C]WAY-100635

region val/val (n=30)

met-carrier (n=21)

p

Anterior cingulate cortex 3.54 ± 1.14

3.63 ± 0.86

0.758

Amygdala 3.98 ± 1.23

4.17 ± 1.03

0.559

Medial cingulate cortex 2.9 ± 0.97

2.98 ± 0.65

0.723

Hippocampus 3.64 ± 1.14

4.12 ± 0.94

0.118

Insula 4.46 ± 1.33

4.64 ± 0.91

0.596

Parahippocampus 5.41 ± 1.64

5.60 ± 1.14

0.596

Posterior cingulate cortex 2.2 ± 0.79

2.25 ± 0.58

0.822

Subgenual anterior cingulate 3.51 ± 0.96

3.85 ± 1.1

0.247

Temporal pole 4.65 ± 1.5

4.75 ± 0.93

0.786

Dorsal raphe nucleus 2.33 ± 0.87

2.29 ± 0.74

0.857

Regions of interest (ROIs) in standardized MNI space (Montreal Neurological Institute) were

calculated by automatic anatomical labeling in both hemispheres and averaged. Data are

given as 5-HT1A BPND means ± standard deviations (SD) for each ROI and compared by

post-hoc student’s t-tests, values correspond to bar charts in Fig.1.

103

Table 3. Post-hoc t-tests comparing serotonin transporter (5-HTT) binding potential

(BPND) according to BDNF Val66Met genotype status in 25 healthy subjects and 16

depressed patients.

region

healthy subjects

MDD patients

val/val (n=19)

met-carrier (n=6)

p

val/val (n=13)

met-carrier (n=3)

p

Anterior cingulate 0.42 ± 0.08 0.40 ± 0.06 0.759

0.38 ± 0.14 0.32 ± 0.15 0.517

Amygdala 1.24 ± 0.13 1.14 ± 0.17 0.167

1.06 ± 0.24 1.14 ± 0.46 0.685

Medial cingulate 0.40 ± 0.07 0.37 ± 0.08 0.431

0.37 ± 0.13 0.30 ± 0.12 0.395

Hippocampus 0.46 ± 0.08 0.41 ± 0.08 0.206

0.40 ± 0.10 0.44 ± 0.11 0.525

N. caudatus 1.84 ± 0.21 1.73 ± 0.22 0.305

1.72 ± 0.32 1.50 ± 0.35 0.309

Putamen 1.88 ± 0.18 1.85 ± 0.27 0.756

1.75 ± 0.28 1.50 ± 0.30 0.248

Thalamus 2.07 ± 0.23 1.88 ± 0.11 0.071

1.88 ± 0.37 1.72 ± 0.45 0.527

Striatum 1.70 ± 0.16 1.66 ± 0.22 0.624

1.58 ± 0.25 1.37 ± 0.28 0.231

Midbrain 2.91 ± 0.33 2.58 ± 0.31 0.040

2.62 ± 0.41 3.20 ± 1.80 0.382*

N. accumbens 1.95 ± 0.3 1.82 ± 0.26 0.327

1.82 ± 0.30 1.67 ± 0.46 0.572

Regions of interest (ROIs) in standardized MNI space (Montreal Neurological Institute) were

calculated by automatic anatomical labeling in both hemispheres and averaged. Data are given as 5-

HTT BPND means ± standard deviations (SD). T-tests or U-test (*) compare differences between

val/val and met-carrier for each ROI.

104

IV. GENERAL DISCUSSION and RAISED QUESTIONS

With this work we investigated a series of issues on the association of neuroplasticity with

serotonin in the living human brain. To this goal, neuroimaging methods such as structural

and functional MRI and PET with the radioligands [11C]DASB and [carbonyl-11C]WAY-100635

were used for the to probe relationships between elevated 5-HT levels, gray matter,

important serotonergic proteins and the neurotrophine brain derived neurotrophic factor. The

here presented results underline that some of the known functions of 5-HT shaping the

neuronal architecture of the brain in embryonic and early post-natal periods are very likely, at

least partly, conserved throughout adulthood.

While neuroplastic effects of the 5-HT1A receptor function were previously discovered in

animals, we here translated this feature into in vivo human research by demonstrating strong

associations between the distribution of the 5-HT1A receptor and regional gray matter

volume. This result was mainly observed in brain regions with high 5HT1A receptor density,

yet restricted to certain regions and not present in others with equaly high 5-HT1A densitiy,

suggesting regional differences in neuroplastic functions. Furthermore, a correlation of 5-

HT1A autoreceptors in the dorsal raphe nuclei, one of the midbrain’s major serotonergic

nucleus, and cortical gray matter in the anterior cingulate cortex undermined the regulatory

function of the raphe nuclei. With the limitation of beeing correlational, these results in any

case justify further longitudinal investigations e.g. into how pharmacological manipulations of

5-HT1A receptor function would be able to effect changes of gray matter. Secondly, we found

that elevated 5-HT levels after selective serotonin reuptake inhibitor administration leads to

strong enhancements of cortical gray matter in the posterior cingulate cortex, which are in

turn associated with altered functional neuronal activity in this region. While this finding goes

along with previous work showing gray matter inceases after selective serotonin reuptake

inhibitor intake in psychiatric patients (Hoexter et al, 2012; Smith et al, 2012; Vermetten et al,

2003), gray matter increases in healthy subjects constitute a novel finding, as well as

adjacent functional alterations as consequence of enhanced gray matter. These results raise

105

many questions. Given that healthy subjects do not exhibit major neuropsychological

changes after 10 days of SSRI intake, one might ask what are the phenotypical correlates of

such strong signal increases. Consequently, this work demands closer investigations of the

biological correlate of VBM results. Several authors question the sensitivity regarding

neuronal changes in VBM studies (Bookstein, 2001; Franklin et al, 2013) and suspect

changes of perfusion and difusion of cerebral blood flow to underlie VBM findings. Indeed,

the huge gap between biological mechanisms and findings of MRI-based structural

neuroimaigng techniques are currently discussed controversially (Draganski & Kherif, 2013;

Erickson, 2013; Fields, 2013; Thomas & Baker, 2013). This work adds to the ongoing

discussion that neuroplastic serotonergic receptors might contribute to structural MRI signal

changes. This supposition provides new testable hypoptheses, that could combine

neurobiological data on neuroplasticity with neuroimaging-based in vivo information.

Finally, we found no evidence for an association between SERT or 5-HT1A availability upon

lifetime alterations of BDNF as produced by a common single nucleotid polymorphism

(Val66Met). This finding is controversial, because two previous studies demonstrated an

effect of this polymorphism on the expression of SERT and 5-HT1A receptors (Henningsson

et al, 2009). The dicrepancy might arise from methodological differences in quantification

using the radiligand [carbonyl-11C]WAY-100635, especially due to reference region data from

an aterial input function by other groups. Given the reported evidence in the introduction

section, BDNF and 5-HT posess many molecular crossconections, but previous studies

failed to report strong effects in one system upon deficites of the other. Reciprocal

compensatory mechanisms might be able to counter-regulate single weak points, and this

fits well to clinical studies generally reporting no clearcut evidence of “single-deficite”-

proteins in psychiatric conditions such as depression, in which BDNF and 5-HT are

considered relevant pathogenetic factors. In these disorders, pathophysiological concepts

comprise entaglements between biological vulnerability by genetical predisposition, multi-hit

neuropathological defictes and environmental adversities (Krishnan & Nestler, 2010;

106

Pittenger & Duman, 2008; Schmidt et al, 2011). Both 5-HT and BDNF would provide ample

neurobiological targets connecting these three concepts, yet until now only a very limited

number of studies encompass this phenomenologically heterogenous spectrum. Especially

studies applying imaging genetics and information on environmental adversities could better

unrevel the impact of these two neurophysiologic systems to pathomechanisms of

depression (Agren et al, 2012; Rabl et al, 2014; Witte et al, 2012). With this latter study we

have provided a groundwork and justification of such studies.

A limitation of this project constitutes the still insufficient resolution of neuroimaging

techniques and the lack of knowledge about exact neurobiological correlates of structural

MRI-based methods. Therefore, due to methodological constraints, it is currently not

possible to detect in vivo in humans how exactly 5-HT exerts it’s neuroplastic functions.

Nevertheless, a combination of MRI and PET in multimodal neuroimaging, as applied in the

first study, is elegantly capable of adding molecular information to the same voxel and thus

bringing neuroimaging closer to neurobiology. The next step, would be study human brain

function with combinations of structural, functional and molecular neuroimaging and in

translational animal models with additional post-mortem data, for examples see (Sagi et al,

2012; Vernon et al, 2011). Additional factors might have influenced our results, including low

power in the molecular imaging genetics study, where statistical power is a trade-off to high

efforts in conducting PET studies and subsequent splitting of study subjects into genotype

groups. Finally, we have to state in this section that binding potential results of [carbonyl-

11C]WAY-100635 vary according to the chosen reference region expecially under

consideration of the arterial input function, and that discrepent results between groups are

thought to be susceptible to heterogenous methodological aproaches, for review see

(Shrestha et al, 2012).

107

V. CONCLUSION and FUTURE PERSPECTIVES

This doctoral thesis was conducted to investigate neuroplastic functions of 5-HT in vivo in

healthy humans. By a combination of structural and functional MRI with molecular imaging of

the serotonergic system including imaging genetics, this work assess the links between 5-

HT and neuroplasticity with multiple methodological approaches. The central statements of

this thesis lies in demonstrating the relationship between structural, molecular, functional

and genetic properties of the human serotonergic system as measured with neuroimaging.

While this improved our knowledge on structural and functional properties of this

neurotransmitter system, future work on pathological alterations of serotonin’s neuroplastic

capabilites is needed. While previous knowledge revealed that 5-HT is implicated in the

pathogenesis of psychiatric diseases such as depression, and serotonergic receptors and

the 5-HT transporter are important targets in psychopharmacology, little is known about the

exact pathomechanisms of depression and the mechanisms of action of antidepressants.

Investigations of deficites in 5-HT mediated neuroplasticity in depression could hence

provide a unification of two previously competing major pathogenetic hypotheses, the

“neuroplasticity hypothesis” and the “monoamine hypothesis”.

Additionally, this work gives rise to still unresolved problems regarding the neurobiology

behind MRI-based neuroimaging methods. Bringing neuroimaging closer to neurobiology

could thereby reduce the gap between sophisticated neuroscientific “bench” aproaches in

animals such as optogenetics or stem cell research and low resolution human “bed side” in

vivo methods. Certainly, a better understanding of neurobiological alterations in vivo would

generate big leaps foreward in the understanding and treatment of psychiatric diseases such

as depression.

108

VI. REFERENCES

Agren T, Furmark T, Eriksson E, Fredrikson M (2012) Human fear reconsolidation and allelic differences in serotonergic and dopaminergic genes. Transl Psychiatry 2: e76

Akbari HM, Whitaker-Azmitia PM, Azmitia EC (1994) Prenatal cocaine decreases the trophic factor S-100 beta and induced microcephaly: reversal by postnatal 5-HT1A receptor agonist. Neurosci Lett 170: 141-144

Akimova E, Lanzenberger R, Kasper S (2009) The serotonin-1A receptor in anxiety disorders. Biol Psychiatry 66: 627-635

Asami T, Bouix S, Whitford TJ, Shenton ME, Salisbury DF, McCarley RW (2012) Longitudinal loss of gray matter volume in patients with first-episode schizophrenia: DARTEL automated analysis and ROI validation. Neuroimage 59: 986-996

Ashburner J (2007) A fast diffeomorphic image registration algorithm. Neuroimage 38: 95-113

Ashburner J (2009) Computational anatomy with the SPM software. Magn Reson Imaging 27: 1163-1174

Ashburner J, Friston KJ (2000) Voxel-based morphometry--the methods. NeuroImage 11: 805-821

Ashburner J, Friston KJ (2009) Computing average shaped tissue probability templates. Neuroimage 45: 333-341

Azmitia EC (1999) Serotonin neurons, neuroplasticity, and homeostasis of neural tissue. Neuropsychopharmacology 21: 33S-45S

Azmitia EC (2001) Modern views on an ancient chemical: serotonin effects on cell proliferation, maturation, and apoptosis. Brain research bulletin 56: 413-424

Azmitia EC, Nixon R (2008) Dystrophic serotonergic axons in neurodegenerative diseases. Brain Res 1217: 185-194

Barkas L, Redhead E, Taylor M, Shtaya A, Hamilton DA, Gray WP (2012) Fluoxetine restores spatial learning but not accelerated forgetting in mesial temporal lobe epilepsy. Brain 135: 2358-2374

Benninghoff J, van der Ven A, Schloesser RJ, Moessner R, Moller HJ, Rujescu D (2012) The complex role of the serotonin transporter in adult neurogenesis and neuroplasticity. A critical review. World J Biol Psychiatry 13: 240-247

109

Berg JM, Tymoczko JL, Stryer L (2002) Biochemistry, 5th ed edn. New York: W.H. Freeman.

Billard T, Le Bars D, Zimmer L (2014) PET radiotracers for molecular imaging of serotonin 5-HT1A receptors. Curr Med Chem 21: 70-81

Black IB (1999) Trophic regulation of synaptic plasticity. J Neurobiol 41: 108-118

Boersma MC, Dresselhaus EC, De Biase LM, Mihalas AB, Bergles DE, Meffert MK (2011) A requirement for nuclear factor-kappaB in developmental and plasticity-associated synaptogenesis. J Neurosci 31: 5414-5425

Bonnin A, Peng W, Hewlett W, Levitt P (2006) Expression mapping of 5-HT1 serotonin receptor subtypes during fetal and early postnatal mouse forebrain development. Neuroscience 141: 781-794

Bookstein FL (2001) "Voxel-based morphometry" should not be used with imperfectly registered images. Neuroimage 14: 1454-1462

Borrelli E, Nestler EJ, Allis CD, Sassone-Corsi P (2008) Decoding the epigenetic language of neuronal plasticity. Neuron 60: 961-974

Brans RG, Kahn RS, Schnack HG, van Baal GC, Posthuma D, van Haren NE, Lepage C, Lerch JP, Collins DL, Evans AC, Boomsma DI, Hulshoff Pol HE (2010) Brain plasticity and intellectual ability are influenced by shared genes. J Neurosci 30: 5519-5524

Castrén E (2005) Is mood chemistry? Nature reviews Neuroscience 6: 241-246

Chao MV (2003) Neurotrophins and their receptors: a convergence point for many signalling pathways. Nat Rev Neurosci 4: 299-309

Chen ZY, Patel PD, Sant G, Meng CX, Teng KK, Hempstead BL, Lee FS (2004) Variant brain-derived neurotrophic factor (BDNF) (Met66) alters the intracellular trafficking and activity-dependent secretion of wild-type BDNF in neurosecretory cells and cortical neurons. J Neurosci 24: 4401-4411

Chételat G, Desgranges B, Landeau B, Mézenge F, Poline JB, de la Sayette V, Viader F, Eustache F, Baron J-C (2008) Direct voxel-based comparison between grey matter hypometabolism and atrophy in Alzheimer's disease. Brain 131: 60-71

Chollet F, Tardy J, Albucher JF, Thalamas C, Berard E, Lamy C, Bejot Y, Deltour S, Jaillard A, Niclot P, Guillon B, Moulin T, Marque P, Pariente J, Arnaud C, Loubinoux I (2011) Fluoxetine for motor recovery after acute ischaemic stroke (FLAME): a randomised placebo-controlled trial. Lancet Neurol 10: 123-130

110

Chow DK, Groszer M, Pribadi M, Machniki M, Carmichael ST, Liu X, Trachtenberg JT (2009) Laminar and compartmental regulation of dendritic growth in mature cortex. Nat Neurosci 12: 116-118

Citri A, Malenka RC (2008) Synaptic plasticity: multiple forms, functions, and mechanisms. Neuropsychopharmacology 33: 18-41

Cowen DS (2007) Serotonin and neuronal growth factors - a convergence of signaling pathways. J Neurochem 101: 1161-1171

Cramer SC, Sur M, Dobkin BH, O'Brien C, Sanger TD, Trojanowski JQ, Rumsey JM, Hicks R, Cameron J, Chen D, Chen WG, Cohen LG, deCharms C, Duffy CJ, Eden GF, Fetz EE, Filart R, Freund M, Grant SJ, Haber S, Kalivas PW, Kolb B, Kramer AF, Lynch M, Mayberg HS, McQuillen PS, Nitkin R, Pascual-Leone A, Reuter-Lorenz P, Schiff N, Sharma A, Shekim L, Stryker M, Sullivan EV, Vinogradov S (2011) Harnessing neuroplasticity for clinical applications. Brain 134: 1591-1609

Daubert EA, Condron BG (2010) Serotonin: a regulator of neuronal morphology and circuitry. Trends Neurosci 33: 424-434

Daubert EA, Heffron DS, Mandell JW, Condron BG (2010) Serotonergic dystrophy induced by excess serotonin. Mol Cell Neurosci 44: 297-306

Dayan E, Cohen LG (2011) Neuroplasticity subserving motor skill learning. Neuron 72: 443-454

De Paola V, Holtmaat A, Knott G, Song S, Wilbrecht L, Caroni P, Svoboda K (2006) Cell type-specific structural plasticity of axonal branches and boutons in the adult neocortex. Neuron 49: 861-875

Delon-Martin C, Plailly J, Fonlupt P, Veyrac A, Royet JP (2013) Perfumers' expertise induces structural reorganization in olfactory brain regions. NeuroImage 68: 55-62

Dorsaint-Pierre R, Penhune VB, Watkins KE, Neelin P, Lerch JP, Bouffard M, Zatorre RJ (2006) Asymmetries of the planum temporale and Heschl's gyrus: relationship to language lateralization. Brain 129: 1164-1176

Draganski B, Gaser C, Busch V, Schuierer G, Bogdahn U, May A (2004) Neuroplasticity: changes in grey matter induced by training. Nature 427: 311-312

Draganski B, Gaser C, Kempermann G, Kuhn HG, Winkler J, Buchel C, May A (2006) Temporal and spatial dynamics of brain structure changes during extensive learning. J Neurosci 26: 6314-6317

111

Draganski B, Kherif F (2013) In vivo assessment of use-dependent brain plasticity-Beyond the "one trick pony" imaging strategy. NeuroImage 73: 255-259

Draganski B, May A (2008) Training-induced structural changes in the adult human brain. Behav Brain Res 192: 137-142

Duncan NW, Gravel P, Wiebking C, Reader AJ, Northoff G (2013) Grey matter density and GABAA binding potential show a positive linear relationship across cortical regions. Neuroscience 235: 226-231

Egan MF, Kojima M, Callicott JH, Goldberg TE, Kolachana BS, Bertolino A, Zaitsev E, Gold B, Goldman D, Dean M, Lu B, Weinberger DR (2003) The BDNF val66met polymorphism affects activity-dependent secretion of BDNF and human memory and hippocampal function. Cell 112: 257-269

Erickson KI (2013) Evidence for structural plasticity in humans: Comment on Thomas and Baker (2012). NeuroImage 73: 237-238

Eshaghi A, Bodini B, Ridgway GR, Garcia-Lorenzo D, Tozer DJ, Sahraian MA, Thompson AJ, Ciccarelli O (2014) Temporal and spatial evolution of grey matter atrophy in primary progressive multiple sclerosis. Neuroimage 86: 257-264

Fields RD (2013) Changes in brain structure during learning: Fact or artifact? Reply to Thomas and Baker. NeuroImage 73: 260-264

Franklin TR, Wang Z, Shin J, Jagannathan K, Suh JJ, Detre JA, O'Brien CP, Childress AR (2013) A VBM study demonstrating 'apparent' effects of a single dose of medication on T1-weighted MRIs. Brain Struct Funct 218: 97-104

Frodl T, Koutsouleris N, Bottlender R, Born C, Jäger M, Mörgenthaler M, Scheuerecker J, Zill P, Baghai T, Schüle C, Rupprecht R, Bondy B, Reiser M, Möller H-J, Meisenzahl EM (2008) Reduced gray matter brain volumes are associated with variants of the serotonin transporter gene in major depression. Mol Psychiatry 13: 1093-1101

Gaser C, Schlaug G (2003) Brain structures differ between musicians and non-musicians. J Neurosci 23: 9240-9245

Gaspar P, Cases O, Maroteaux L (2003) The developmental role of serotonin: news from mouse molecular genetics. Nat Rev Neurosci 4: 1002-1012

Gentry JJ, Barker PA, Carter BD (2004) The p75 neurotrophin receptor: multiple interactors and numerous functions. Prog Brain Res 146: 25-39

112

Getz A, Xu F, Zaidi W, Syed NI (2011) The antidepressant fluoxetine but not citalopram suppresses synapse formation and synaptic transmission between Lymnaea neurons by perturbing presynaptic and postsynaptic machinery. Eur J Neurosci 34: 221-234

Good CD, Johnsrude IS, Ashburner J, Henson RN, Friston KJ, Frackowiak RS (2001) A voxel-based morphometric study of ageing in 465 normal adult human brains. Neuroimage 14: 21-36

Greve DN, Svarer C, Fisher PM, Feng L, Hansen AE, Baare W, Rosen B, Fischl B, Knudsen GM (2014) Cortical surface-based analysis reduces bias and variance in kinetic modeling of brain PET data. Neuroimage 92: 225-236

Gunn RN, Gunn SR, Cunningham VJ (2001) Positron emission tomography compartmental models. J Cereb Blood Flow Metab 21: 635-652

Haacke EM (1999) Magnetic resonance imaging : physical principles and sequence design, New York: Wiley.

Hacker MP, Messer WS, Bachmann KA. (2009) Pharmacology principles and practice. Academic Press/Elsevier,, Amsterdam ; Boston, pp. 1 online resource (xi, 594 p.).

Haeusler D, Mien LK, Nics L, Ungersboeck J, Philippe C, Lanzenberger RR, Kletter K, Dudczak R, Mitterhauser M, Wadsak W (2009) Simple and rapid preparation of [11C]DASB with high quality and reliability for routine applications. Appl Radiat Isot 67: 1654-1660

Hajszan T, MacLusky NJ, Leranth C (2005) Short-term treatment with the antidepressant fluoxetine triggers pyramidal dendritic spine synapse formation in rat hippocampus. Eur J Neurosci 21: 1299-1303

Henningsson S, Borg J, Lundberg J, Bah J, Lindström M, Ryding E, Jovanovic H, Saijo T, Inoue M, Rosén I, Träskman-Bendz L, Farde L, Eriksson E (2009) Genetic variation in brain-derived neurotrophic factor is associated with serotonin transporter but not serotonin-1A receptor availability in men. Biological Psychiatry 66: 477-485

Hodges MR, Wehner M, Aungst J, Smith JC, Richerson GB (2009) Transgenic mice lacking serotonin neurons have severe apnea and high mortality during development. J Neurosci 29: 10341-10349

Hoexter MQ, de Souza Duran FL, D'Alcante CC, Dougherty DD, Shavitt RG, Lopes AC, Diniz JB, Deckersbach T, Batistuzzo MC, Bressan RA, Miguel EC, Busatto GF (2012) Gray matter volumes in obsessive-compulsive disorder before and after fluoxetine or cognitive-behavior therapy: a randomized clinical trial. Neuropsychopharmacology 37: 734-745

Hoflich A, Baldinger P, Savli M, Lanzenberger R, Kasper S (2012) Imaging treatment effects in depression. Rev Neurosci 23: 227-252

113

Holtmaat A, Randall J, Cane M (2013) Optical imaging of structural and functional synaptic plasticity in vivo. Eur J Pharmacol 719: 128-136

Holtmaat A, Svoboda K (2009) Experience-dependent structural synaptic plasticity in the mammalian brain. Nat Rev Neurosci 10: 647-658

Homberg JR (2012) Serotonin and decision making processes. Neurosci Biobehav Rev 36: 218-236

Hornung JP (2010) The neuroanatomy of the serotonergic system. In Handbook of the behavioral neurobiology of serotonin, pp 51-64. London: Academic

Houle S, Ginovart N, Hussey D, Meyer JH, Wilson AA (2000) Imaging the serotonin transporter with positron emission tomography: initial human studies with [11C]DAPP and [11C]DASB. Eur J Nucl Med 27: 1719-1722

Huang Y, Zheng MQ, Gerdes JM (2010) Development of effective PET and SPECT imaging agents for the serotonin transporter: has a twenty-year journey reached its destination? Current topics in medicinal chemistry 10: 1499-1526

Hyde KL, Lerch J, Norton A, Forgeard M, Winner E, Evans AC, Schlaug G (2009) The effects of musical training on structural brain development: a longitudinal study. Ann N Y Acad Sci 1169: 182-186

Ichise M, Meyer JH, Yonekura Y (2001) An introduction to PET and SPECT neuroreceptor quantification models. J Nucl Med 42: 755-763

Innis RB, Cunningham VJ, Delforge J, Fujita M, Gjedde A, Gunn RN, Holden J, Houle S, Huang SC, Ichise M, Iida H, Ito H, Kimura Y, Koeppe RA, Knudsen GM, Knuuti J, Lammertsma AA, Laruelle M, Logan J, Maguire RP, Mintun MA, Morris ED, Parsey R, Price JC, Slifstein M, Sossi V, Suhara T, Votaw JR, Wong DF, Carson RE (2007) Consensus nomenclature for in vivo imaging of reversibly binding radioligands. J Cereb Blood Flow Metab 27: 1533-1539

Ishii K, Sasaki H, Kono AK, Miyamoto N, Fukuda T, Mori E (2005) Comparison of gray matter and metabolic reduction in mild Alzheimer's disease using FDG-PET and voxel-based morphometric MR studies. Eur J Nucl Med Mol Imaging 32: 959-963

Jack CR, Lowe VJ, Senjem ML, Weigand SD, Kemp BJ, Shiung MM, Knopman DS, Boeve BF, Klunk WE, Mathis CA, Petersen RC (2008) 11C PiB and structural MRI provide complementary information in imaging of Alzheimer's disease and amnestic mild cognitive impairment. Brain 131: 665-680

Jeffery KJ, Reid IC (1997) Modifiable neuronal connections: an overview for psychiatrists. Am J Psychiatry 154: 156-164

114

Johansen-Berg H (2012) The future of functionally-related structural change assessment. Neuroimage 62: 1293-1298

Kalpouzos G, Chételat G, Baron J-C, Landeau B, Mevel K, Godeau C, Barré L, Constans J-M, Viader F, Eustache F, Desgranges B (2009) Voxel-based mapping of brain gray matter volume and glucose metabolism profiles in normal aging. Neurobiol Aging 30: 112-124

Kanda T, Ishii K, Uemura T, Miyamoto N, Yoshikawa T, Kono AK, Mori E (2008) Comparison of grey matter and metabolic reductions in frontotemporal dementia using FDG-PET and voxel-based morphometric MR studies. Eur J Nucl Med Mol Imaging 35: 2227-2234

Kandel ER (1981) Calcium and the control of synaptic strength by learning. Nature 293: 697-700

Kandel ER (2001) The molecular biology of memory storage: a dialogue between genes and synapses. Science 294: 1030-1038

Kandel ER (2004) The molecular biology of memory storage: a dialog between genes and synapses. Biosci Rep 24: 475-522

Kandel ER, Schwartz JH (1982) Molecular biology of learning: modulation of transmitter release. Science 218: 433-443

Karabeg MM, Grauthoff S, Kollert SY, Weidner M, Heiming RS, Jansen F, Popp S, Kaiser S, Lesch KP, Sachser N, Schmitt AG, Lewejohann L (2013) 5-HTT deficiency affects neuroplasticity and increases stress sensitivity resulting in altered spatial learning performance in the Morris water maze but not in the Barnes maze. PLoS One 8: e78238

Karpova NN, Pickenhagen A, Lindholm J, Tiraboschi E, Kulesskaya N, Agustsdottir A, Antila H, Popova D, Akamine Y, Bahi A, Sullivan R, Hen R, Drew LJ, Castren E (2011) Fear erasure in mice requires synergy between antidepressant drugs and extinction training. Science 334: 1731-1734

Kasai H, Fukuda M, Watanabe S, Hayashi-Takagi A, Noguchi J (2010) Structural dynamics of dendritic spines in memory and cognition. Trends Neurosci 33: 121-129

Kempton MJ, Salvador Z, Munafo MR, Geddes JR, Simmons A, Frangou S, Williams SC (2011) Structural neuroimaging studies in major depressive disorder. Meta-analysis and comparison with bipolar disorder. Arch Gen Psychiatry 68: 675-690

Klein A, Andersson J, Ardekani BA, Ashburner J, Avants B, Chiang M-C, Christensen GE, Collins DL, Gee J, Hellier P, Song JH, Jenkinson M, Lepage C, Rueckert D, Thompson P, Vercauteren T, Woods RP, Mann JJ, Parsey RV (2009) Evaluation of 14 nonlinear deformation algorithms applied to human brain MRI registration. Neuroimage 46: 786-802

115

Koponen E, Rantamaki T, Voikar V, Saarelainen T, MacDonald E, Castren E (2005) Enhanced BDNF signaling is associated with an antidepressant-like behavioral response and changes in brain monoamines. Cell Mol Neurobiol 25: 973-980

Kozisek ME, Middlemas D, Bylund DB (2008) Brain-derived neurotrophic factor and its receptor tropomyosin-related kinase B in the mechanism of action of antidepressant therapies. Pharmacol Ther 117: 30-51

Kranz GS, Kasper S, Lanzenberger R (2010) Reward and the serotonergic system. Neuroscience 166: 1023-1035

Krishnan V, Nestler EJ (2010) Linking molecules to mood: new insight into the biology of depression. Am J Psychiatry 167: 1305-1320

la Fougère C, Grant S, Kostikov A, Schirrmacher R, Gravel P, Schipper H, Reader A, Evans A, Thiel A (2010) Where in-vivo imaging meets cytoarchitectonics: The relationship between cortical thickness and neuronal density measured with high-resolution [(18)F]flumazenil-PET. Neuroimage

Lan MJ, Ogden RT, Huang YY, Oquendo MA, Sullivan GM, Miller J, Milak M, Mann JJ, Parsey RV (2014) Genetic variation in brain-derived neurotrophic factor val66met allele is associated with altered serotonin-1A receptor binding in human brain. NeuroImage

Laruelle M (2000) Imaging synaptic neurotransmission with in vivo binding competition techniques: a critical review. J Cereb Blood Flow Metab 20: 423-451

Laruelle M, Slifstein M, Huang Y (2003) Relationships between radiotracer properties and image quality in molecular imaging of the brain with positron emission tomography. Mol Imaging Biol 5: 363-375

Lenglet C, Abosch A, Yacoub E, De Martino F, Sapiro G, Harel N (2012) Comprehensive in vivo mapping of the human basal ganglia and thalamic connectome in individuals using 7T MRI. PLoS One 7: e29153

Lesch KP, Waider J (2012) Serotonin in the modulation of neural plasticity and networks: implications for neurodevelopmental disorders. Neuron 76: 175-191

Leventer RJ, Jansen A, Pilz DT, Stoodley N, Marini C, Dubeau F, Malone J, Mitchell LA, Mandelstam S, Scheffer IE, Berkovic SF, Andermann F, Andermann E, Guerrini R, Dobyns WB (2010) Clinical and imaging heterogeneity of polymicrogyria: a study of 328 patients. Brain 133: 1415-1427

Logan J, Fowler JS, Volkow ND, Wang GJ, Ding YS, Alexoff DL (1996) Distribution volume ratios without blood sampling from graphical analysis of PET data. J Cereb Blood Flow Metab 16: 834-840

116

Lusebrink F, Wollrab A, Speck O (2013) Cortical thickness determination of the human brain using high resolution 3T and 7T MRI data. Neuroimage 70: 122-131

Macqueen G, Frodl T (2010) The hippocampus in major depression: evidence for the convergence of the bench and bedside in psychiatric research? Mol Psychiatry

Maguire EA, Gadian DG, Johnsrude IS, Good CD, Ashburner J, Frackowiak RS, Frith CD (2000) Navigation-related structural change in the hippocampi of taxi drivers. Proc Natl Acad Sci U S A 97: 4398-4403

Maguire EA, Woollett K, Spiers HJ (2006) London taxi drivers and bus drivers: a structural MRI and neuropsychological analysis. Hippocampus 16: 1091-1101

Mahar I, Bambico FR, Mechawar N, Nobrega JN (2014) Stress, serotonin, and hippocampal neurogenesis in relation to depression and antidepressant effects. Neurosci Biobehav Rev 38: 173-192

Malenka RC, Nicoll RA (1999) Long-term potentiation--a decade of progress? Science 285: 1870-1874

Marques JP, Kober T, Krueger G, van der Zwaag W, Van de Moortele P-F, Gruetter R (2010) MP2RAGE, a self bias-field corrected sequence for improved segmentation and T1-mapping at high field. Neuroimage 49: 1271-1281

Martel J-C, Leduc N, Ormière A-M, Faucillon V, Danty N, Culie C, Cussac D, Newman-Tancredi A (2007) WAY-100635 has high selectivity for serotonin 5-HT(1A) versus dopamine D(4) receptors. Eur J Pharmacol 574: 15-19

Martensson J, Eriksson J, Bodammer NC, Lindgren M, Johansson M, Nyberg L, Lovden M (2012) Growth of language-related brain areas after foreign language learning. NeuroImage 63: 240-244

Mathew RJ, Partain CL (1985) Midsagittal sections of the cerebellar vermis and fourth ventricle obtained with magnetic resonance imaging of schizophrenic patients. Am J Psychiatry 142: 970-971

Mattson MP (2008) Glutamate and neurotrophic factors in neuronal plasticity and disease. Ann N Y Acad Sci 1144: 97-112

Matuskey D, Pittman B, Planeta-Wilson B, Walderhaug E, Henry S, Gallezot JD, Nabulsi N, Ding YS, Bhagwagar Z, Malison R, Carson RE, Neumeister A (2012) Age effects on serotonin receptor 1B as assessed by PET. J Nucl Med 53: 1411-1414

117

May A (2011) Experience-dependent structural plasticity in the adult human brain. Trends Cogn Sci 15: 475-482

May A, Hajak G, Ganssbauer S, Steffens T, Langguth B, Kleinjung T, Eichhammer P (2006) Structural Brain Alterations following 5 Days of Intervention: Dynamic Aspects of Neuroplasticity. Cerebral Cortex 17: 205-210

McClung CA, Nestler EJ (2008) Neuroplasticity mediated by altered gene expression. Neuropsychopharmacology 33: 3-17

Mead GE, Hsieh CF, Lee R, Kutlubaev MA, Claxton A, Hankey GJ, Hackett ML (2012) Selective serotonin reuptake inhibitors (SSRIs) for stroke recovery. The Cochrane database of systematic reviews 11: CD009286

Mehta M, Ahmed Z, Fernando SS, Cano-Sanchez P, Adayev T, Ziemnicka D, Wieraszko A, Banerjee P (2007) Plasticity of 5-HT 1A receptor-mediated signaling during early postnatal brain development. J Neurochem 101: 918-928

Michaelis L. MML (1913) Die Kinetik der Invertinwirkung. Biochem Z: 333-336

Mintun MA, Raichle ME, Kilbourn MR, Wooten GF, Welch MJ (1984) A quantitative model for the in vivo assessment of drug binding sites with positron emission tomography. Annals of neurology 15: 217-227

Moiseiwitsch JR, Lauder JM (1995) Serotonin regulates mouse cranial neural crest migration. Proc Natl Acad Sci U S A 92: 7182-7186

Mugler JP, 3rd, Brookeman JR (1990) Three-dimensional magnetization-prepared rapid gradient-echo imaging (3D MP RAGE). Magnetic resonance in medicine : official journal of the Society of Magnetic Resonance in Medicine / Society of Magnetic Resonance in Medicine 15: 152-157

Nestler EJ, Barrot M, DiLeone RJ, Eisch AJ, Gold SJ, Monteggia LM (2002) Neurobiology of depression. Neuron 34: 13-25

Nibuya M, Morinobu S, Duman RS (1995) Regulation of BDNF and trkB mRNA in rat brain by chronic electroconvulsive seizure and antidepressant drug treatments. J Neurosci 15: 7539-7547

Nitsche MA, Kuo M-F, Karrasch R, Wächter B, Liebetanz D, Paulus W (2009) Serotonin affects transcranial direct current-induced neuroplasticity in humans. Biol Psychiatry 66: 503-508

Oh H, Mormino EC, Madison C, Hayenga A, Smiljic A, Jagust WJ (2010) β-Amyloid affects frontal and posterior brain networks in normal aging. Neuroimage

118

Parsey RV, Slifstein M, Hwang DR, Abi-Dargham A, Simpson N, Mawlawi O, Guo NN, Van Heertum R, Mann JJ, Laruelle M (2000) Validation and reproducibility of measurement of 5-HT1A receptor parameters with [carbonyl-11C]WAY-100635 in humans: comparison of arterial and reference tisssue input functions. J Cereb Blood Flow Metab 20: 1111-1133

Paterson LM, Tyacke RJ, Nutt DJ, Knudsen GM (2010) Measuring endogenous 5-HT release by emission tomography: promises and pitfalls. J Cereb Blood Flow Metab 30: 1682-1706

Pereira AC, Huddleston DE, Brickman AM, Sosunov AA, Hen R, McKhann GM, Sloan R, Gage FH, Brown TR, Small SA (2007) An in vivo correlate of exercise-induced neurogenesis in the adult dentate gyrus. Proc Natl Acad Sci U S A 104: 5638-5643

Pike VW (2009) PET radiotracers: crossing the blood-brain barrier and surviving metabolism. Trends Pharmacol Sci 30: 431-440

Pilar-Cuellar F, Vidal R, Diaz A, Castro E, dos Anjos S, Pascual-Brazo J, Linge R, Vargas V, Blanco H, Martinez-Villayandre B, Pazos A, Valdizan EM (2013) Neural plasticity and proliferation in the generation of antidepressant effects: hippocampal implication. Neural plasticity 2013: 537265

Pittenger C, Duman RS (2008) Stress, depression, and neuroplasticity: a convergence of mechanisms. Neuropsychopharmacology 33: 88-109

Piubelli C, Vighini M, Mathe AA, Domenici E, Carboni L (2011) Escitalopram affects cytoskeleton and synaptic plasticity pathways in a rat gene-environment interaction model of depression as revealed by proteomics. Part II: environmental challenge. Int J Neuropsychopharmacol 14: 834-855

Polter AM, Li X (2010) 5-HT1A receptor-regulated signal transduction pathways in brain. Cellular Signalling 22: 1406-1412

Protopopescu X, Butler T, Pan H, Root J, Altemus M, Polanecsky M, McEwen B, Silbersweig D, Stern E (2008) Hippocampal structural changes across the menstrual cycle. Hippocampus 18: 985-988

Rabl U, Meyer BM, Diers K, Bartova L, Berger A, Mandorfer D, Popovic A, Scharinger C, Huemer J, Kalcher K, Pail G, Haslacher H, Perkmann T, Windischberger C, Brocke B, Sitte HH, Pollak DD, Dreher JC, Kasper S, Praschak-Rieder N, Moser E, Esterbauer H, Pezawas L (2014) Additive gene-environment effects on hippocampal structure in healthy humans. J Neurosci 34: 9917-9926

Rami-Mark C, Ungersboeck J, Haeusler D, Nics L, Philippe C, Mitterhauser M, Willeit M, Lanzenberger R, Karanikas G, Wadsak W (2013) Reliable set-up for in-loop (1)(1)C-carboxylations using Grignard reactions for the preparation of [carbonyl-(1)(1)C]WAY-100635 and [(1)(1)C]-(+)-PHNO. Appl Radiat Isot 82: 75-80

119

Rantamaki T, Hendolin P, Kankaanpaa A, Mijatovic J, Piepponen P, Domenici E, Chao MV, Mannisto PT, Castren E (2007) Pharmacologically diverse antidepressants rapidly activate brain-derived neurotrophic factor receptor TrkB and induce phospholipase-Cgamma signaling pathways in mouse brain. Neuropsychopharmacology 32: 2152-2162

Rapoport JL, Gogtay N (2008) Brain neuroplasticity in healthy, hyperactive and psychotic children: insights from neuroimaging. Neuropsychopharmacology 33: 181-197

Riccio O, Potter G, Walzer C, Vallet P, Szabó G, Vutskits L, Kiss JZ, Dayer AG (2009) Excess of serotonin affects embryonic interneuron migration through activation of the serotonin receptor 6. Mol Psychiatry 14: 280-290

Rubenstein JL (1998) Development of serotonergic neurons and their projections. Biol Psychiatry 44: 145-150

Sagi Y, Tavor I, Hofstetter S, Tzur-Moryosef S, Blumenfeld-Katzir T, Assaf Y (2012) Learning in the fast lane: new insights into neuroplasticity. Neuron 73: 1195-1203

Santarelli L, Saxe M, Gross C, Surget A, Battaglia F, Dulawa S, Weisstaub N, Lee J, Duman R, Arancio O, Belzung C, Hen R (2003) Requirement of hippocampal neurogenesis for the behavioral effects of antidepressants. Science 301: 805-809

Sarkamo T, Ripolles P, Vepsalainen H, Autti T, Silvennoinen HM, Salli E, Laitinen S, Forsblom A, Soinila S, Rodriguez-Fornells A (2014) Structural changes induced by daily music listening in the recovering brain after middle cerebral artery stroke: a voxel-based morphometry study. Front Hum Neurosci 8: 245

Saulin A, Savli M, Lanzenberger R (2012) Serotonin and molecular neuroimaging in humans using PET. Amino Acids 42: 2039-2057

Savli M, Bauer A, Mitterhauser M, Ding YS, Hahn A, Kroll T, Neumeister A, Haeusler D, Ungersboeck J, Henry S, Isfahani SA, Rattay F, Wadsak W, Kasper S, Lanzenberger R (2012) Normative database of the serotonergic system in healthy subjects using multi-tracer PET. NeuroImage 63: 447-459

Schlaug G, Norton A, Overy K, Winner E (2005) Effects of music training on the child's brain and cognitive development. Ann N Y Acad Sci 1060: 219-230

Schmidt HD, Shelton RC, Duman RS (2011) Functional biomarkers of depression: diagnosis, treatment, and pathophysiology. Neuropsychopharmacology 36: 2375-2394

Schneider F, Fink GR (2013) Funktionelle MRT in Psychiatrie und Neurologie, 2., überarbeitete und aktualisierte Auflage 2013 edn. Berlin, Heidelberg: Springer.

120

Scholz J, Klein MC, Behrens TEJ, Johansen-Berg H (2009) Training induces changes in white-matter architecture. Nat Neurosci 12: 1370-1371

Selvaraj S, Turkheimer F, Rosso L, Faulkner P, Mouchlianitis E, Roiser JP, McGuire P, Cowen PJ, Howes O (2012) Measuring endogenous changes in serotonergic neurotransmission in humans: a [11C]CUMI-101 PET challenge study. Mol Psychiatry 17: 1254-1260

Serra-Millàs M, López-Vílchez I, Navarro V, Galán A-M, Escolar G, Penadés R, Catalán R, Fañanás L, Arias B, Gastó C (2011) Changes in plasma and platelet BDNF levels induced by S-citalopram in major depression. Psychopharmacology

Shrestha S, Hirvonen J, Hines CS, Henter ID, Svenningsson P, Pike VW, Innis RB (2012) Serotonin-1A receptors in major depression quantified using PET: Controversies, confounds, and recommendations. NeuroImage 59: 3243-3251

Singh C, Bortolato M, Bali N, Godar SC, Scott AL, Chen K, Thompson RF, Shih JC (2013) Cognitive abnormalities and hippocampal alterations in monoamine oxidase A and B knockout mice. Proc Natl Acad Sci U S A 110: 12816-12821

Slifstein M, Parsey RV, Laruelle M (2000) Derivation of [(11)C]WAY-100635 binding parameters with reference tissue models: effect of violations of model assumptions. Nucl Med Biol 27: 487-492

Smith R, Chen K, Baxter L, Fort C, Lane RD (2012) Antidepressant effects of sertraline associated with volume increases in dorsolateral prefrontal cortex. J Affect Disord

Tanti A, Rainer Q, Minier F, Surget A, Belzung C (2012) Differential environmental regulation of neurogenesis along the septo-temporal axis of the hippocampus. Neuropharmacology 63: 374-384

Tardito D, Perez J, Tiraboschi E, Musazzi L, Racagni G, Popoli M (2006) Signaling pathways regulating gene expression, neuroplasticity, and neurotrophic mechanisms in the action of antidepressants: a critical overview. Pharmacol Rev 58: 115-134

Thomas C, Baker CI (2013) Teaching an adult brain new tricks: A critical review of evidence for training-dependent structural plasticity in humans. NeuroImage 73: 225-236

Tost H, Braus DF, Hakimi S, Ruf M, Vollmert C, Hohn F, Meyer-Lindenberg A (2010) Acute D2 receptor blockade induces rapid, reversible remodeling in human cortical-striatal circuits. Nat Neurosci 13: 920-922

Trachtenberg JT, Chen BE, Knott GW, Feng G, Sanes JR, Welker E, Svoboda K (2002) Long-term in vivo imaging of experience-dependent synaptic plasticity in adult cortex. Nature 420: 788-794

121

Turkheimer FE, Veronese M, Dunn J (2014) Experimental design and practical data analysis in positron emission tomography.

Upton AL, Salichon N, Lebrand C, Ravary A, Blakely R, Seif I, Gaspar P (1999) Excess of serotonin (5-HT) alters the segregation of ispilateral and contralateral retinal projections in monoamine oxidase A knock-out mice: possible role of 5-HT uptake in retinal ganglion cells during development. J Neurosci 19: 7007-7024

van Tol M-J, van der Wee NJA, van den Heuvel OA, Nielen MMA, Demenescu LR, Aleman A, Renken R, van Buchem MA, Zitman FG, Veltman DJ (2010) Regional brain volume in depression and anxiety disorders. Arch Gen Psychiatry 67: 1002-1011

Vermetten E, Vythilingam M, Southwick SM, Charney DS, Bremner JD (2003) Long-term treatment with paroxetine increases verbal declarative memory and hippocampal volume in posttraumatic stress disorder. Biol Psychiatry 54: 693-702

Vernon AC, Natesan S, Modo M, Kapur S (2011) Effect of chronic antipsychotic treatment on brain structure: a serial magnetic resonance imaging study with ex vivo and postmortem confirmation. Biol Psychiatry 69: 936-944

Vetencourt JFM, Sale A, Viegi A, Baroncelli L, De Pasquale R, O'Leary OF, Castrén E, Maffei L (2008) The antidepressant fluoxetine restores plasticity in the adult visual cortex. Science 320: 385-388

Vetencourt JFM, Tiraboschi E, Spolidoro M, Castrén E, Maffei L (2011) Serotonin triggers a transient epigenetic mechanism that reinstates adult visual cortex plasticity in rats. Eur J Neurosci 33: 49-57

Villemagne VL, Burnham S, Bourgeat P, Brown B, Ellis KA, Salvado O, Szoeke C, Macaulay SL, Martins R, Maruff P, Ames D, Rowe CC, Masters CL, Australian Imaging B, Lifestyle Research G (2013) Amyloid beta deposition, neurodegeneration, and cognitive decline in sporadic Alzheimer's disease: a prospective cohort study. Lancet Neurol 12: 357-367

Vitalis T, Cases O, Gillies K, Hanoun N, Hamon M, Seif I, Gaspar P, Kind P, Price DJ (2002) Interactions between TrkB signaling and serotonin excess in the developing murine somatosensory cortex: a role in tangential and radial organization of thalamocortical axons. J Neurosci 22: 4987-5000

Vitalis T, Cases O, Passemard S, Callebert J, Parnavelas JG (2007) Embryonic depletion of serotonin affects cortical development. Eur J Neurosci 26: 331-344

Wadsak W. MLK, Ettlinger D.E., Lanzenberger R., Haeusler D., Dudczak R., Kletter K., Mitterhauser M. (2007) Simple and fully automated preparation of [carbonyl-11C]WAY-100635. Radiochimica Acta 95: 1-6

122

Warner-Schmidt JL, Chen EY, Zhang X, Marshall JJ, Morozov A, Svenningsson P, Greengard P (2010) A role for p11 in the antidepressant action of brain-derived neurotrophic factor. Biological Psychiatry 68: 528-535

Weinberger DR, Bigelow LB, Kleinman JE, Klein ST, Rosenblatt JE, Wyatt RJ (1980) Cerebral ventricular enlargement in chronic schizophrenia. An association with poor response to treatment. Arch Gen Psychiatry 37: 11-13

Weinberger DR, Torrey EF, Wyatt RJ (1979) Cerebellar atrophy in chronic schizophrenia. Lancet 1: 718-719

Weishaupt D, Köchli VD, Marincek B (2006) How does MRI work? : an introduction to the physics and function of magnetic resonance imaging, 2nd edn. Berlin ; New York: Springer.

Whitaker-Azmitia PM (1998) Role of the neurotrophic properties of serotonin in the delay of brain maturation induced by cocaine. Ann N Y Acad Sci 846: 158-164

Whitaker-Azmitia PM (2001) Serotonin and brain development: role in human developmental diseases. Brain research bulletin 56: 479-485

Whitaker-Azmitia PM, Druse M, Walker P, Lauder JM (1996) Serotonin as a developmental signal. Behav Brain Res 73: 19-29

Wilson CC, Faber KM, Haring JH (1998) Serotonin regulates synaptic connections in the dentate molecular layer of adult rats via 5-HT1a receptors: evidence for a glial mechanism. Brain Res 782: 235-239

Witte AV, Kurten J, Jansen S, Schirmacher A, Brand E, Sommer J, Floel A (2012) Interaction of BDNF and COMT polymorphisms on paired-associative stimulation-induced cortical plasticity. J Neurosci 32: 4553-4561

Wonderlick JS, Ziegler DA, Hosseini-Varnamkhasti P, Locascio JJ, Bakkour A, van der Kouwe A, Triantafyllou C, Corkin S, Dickerson BC (2009) Reliability of MRI-derived cortical and subcortical morphometric measures: effects of pulse sequence, voxel geometry, and parallel imaging. Neuroimage 44: 1324-1333

Woodward ND, Zald DH, Ding Z, Riccardi P, Ansari MS, Baldwin RM, Cowan RL, Li R, Kessler RM (2009) Cerebral morphology and dopamine D2/D3 receptor distribution in humans: a combined [18F]fallypride and voxel-based morphometry study. Neuroimage 46: 31-38

Wright IC, McGuire PK, Poline JB, Travere JM, Murray RM, Frith CD, Frackowiak RS, Friston KJ (1995) A voxel-based method for the statistical analysis of gray and white matter density applied to schizophrenia. Neuroimage 2: 244-252

123

Wu S, Ogden RT, Mann JJ, Parsey RV (2007) Optimal metabolite curve fitting for kinetic modeling of 11C-WAY-100635. J Nucl Med 48: 926-931

Yan Y, Sheppard PC, Kasper S, Lin L, Hoare S, Kapoor A, Dodd JG, Duckworth ML, Matusik RJ (1997) Large fragment of the probasin promoter targets high levels of transgene expression to the prostate of transgenic mice. Prostate 32: 129-139

Zatorre RJ, Fields RD, Johansen-Berg H (2012) Plasticity in gray and white: neuroimaging changes in brain structure during learning. Nat Neurosci 15: 528-536

124

APPENDIX – CURRICULUM VITAE

Dr. med. univ. Christoph Kraus

Schilfgasse 2, 2452 Mannersdorf Weinbergstraße 6, 7100 Neusiedl am See mobile: +43 664 4147931 Email: [email protected] Department of Psychiatry and Psychotherapy Medical University of Vienna Waehringer Guertel 18-20 A – 1090 Vienna, Austria

Personal Details: 1.7.1983 born in Eisenstadt

unmarried

Austrian

Primary Education:

1989-1993 Primary School

1993-2001 Gymnasium Neusiedl am See, Matura, 7.6.2001

Higher Education:

2002-2009 Medical Studies at the Medical University of Vienna

2007-2009 Diploma thesis at the Department of Psychoanalysis and

Psychotherapy under supervison from Ao. Univ. Prof. Mag. Dr. Jandl-

Jager Elisabeth entitled “Violence in childhood and recognition in the

medical system of Austria”.

17.9.2009 M.D. degree of the Medical University of Vienna

2012 – present PhD Thesis in Clinical Neurosciences (N790). Title: Serotonin and

Neuroplasticity – Investigated in vivo by Positron Emission

Tomography and structural Magnetic Resonance Imaging. Supervisor:

A/Prof. Rupert Lanzenberger, Mentors: Prof. Siegfried Kasper, A/Prof.

Wolfgang Wadsak

125

Professional Education:

1.9.2011–present Clinical Training, Medical University of Vienna, Department of

Psychiatry and Psychotherapy

1.10.2012–1.10.2013 Clinical Training, Medical University of Vienna with emphasis on

treatment of forensic psychiatric Patients in the Prison “Justizanstalt

Josefstadt”

1.3.2014–30.9.2014 Psychiatric consultant service at the Vienna General Hospital (AKH)

1.10.2014-present Training in Neurology, Department of Neurology, Medical University of

Vienna, as required for specialist training

Scientific Collaborations:

2009 – present staff member in the „NEUROIMAGING LABs (NIL)“ (head Assoc.-Prof. PD Dr. Rupert Lanzenberger), Department of Psychiatry and Psychotherapy, MUW. Collaboration and Coinvestigator in clinical projects focused on neuroimaging and genetics:

2009 – 2011 Coinvestigator: Networks of Anxiety: Connectivity Analysis in Social Phobia using Functional Magnetic Resonance Imaging, OeNB procet number 12982, EK 619/2007, Principal Investigator: Assoc.-Prof. Priv.-Doz. Dipl.-Ing. Dr. Christian Windischberger, MUW Austria.

2009 – 2011 Coinvestigator: The influence of hormone replacement therapy on the cerebral serotonin-1A receptor distribution and mood in postmenopausal women. A longitudinal study using Positron Emission Tomography (PET) and the radioligand [carbonyl-11C]WAY-100635. Principal Investigator: O. Univ. Prof. Dr. h.c. mult Dr. med. S. Kasper, MUW Austria.

2010 – 2011 Coinvestigator: Effects of electroconvulsive therapy on serotonin-1A receptor binding in major depression. A longitudinal study using Positron Emission Tomography (PET) and the radioligand [carbonyl-11C]WAY-100635. Principal Investigator: Ao. Univ. Prof. Dr. Richard Frey, MUW Austria.

2010 – 2014 Coinvestigator: The influence of sex steroid hormones onserotonin transporter bindingin the human brain investigated by PET. OeNB project number 13214, EK 620/2008. Principal Investigator: Assoc.-Prof. Priv.-Doz. Dr. Rupert Lanzenberger, MUW Austria.

2011 – 2014 Coinvestigator: The Serotonin Transporter in Attention Deficit Hyperactivity Disorder Investigated with Positron Emission Tomography. OeNB project number AP13675ONB, EK 784/2009, Principal Investigator: Priv. Doz. Mag. Dr. Markus Mitterhauser, Austria.

126

2012 – present Coinvestigator: Multimodal Assessment of Neurobiological Markers for Psychiatric Disorders (MAN-BIOPSY). Research cluster “Multimodal Neuroimaging in clinical neurosciences (MMI-CNS), Medical University Vienna, University of Vienna. Principal Investigators: Assoc.-Prof. Priv.-Doz. Dr. Rupert Lanzenberger, Univ. Prof. Mag. Dr. Claus LammPhD

Other Universitary Acitvities:

2008 Initiation of the course: „Medizinsoziologie mit wechselndem Schwerpunkten: Balint-Gruppe, wissenschaftliche Methoden, Traumforschung“, together with Ao. Univ. Prof. Mag. Dr. Jandl-Jager in winterterm 08.

2009 Initiation of the course “Talks about death and dying“, starting in winterterm 09/10, togehter with Univ. Prof. Dr. Pötter, Univ. Prof. Dr. Watzke, Mag. Dr. Hladschik-Kermer, Mag. Kirchheiner

2012 – present Representative of resident psychiatrists at the Department of Psychiatry and Psychotherapy

Further Education:

2009 – 2012 Psychotherapeutic propaedeutics – H.O.P.P. Vienna 2012 Statistical Parametric Mapping Course, Center for Experimental

Medicine, Department of System Neuroscience UKE Eppendorf, Hamburg.

2012 PMOD basic Application and Small Animal Imaging Processing

Courses, Zürich 2013 – present Training in Cognitive Behavioural Therapy

1. FELLOWSHIPS

Young Scientist Association (YSA) of the Medical University of Vienna

Austrian Medical Chamber

Austrian Psychiatric and Psychotherapeutic Association

European College of Neuropsychopharmacology (ECNP)

2. TEACHING

2013 – now Student courses in clinical psychiatry and psychopharmacology at the Medical University of Vienna

127

2014 – now Nurse Training in clinical psychiatry at the School of Nursery of the Viennese Hospital Association

2015 Thesis Completion of Dr. Patrick Köck, Thesis supervision cand. med. Clauddia Winkler

3. REVIEWER FOR MANUSCRIPTS:

Molecules [IF 2014: 2.42]

World Journal of Biological Psychiatry [IF 2014: 4.23]

International Journal of Neuropsychopharmacology [IF 2014: 4.0]

Journal of Psychiatry and Neuroscience [IF 2014: 7.49]

4. PRIZES

Best Poster Award, 7th PhD Sympusium MUW, 2011

Best Poster Award, 8th PhD Symposium MUW, 2012

BSM – ÖGN – Mallinckrodt Förderungspreis Nuklearmedizin, 2012

Rafaelsen Young Investigator’s award by the International College of

Neuropsychopharmacology (CINP) 2014

WFSBP Educational Grant, 2015

ECNP Travel Award, 2015

5. CHAIRS Co-Chair, Serotonin and Neuroplasticity at the WFSBP Conference 2015

Young Programme Sub-committee for the Seoul Congress

6. SCIENTIFIC FIELD OF WORK

Serotonin and Neuroplasticity, Neuroimaging of Neuropsychiatric disorders: ADHD,

Depression, Anxiety Disorders, Genetics in Neuroimaing

128

7. PUBLICATION LIST

7.1 Articles

7.1.1 1ST Authorships:

1. Baldinger P, Kraus C, Rami-Mark C, Gryglewsky G, Kranz GS, Haeusler D, Hahn A, Wadsak W, Mitterhauser M, Rujescu D, Kasper S, Lanzenberger R. Interaction between 5-HTTLPR and 5-HT1B genotype status enhances serotonin-1A receptor binding. NeuroImage 2015 May 15;111:505-512. Epub 2015, [2014, IF: 6.36]

2. Kraus C, Baldinger P, Rami-Mark C, Gryglewsky G, Kranz GS, Haeusler D, Hahn A, Wadsak W, Mitterhauser M, Rujescu D, Kasper S, Lanzenberger R. Exploring the impact of BDNF Val66Met genotype on serotonin transporter and serotonin-1A receptor binding, PLOS-One, 2014 Sep 4;9(9) [2014: 3.23]

3. Kraus C, Ganger S, Losak J, Hahn A, Savli M, Kranz GS, Baldinger P, Windischberger C, Kasper S, Lanzenberger R, Gray matter and intrinsic network changes in the posterior cingulate cortex after selective serotonin reuptake inhibitor intake. NeuroImage 2014; 84:236-244. Epub 2013 Aug 26 [2014, IF: 6.36]

4. Kraus C, Hahn A, Savli M, Kranz GS, Baldinger P, Höflich A, Spindelegger C, Ungersböck J, Häusler D, Mitterhauser M, Windischberger C, Wadsak W, Kasper S, Lanzenberger R. Serotonin-1A receptor binding is positively associated gray matter volume – A multimodal neuroimaging study combining PET and structural MRI. NeuroImage 2012 Nov 15;63(3):1091-1098. Epub 2012 Jul 23 [2014, IF: 6.36]

5. Kraus C, Jandl-Jager E. Awareness and knowledge of child abuse amongst physicians - a descriptive study by a sample of rural Austria. Wien Klin Wochenschr. 2011 Jun;123(11-12):340-9. Epub 2011 May 4. [2014, IF: 0.84]

7.1.2 Co-authorships:

6. Seidel EM, Pfabigan D, Hahn A, Sladky R, Grahl A, Paul K, Kraus C, Küblböck M, Kranz G, Hummer A, Lanzenberger R, Windischberger C, Lamm C. Uncertainty during pain anticipation: The adaptive value of preparatory processes. Human Brain Mapping,. 2014 Oct 16. [Epub ahead of print], [2014, IF: 5.97]

7. Pfabigan D, Seidel EM, Sladky R, Hahn A, Paul K, Grahl A, Küblböck M, Kraus C, Hummer A, Kranz G, Windischberger C, Lanzenberger R, Lamm C, P300 amplitude variation is related to ventral striatum BOLD response during gain and loss anticipation: An EEG and fMRI experiment. NeuroImage 2014 Aug 1;96:12-21, Epub 2014 Apr 6 [2014, IF: 6.36].

8. Hahn A, Haeusler D, Kraus C, Höflich A, Kranz GS, Baldinger P, Savli M, Mitterhauser M, Wadsak W, Karanikas G, Kasper S, Lanzenberger R. Attenuated serotonin transporter association between dorsal raphe and ventral striatum in major depression. Human Brain Mapping Hum Brain Mapp. 2014 Aug;35(8):3857-66. Epub 2014 Jan 17. [2014, IF: 5.97]

129

9. Sladky R, Höflich A, Küblböck M, Kraus C, Baldinger P, Moser E, Lanzenberger R, Windischberger C. Disrupted efective connectivity between the amygdala and orbitofrontal cortex in social anxiety disorder during emotion discrimination revealed by dynamic causal modeling for fMRI, Cerebral Cortex, Epub 2013 Oct 9 [2014, IF: 8.66]

10. Baldinger P, Hahn A, Mitterhauser M, Kranz G, Friedl M, Wadsak W, Kraus C, Ungersböck J, Hartmann A, Giegling I, Rujescu D, Kasper S, Lanzenberger R. Impact of COMT genotype on serotonin-1A receptor binding investigated with PET. Brain Structure and Function 2014 Nov;219(6):2017-28. Epub 2013 Aug 9. [2014, IF: 5.62]

11. Hahn A, Kranz GS, Seidel EM, Sladky R, Kraus C, Küblböck M, Pfabigan DM, Hummer A, Grahl A, Ganger S, Windischberger C, Lamm C, Lanzenberger R. Comparing neural response to painful electrical stimulation with functional MRI at 3 and 7 Tesla. NeuroImage 2013 Nov 15;82:336-343. Epub 2013 Jun 12 [2014, IF: 6.36]

12. Kranz GS, Hahn A, Baldinger P, Häusler D, Philippe C, Kaufmann U, Wadsak W, Savli M, Höflich A, Kraus C, Vanicek T, Mitterhauser M, Kasper S, Lanzenberger R. Cerebral serotonin transporter asymmetry in males and male-to-female transsexuals: a PET study with [11C]DASB. Brain Structure and Function. 2014 Jan;219(1):171-83. Epub 2012 Dec 9 [2014, IF: 5.62]

13. Hahn A, Nics L, Baldinger P, Wadsak W, Savli M, Kraus C, Birkfellner W, Ungersboeck J, Haeusler D, Mitterhauser M, Karanikas G, Kasper S, Frey R, Lanzenberger R. Application of image-derived and venous input functions in major depression using [carbonyl-11C]WAY-100635. Nuclear Medicine and Biology 2013 Apr;40(3):371-7 [2014, IF: 2.41].

14. Hahn A, Wadsak W, Windischberger C, Baldinger P, Höflich A, Losak J, Nics L, Philippe C, Kranz GS, Kraus C, Mitterhauser M, Karanikas G, Kasper S, Lanzenberger R. Differential modulation of self-referential processing in the default mode network via serotonin-1A receptors. Proceedings of the National Academy of Sciences (PNAS) 2012 Feb 14;109(7):2619-24. Epub 2012 Jan 30. [2014, IF: 9.67]

15. Sladky R, Höflich A, Atanelov J, Kraus C, Baldinger P, Moser E, Lanzenberger R, Windischberger C. Increased neural habituation in the amygdala and orbitofrontal cortex in social anxiety disorder revealed by FMRI. PLOS One. 2012;7(11):e50050. Epub 2012 Nov 29. [2014, IF: 3.23]

7.2 Abstracts

7.2.1 1ST Authorships:

1. Kraus C, Savli M, Hahn A, Baldinger P, Höflich A, Mitterhauser M, Wadsak W, Windischberger C, Kasper S, Lanzenberger R. Serotonin-1A binding in the subgenual anterior cingulate cortex is associated with regional grey matter volume in striatum and temporal areas. 19th European Congress of Psychiatry (EPA), March 12-15, 2011, Vienna, Austria, European Psychiatry 2011, 26(1): P02-338

130

2. Kraus C, Hahn A, Savli M, Mitterhauser M, Wadsak W, Windischberger C, Kasper S, Lanzenberger R. A positive Correlation Between Inhibitory Serotonergic Neurotransmission and Grey Matter Volume. 7th PhD Symposium, 15-16th June 2011, Vienna Austria

3. Kraus C, Savli M, Hahn A, Höflich A, Baldinger P, Mitterhauser M, Wadsak W, Kasper S, Lanzenberger R. Molecular imaging of the serotonergic system – Serotonin, an important effector in modulating emotions. 12th International Neuropsychoanalysis Congress, June 24 – 26, 2011, Berlin, Germany

4. Kraus C, Hahn A, Savli M, Höflich A, Baldinger P, Kranz GS, Mitterhauser M, Wadsak W, Kasper S, Lanzenberger R. Serotonin-A receptor binding in the dorsal raphe nucleus is associated with hippocampal grey matter volume. 24th European College of Neuropsychopharmacology (ECNP) Congress, 3-7 September 2011, Paris, France European Neuropsychopharmacology,Vol 21, Suppl. 3, S318-319

5. Kraus C, Hahn A, Savli M, Baldinger P, Höflich A, Kranz GS, Losak J, Mitterhauser M, Wadsak W, Windischberger C, Kasper S, Lanzenberger R. Multimodal neuroimaging detects serotonin-1A receptor mediated neuroplasticity in humans. 24th IGB Workshop, Regulation of Neural Gene Expression from Development to Disease, 16-19 October 2011 Capri, Italy

6. Kraus C, Savli M, Hahn A, Baldinger P, Höflich A, Ungersboeck J, Mitterhauser M, Windischberger C, Wadsak W, Kasper S, Lanzenberger R. Serotonin-1A receptor related morphogenic signaling is associated with regional brain volumes and network neuroplasticity 20th EPA European Congress of Psychiatry, Prague, 3-6 March 2012

7. Kraus C, Vanicek T, Baldinger P, Hartmann A, Wadsak W, Lanzenberger R. Relationship between 5-HT1B receptor SNPs and 5-HT1A receptor BPND in healthy subjects measured by PET. 8th International Imaging Genetics Conference, Irvine, California, USA, January 16th–17th, 2012

8. Kraus C, Savli M, Hahn A, Höflich A, Baldinger P, Wadsak W, Windischberger C, Mitterhauser M, Kasper S, Lanzenberger R. Multimodal neuroimaging with PET and MRI to investigate the relation between serotonergic neurotransmission and regional brain volumes. ECNP Workshop on Neuropsychopharmacology for Young Scientists in Europe, 15-18 March 2012, Nice, France European Neuropsychopharmacology, Vol xx, Suppl. x, March 2012, Sxx(P.x.xxx)

9. Kraus C, Mitterhauser M, Bauer A, Ding Y-S, Henry S, Rattay F, Savli M, Lanzenberger R. A Normative database of the serotonergic system in healthy subjects using multi-tracer PET 10th PhD Symposium, June 13-14, 2012, Vienna, Austria

131

10. Kraus C, Vanicek T, Wadsak W, Baldinger P, Hartmann A, Mitterhauser M, Ungersboeck J, Rujescu D, Kasper S, Lanzenberger R. 5-HT1B receptor gene status alters 5-HT1A binding as investigated in vivo by PET and [carbonyl-11C]WAY-100635 25th European College of Neuropsychopharmacology (ECNP) Congress, 13-17 October 2012, Vienna, Austria European Neuropsychopharmacology,Vol 2x, Suppl. x, Sxxx-xxx

11. Kraus C, Ganger S, Losak J, Hahn A, Savli M, Spies M, Baldinger P, Windischberger C, Kassper S, Lanzenberger R. Rapid gray matter increases and resting state network changes after selective serotonin reuptake inhibitor administration. 11th World Congress of Biological Psychiatry (WFSBP), June 23rd- 27th, 2013, Kyoto, Japan

12. Kraus C, Kranz GS, Küblböck M, Pfabigan D, Hahn A, Sladky R, Seidel EM, Hummer A, Paul K, Ganger S, Windischberger C, Lamm C, Lanzenberger R. Reward anticipation maps comparing high and ultrahigh field functional MRI. 19th Annual Meeting of the Organization for Human Brain Mapping (HBM), June 16-20, 2012, Seattle, USA

13. Kraus C, Baldinger P, Hahn A, Rami-Mark C, Wadsak W, Mitterhauser M, Kasper S, Lanzenberger R. Brain derived neurotrophic factor genotype status impacts on hippocampal serotonin-1A receptor binding. 13th Meeting of the Austrian Neuroscience Association (ANA) Vienna, 16–19 September 2013

14. Kraus C, Kranz GS, Pfabigan DM, Hoffmann A, Hahn A, Seidel EM, Küblböck M, Spies M, Paul K, Sladky R, Kasper S, Windischberger C, Lamm C, Lanzenberger R. Parahippocampal and insular gray matter volume correlates with empathy. 29th

CINP–World Congress of Neuropsychopharmacology, 22-26 June 2014, Vancouver, Canada

15. Kraus C, Stürkat IL, Sladky R, Hahn A, Pfabigan D, Tik M, Köck P, Windischberger C, Lamm C, Lanzenberger R. Altered structural plasticity in acute and remitted depressive patients investigated with ultra-high field magnetic resonance imaging. 28th ECNP Congress 29.8. – 1.9. 2015, Amsterdam.

7.2.2 Co-authorships:

1. Baldinger P, Savli M, Kranz GS, Höflich A, Kraus C, Windischberger C, Kasper S, Lanzenberger R. Are there structural brain changes following 10 days of SSRI administration investigated by voxel-based morphometry? 19th European Congress of Psychiatry (EPA), March 12-15, 2011, Vienna, Austria European Psychiatry 2011, 26(1): P02-317

132

2. Höflich A, Philippe C, Savli M, Baldinger P, Kranz GS, Müller S, Häusler D, Zgud S, Kraus C, Wadsak W, Mitterhauser M, Lanzenberger R, Kasper S. Prediction of steady-state occupancy of the serotonin transporter based on single-dose occupancy: A [11C]DASB PET study. 19th European Congress of Psychiatry (EPA), March 12-15, 2011, Vienna, Austria European Psychiatry 2011, 26(1): P02-333

3. Sladky R, Kraus C, Tröstl J, Kasper S, Lanzenberger R, Moser E, Windischberger C. Orbitofrontal hyperactivity in social anxiety disorder patients: An fMRI study. 19th European Congress of Psychiatry (EPA), March 12-15, 2011, Vienna, Austria European Psychiatry 2011, 26(1): P01-179

4. Tröstl J, Sladky R, Hummer A, Kraus C, Moser E, Kasper S, Lanzenber R, Windischberger C. Reduced connectivity in the uncinate fiber tract between the frontal cortex and limbic subcortical areas in social phobia. 19th European Congress of Psychiatry (EPA), March 12-15, 2011, Vienna, Austria European Psychiatry 2011, 26(1): P01-182

5. Savli M, Hahn A, Häusler D, Baldinger P, Höflich A, Kraus C, Wadsak W, Mitterhauser M, Lanzenberger R, Dudczak R, Kasper S. Can the Median Raphe Nucleus predict Clinical Outcome in Patients with Major Depression? A [11C]DASB PET study Talk (Young Scientists award session) and Abstract (CD) YS-02-001 10th World Congress of Biolog. Psychiatry (WFSBP), 29.6.– 2.7.2011, Prague, Czech Republic

6. Hahn A, Lanzenberger R, Häusler D, Philippe C, Savli M, Baldinger P, Höflich A, Kraus C, Akimova E, Mitterhauser M, Wadsak W, Kasper S. Reduced Serotonin Transporter Association between Raphe Region and Ventral Striatum in Major Depressive Disorder. Talk (Free Communication) and Abstract (CD) FC-06-002 10th World Congress of Biolog. Psychiatry (WFSBP), 29.6.– 2.7.2011, Prague, Czech Republic

7. Savli M, Hahn A, Häusler D, Philippe C, Baldinger P, Höflich A, Kraus C, Kranz GS, Zgud S, Akimova E, Wadsak W, Mitterhauser M, Lanzenberger R, Dudczak R, Kasper S. The Impact of Software Motion Correction on PET Drug Occupancy Studies. 10th International Conference on Quantification of Brain Function with PET, May 24-28, 2011, Barcelona, Spain

8. Kranz GS, Kaufmann U, Ungersböck J, Hahn A, Stein P, Baldinger P, Höflich A, Zgud S, Kraus C, Losak J, Mitterhauser M, Wadsak W, Kasper S, Lanzenberger R. Estrogen and progesterone treatment affects serotoninergic neurotransmission in postmenopausal women. 17th Annual Meeting of the Organization for Human Brain Mapping, June 26-30, 2011, Quebec City, Canada

9. Sladky R, Kraus C, Tröstl J, Kasper S, Lanzenberger R, Moser E, Windischberger C. Orbitofrontal hyperactivity and habituation in social anxiety disorder patients: an

133

fMRI study. 17th Annual Meeting of the Organization for Human Brain Mapping, June 26-30, 2011, Quebec City, Canada

10. Tröstl J, Sladky R, Hummer A, Kraus C, Moser E, Kasper S, Lanzenberger R, Windischberger C. White Matter Alterations in Social Anxiety Disorder: a DTI study. 17th Annual Meeting of the Organization for Human Brain Mapping, June 26-30, 2011, Quebec City, Canada

11. Höflich A, Hahn A, Kraus C, Baldinger P, Kranz GS, Windischberger C, Kasper S, Lanzenberger R. Resting-state functional connectivity of the raphe nuclei. 24th European College of Neuropsychopharmacology (ECNP) Congress, 3-7 September 2011, Paris, France European Neuropsychopharmacology,Vol 21, Suppl. 3, S319-320

12. Savli M, Baldinger P, Kranz GS, Höflich A, Kraus C, Losak J, Windischberger C, Kasper S, Lanzenberger R. Rapid gray matter density changes after selective serotonin reuptake inhibitor administration revealed by voxel-based-morphometry. 24th European College of Neuropsychopharmacology (ECNP) Congress, 3-7 September 2011, Paris, France European Neuropsychopharmacology,Vol 21, Suppl. 3, S312

13. Savli M, Hahn A, Häusler D, Baldinger P, Höflich A, Kraus C, Wadsak W, Mitterhauser M, Lanzenberger R, Dudczak R, Kasper S. The Impact of Median Raphe Nucleus Serotonin Transporter Binding on Depression: A [11C]DASB PET study. (Abstract and Talk). Annual Meeting of the International Society of NeuroImaging in Psychiatry (ISNIP), 07-10.09.2011, Heidelberg, Germany.

14. Tröstl J, Sladky R, Hummer A, Kraus C, Moser E, Kasper S, Lanzenberger R, Windischberger C. DTI of White Matter Alterations in the Uncinate Fasciculus of Social Phobia Patients. (Talk) 28th Annual Scientific Meeting of the European Society for Magnetic Resonance in Medicine and Biology (ESMRM), October 6-8, 2011, Leipzig, Germany.

15. Baldinger P, Kraus C, Friedl M, Haeusler D, Hartmann A, Mitterhauser M, Kranz G, Rujescu D, Kasper S, Lanzenberger R. 5-HT1A receptor genotype is associated with 5-HT1A receptor binding in the healthy human brain measured by PET. ECNP Workshop on Neuropsychopharmacology for Young Scientists in Europe, 15-18 March 2012, Nice, France European Neuropsychopharmacology, Vol xx, Suppl. x, March 2012, Sxx(P.x.xxx)

16. Hahn A, Wadsak W, Windischberger C, Baldinger P, Höflich A, Losak J, Nics L, Ungersböck J, Kranz GS, Kraus C, Mitterhauser M, Karanikas G, Kasper S, Lanzenberger R. Default mode network is modulated by serotonin-1A receptors. 18th Annual Meeting of the Organization for Human Brain Mapping (HBM), June 10-14, 2012, Beijing, China NeuroImage Volume xx, Supplement x, August 2012, Pages Sxxx-Sxxx

134

17. Sladky R, Höfich A, Tröstl J, Kraus C, Baldinger P, Moser E, Lanzenberger R, Windischberger C. Increased neural habituation in the amygdala and orbitofrontal cortex in social anxiety disorder revealed by fMRI. 18th Annual Meeting of the Organization for Human Brain Mapping (HBM), June 10-14 2012, Beijing, China NeuroImage Volume xx, Supplement x, August 2012, Pages Sxx-Sx

18. Lanzenberger R, Hahn A, Ungersböck J, Friedl M, Baldinger P, Philippe C, Nics L, Kranz GS, Kraus C, Häusler D, Hartmann A, Savli M, Vanicek T, Mitterhauser M, Wadsak W, Rujescu D. Kasper S. A genetic variation of the serotonin-1B receptor affects serotonin-1A receptor in vivo binding. 18th Annual Meeting of the Organization for Human Brain Mapping (HBM), June 10-14 2012, Beijing, China NeuroImage Volume xx, Supplement x, August 2012, Pages Sxx-Sx

19. Lanzenberger R. Mitterhauser M, Hahn A, Baldinger P, Friedl M, Kraus C, Pichler S, Rujescu D, Wadsak W, Kasper S. Molecular imaging genetics of the serotonin-1A receptor investigating the common rs6295 single nucleotide polymorphism. The 9th International Symposium on Functional Neuroreceptor Mapping of the Living Brain, August 9-11, 2012, Baltimore, Maryland, USA Journal of Cerebral Blood Flow and Metabolism 2012, Suppl. Xx

20. Baldinger P, Hahn A, Wadsak W, Friedl M, Kraus C, Mitterhauser M, Ungersböck J, Rujescu D, Kasper S, Lanzenberger R. Association between Catechol-O-methyltransferase Genotyp and Serotonin-1A Receptor Binding measured via Positron Emission Tomography 25th European College of Neuropsychopharmacology (ECNP) Congress, 13-17 October 2012, Vienna, Austria European Neuropsychopharmacology,Vol 2x, Suppl. x, Sxxx-xxx

21. Baldinger P, Hahn A, Mitterhauser M, Kraus C, Wadsak W, Rujescu D, Kasper S, Lanzenberger R. Genotyp of serotonin-1B receptor affects serotonin-1A receptor

binding in vivo 10th PhD Symposium, June 13-14, 2012, Vienna, Austria

22. Höflich A, Hahn A, Atanelov J, Baldinger P, Kraus C, Windischberger C, Kasper S, Lanzenberger R. Influence of ketamine on resting-state functional connectivity in healthy volunteers - A fMRI study. 10th PhD Symposium, June 13-14, 2012, Vienna, Austria

23. Seidel EM, Pfabigan D, Hahn A, Sladky R, Grahl A, Kraus C, Kueblboeck M, Kranz G, Hummer A, Lanzenberger R, Windischberger C, Lamm C. Uncertainty during pain anticipation – An fMRI and EEG experiment. 19th Annual Meeting of the Organization for Human Brain Mapping (HBM), June 16-20, 2012, Seattle, USA

24. Hahn A, Kranz G, Seidel EM, Sladky R, Kraus C, Kueblboeck M, Pfabigan D, Hummer A, Grahl A, Ganger S, Windischberger C, Lamm C, Lanzenberger R.

135

Imaging the periaqueductal gray during pain processing at 3 and 7 Tesla functional MRI. 19th Annual Meeting of the Organization for Human Brain Mapping (HBM), June 16-20, 2012, Seattle, USA

25. Pfabigan D, Seidel EM, Sladky R., Hahn A, Paul K, Kueblboeck M, Kraus C, Hummer A, Kranz G, Windischberger C, Lanzenberger R, Lamm C. A multimodal study on gain and loss anticipation combining fMRI and EEG. 19th Annual Meeting of the Organization for Human Brain Mapping (HBM), June 16-20, 2012, Seattle, USA

26. Sladky R, Höflich A., Kuebelboeck M, Kraus C, Baldinger P, Moser E, Lanzenberger R, Windischberger C. Disrupted effective connectivity between amygdala and OFC in social anxiety disorder revealed by DCM. 19th Annual Meeting of the Organization for Human Brain Mapping (HBM), June 16-20, 2012, Seattle, USA

27. Ganger S, Hahn A, Sladky R, Kueblboeck M, Kranz G, Höflich A, Kraus C, Losak J, Spies M, Windischberger C, Lanzenberger R. Comparing techniques for resting state extraction from task data. 19th Annual Meeting of the Organization for Human Brain Mapping (HBM), June 16-20, 2012, Seattle, USA

28. Küblböck M, Hummer A, Hahn A, Hoffmann A, Kraus C, Woletz M, Komorowski A, Lanzenberger R, Lamm C, Windischberger C. Reduction in vascular confounds of 3T and 7T fMRI group analysis results using the RESCALE method. 20th Annual Meeting of the Organization for Human Brain Mapping (HBM), June 8-12, 2014, Hamburg, Germany.

29. Hoffmann A, Sladky R, Spies M, Küblböck M, Höflich A, Hummer A, Kranz GS, Woletz M, Lamm C, Lanzenberger R, Windischberger C. The Default Mode Network’s Frequency-dependency. 20th Annual Meeting of the Organization for Human Brain Mapping (HBM), June 8-12, 2014, Hamburg, Germany.

30. Woletz M, Hoffmann A, Ganger S, Seiger R, Hahn A, Lamm C, Lanzenberger R, Windischberger C. Slice-timing correction for multi-band images in SPM. 20th Annual Meeting of the Organization for Human Brain Mapping (HBM), June 8-12, 2014, Hamburg, Germany.

31. Sladky R, Minkova L, Höflich A, Kraus C, Baldinger P, Moser M, Lanzenberger R., Windischberger C. Task-dependent modulation of amygdalar connectivity in social anxiety disorder patients and healthy subjects. 20th Annual Meeting of the Organization for Human Brain Mapping (HBM), June 8-12, 2014, Hamburg, Germany.

32. Sladky R , Kraus C, Stürkat IL, Hoffmann A, Lamplmair D, Tik M, Spies M, Pfabigan D, Lamm C, Lanzenberger R, Windischberger C. Effective connectivity of amygdalar sub regions and OFC in acute and remitted MDD patients at 7T. 21st Annual Meeting of the Organization for Human Brain Mapping (HBM), June 14-18 2015, Honolulu, Hawaii, USA

136

33. The influence of high‐ dose estradiol administration on limbic brain structures and

the ventricular system Seiger R, Hahn A, Hummer A, Kranz GS, Ganger S, Woletz M, Kraus C, Sladky R, Kasper S, Windischberger C, Lanzenberger R. DGPPN Kongress – Deutsche Gesellschaft für Psychiatrie und Psychotherapie, Psychosomatik und Nervenheilkunde, November 25‐ 28 2015, Berlin

7.3 Lectures

1. Transkranielle Magnetstimulation als neue Behandlungsmethode bei Sozial Phobie. Fortbildungsveranstaltung der Klinischen Abteilung für Allgemeine Psychiatrie in Wien (Scientific seminar at the Department for Psychiatry and Pschotherapy, 21.10.10).

2. Einfluss der serotonergen Transmission auf die Struktur der Grauen Substanz. Winterseminar “Biologische Psychiatrie”, Oberlech 21.3.11 (Scientific seminar at the at the Wintersymposium “Biological Psychiatry, Oberlech, 21.03.11).

3. Serotonin-1A receptor distribution in the subgenual anterior cingulate cortex is associated with regional grey matter volume in the striatum and temporal areas. 19th European Congress of Psychiatry (EPA), March 12-15, 2011, Vienna, Austria, e-Poster presentation.

4. Inhibitory serotonergic neurotransmission correlates positively with cortical grey matter volume as revealed by PET and MRI. World Congress of Biological Psychiatry (WFSBP), May 31st, 2011, Prague, Czech Republic, free communication.

5. Serotonin-1a receptor related morphogenic signaling is associated with regional brain volumes and network neuroplasticity. 20th European Congress of Psychiatry, March 3-6, 2012, Prague, oral presentation.

6. Gen–Umwelt Interaktionen – Erfahrungsbedingte Plastizität in Gesundheit und psychiatrischen Erkrankungen, Winterseminar “Biologische Psychiatrie”, Oberlech 11.3.2013 (Scientific seminar at the at the Wintersymposium “Biological Psychiatry, Oberlech).

7. Rapid gray matter increases and resting state network changes after selective serotonin reuptake inhibitor administration, 11th World Congress of Biological Psychiatry (WFSBP), 26.6.2013, Kyoto, Japan, free communication.

8. Brain derived neurotrophic factor genotype status impacts on hippocampal serotonin-1A receptor binding, 13th Meeting of the Austrian Neuroscience Association (ANA),Vienna, 16.9.2013, oral presentation

9. Die Bedeutung des glutamatergen Systems bei psychiatrischern Erkrankungen, ÖGBP, 15.11.2013, Official Meeting of the Austrian Society for Neuropsychopharmacologgy and Biological Psychiatry.

10. Interaktionen zwischen Serotonintransporter und Serotonin-1B Rezeptor-Genotypen auf die Expression des Serotonin-1A Rezeptors, Winterseminar “Biologische Psychiatrie”, Oberlech (Scientific seminar at the at the Wintersymposium “Biological Psychiatry, Oberlech, 23.03.2014

137

11. Interaction between serotonin transporter and 1B receptor genotype status impacts on serotonin 1A receptor binding, Oral Presentation, 22.5.2014, The 10th International Symposium on Functional NeuroReceptor Mapping of the Living Brain, Egmond aan Zee, The Netherlands

12. Serotonin and Neuroplasticity, Young Scientists Section at DEVELAGE-Symposium, Medical University of Vienna, 27.11.2014

13. Prädiktion des Behandlungserfolges bei Depression mit funktioneller MRT, (Scientific seminar at the at the Wintersymposium “Biological Psychiatry, Oberlech, 17.3.2015

14. Serotonin and Neuroplasticity, Interdisciplinary Meeting at the Center of Brain Science, 27.3.2015

02/07/2015