modeling interneuron dysfunction in schizophrenia

7
Fax +41 61 306 12 34 E-Mail [email protected] www.karger.com Dev Neurosci 2012;34:152–158 DOI: 10.1159/000336731 Modeling Interneuron Dysfunction in Schizophrenia Martin J. Schmidt Karoly Mirnics Department of Psychiatry, Vanderbilt Kennedy Center, Vanderbilt University, Nashville, Tenn., USA cinations and delusions, negative symptoms including social withdrawal, anhedonia, and avolition among oth- ers, and cognitive symptoms including deficits in work- ing memory, disorganized thought, and attention. The cause(s) of schizophrenia remain(s) elusive although ge- netic and environmental disruptions in neurodevel- opment remain a consistent focus [2, 3]. Patients typi- cally first experience symptoms in adolescence or early adulthood and as many as 60% experience impairment throughout life [4]. In addition to the behavioral impact, cardiovascular disease and metabolic syndromes, includ- ing weight gain and diabetes, contribute to a mortality rate that is 2.5 times higher than the general population [5–7]. The duration and severity of the illness represent a significant burden to the patient, his/her family, the health care system, and society at large. In fact, the World Health Organization ranks schizophrenia as the third most costly neuropsychiatric illness behind only unipo- lar depression and alcohol abuse [8]. The financial and personal toll of the illness reaches from patient to popula- tion and working towards a better understanding of its development will enable more effective treatments that alleviate that strain. Two people who have influenced schizophrenia re- search and treatment from the beginning had different interpretations of its origin. Emil Kraepelin referred to it as dementia praecox linking it to other dementias with defined neuropathology like Alzheimer’s dementia. He was convinced that schizophrenia was a disorder of the brain and devoted himself to looking for pathogens and/ Key Words Schizophrenia Gene expression GAD67 Interneurons Mouse behavior Abstract Schizophrenia is a debilitating neurodevelopmental disorder affecting approximately 1% of the population and imposing a significant burden on society. One of the most replicated and well-established postmortem findings is a deficit in the ex- pression of the gene encoding the 67-kDa isoform of glutam- ic acid decarboxylase (GAD67), the primary GABA-producing enzyme in the brain. GAD67 is expressed in various classes of interneurons, with vastly different morphological, molecular, and physiological properties. Importantly, GABA system defi- cits in schizophrenia encompass multiple interneuronal sub- types, raising several important questions. First, do different classes of interneurons regulate different aspects of behavior? Second, can we model cell-type-specific GABAergic deficits in mice, and will the rodent findings translate to human physiol- ogy? Finally, will this knowledge open the door to knowledge- based approaches to treat schizophrenia? Copyright © 2012 S. Karger AG, Basel Introduction Schizophrenia is a debilitating disorder affecting ap- proximately 1% of the population [1]. Its symptoms fall into three domains: positive symptoms including hallu- Published online: May 8, 2012 Martin J. Schmidt 8128 Medical Research Building III 465 21st Avenue South Nashville, TN 37232-8548 (USA) Tel. +1 615 936 2014, E-Mail martin.j.schmidt  @  vanderbilt.edu © 2012 S. Karger AG, Basel 0378–5866/12/0343–0152$38.00/0 Accessible online at: www.karger.com/dne

Upload: karoly

Post on 03-Dec-2016

214 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Modeling Interneuron Dysfunction in Schizophrenia

Fax +41 61 306 12 34E-Mail [email protected]

Dev Neurosci 2012;34:152–158 DOI: 10.1159/000336731

Modeling Interneuron Dysfunction in Schizophrenia

Martin J. Schmidt Karoly Mirnics

Department of Psychiatry, Vanderbilt Kennedy Center, Vanderbilt University, Nashville, Tenn. , USA

cinations and delusions, negative symptoms including social withdrawal, anhedonia, and avolition among oth-ers, and cognitive symptoms including deficits in work-ing memory, disorganized thought, and attention. The cause(s) of schizophrenia remain(s) elusive although ge-netic and environmental disruptions in neurodevel-opment remain a consistent focus [2, 3] . Patients typi-cally first experience symptoms in adolescence or early adulthood and as many as 60% experience impairment throughout life [4] . In addition to the behavioral impact, cardiovascular disease and metabolic syndromes, includ-ing weight gain and diabetes, contribute to a mortality rate that is 2.5 times higher than the general population [5–7] . The duration and severity of the illness represent a significant burden to the patient, his/her family, the health care system, and society at large. In fact, the World Health Organization ranks schizophrenia as the third most costly neuropsychiatric illness behind only unipo-lar depression and alcohol abuse [8] . The financial and personal toll of the illness reaches from patient to popula-tion and working towards a better understanding of its development will enable more effective treatments that alleviate that strain.

Two people who have influenced schizophrenia re-search and treatment from the beginning had different interpretations of its origin. Emil Kraepelin referred to it as dementia praecox linking it to other dementias with defined neuropathology like Alzheimer’s dementia. He was convinced that schizophrenia was a disorder of the brain and devoted himself to looking for pathogens and/

Key Words

Schizophrenia � Gene expression � GAD67 � Interneurons � Mouse behavior

Abstract

Schizophrenia is a debilitating neurodevelopmental disorder affecting approximately 1% of the population and imposing a significant burden on society. One of the most replicated and well-established postmortem findings is a deficit in the ex-pression of the gene encoding the 67-kDa isoform of glutam-ic acid decarboxylase (GAD67), the primary GABA-producing enzyme in the brain. GAD67 is expressed in various classes of interneurons, with vastly different morphological, molecular, and physiological properties. Importantly, GABA system defi-cits in schizophrenia encompass multiple interneuronal sub-types, raising several important questions. First, do different classes of interneurons regulate different aspects of behavior? Second, can we model cell-type-specific GABAergic deficits in mice, and will the rodent findings translate to human physiol-ogy? Finally, will this knowledge open the door to knowledge-based approaches to treat schizophrenia?

Copyright © 2012 S. Karger AG, Basel

Introduction

Schizophrenia is a debilitating disorder affecting ap-proximately 1% of the population [1] . Its symptoms fall into three domains: positive symptoms including hallu-

Published online: May 8, 2012

Martin J. Schmidt 8128 Medical Research Building III 465 21st Avenue South Nashville, TN 37232-8548 (USA) Tel. +1 615 936 2014, E-Mail martin.j.schmidt   @   vanderbilt.edu

© 2012 S. Karger AG, Basel0378–5866/12/0343–0152$38.00/0

Accessible online at:www.karger.com/dne

Page 2: Modeling Interneuron Dysfunction in Schizophrenia

Modeling Interneuron Dysfunction in Schizophrenia

Dev Neurosci 2012;34:152–158 153

or pathology that might explain its symptoms [9] . In con-trast, Eugen Bleuler described it as schizophrenia, or ‘split mind’, and believed connecting with patients individu-ally was more beneficial to understanding the illness than studying neurobiology [10] . Psychosis, the defining fea-ture of schizophrenia, is impairment in distinguishing reality amongst hallucinations and delusions. This pre-sents a problem for researchers interested in mental ill-ness. Is it possible to quantify reality and study its neuro-biology scientifically?

Interestingly, a revolution in experimental psychology was taking place at about the same time Kraepelin and Bleuler were consolidating their observations. J.B. Watson [11] detailed his displeasure with the existing study of the mind in an article published in 1913. In his view, psycho-logical processes can be studied as a science only when subjective processes of introspection, consciousness, and the mind are excluded [11] . Watson’s ‘behaviorism’ would later be extended by scientists like B.F. Skinner to incor-porate those complicated ‘internal’ processes that have quantifiable outcomes such as value judgments, motiva-tion, and decision-making, which are now also thought to be dysfunctional in schizophrenia. No causal pathology exists for schizophrenia [12] . However, alterations in neu-ral connectivity and gene expression are being identified and advances in molecular biology have made it possible to tease apart the meaning of these insults in animal mod-els. Although we will never recapitulate psychosis in any model, incorporating classical views of schizophrenia and behavior with modern molecular biology allows for the empirical analysis of molecular genetic dysfunction, its effects on the brain, and on behavior.

GABA-Associated Deficits in Schizophrenia

Discovering that GABA controls dopamine release in striatal and mesolimbic circuits prompted researchers in the 1970s to theorize that GABA dysfunction could cause schizophrenia [13, 14] . GABA-associated deficits emerged in the clinical literature with the publication of studies that found reduced GABA content [15] , increased GABA receptor subunit protein levels [16, 17] but decreased re-ceptor mRNA [18] , decreased GABA transporter [19] protein levels, and altered interneuron densities [20–22] . In 1995, Akbarian et al. [23] published the first studies of gene expression in postmortem schizophrenic brain tis-sue and found a decrease in the 67-kDa isoform of glu-tamic acid decarboxylase (GAD67) mRNA in the pre-frontal cortex that cell loss could not account for. GAD67

is the primary enzyme responsible for the production of GABA in the brain [24] . The GAD67 expression deficit has become one of the most consistently replicated gene expression findings in schizophrenia across many differ-ent brain regions, patient cohorts, methods, and investi-gators, which is remarkable given the complex genetics and diverse presentation of symptoms seen in patients [25–35] .

How might this deficit develop? Several studies of the gene that encodes GAD67, GAD1, have yielded a number of single-nucleotide polymorphisms that are found more frequently in schizophrenic patients than controls [36–38] . The majority of single-nucleotide polymorphisms in each study was found in gene regulatory sequences sug-gesting a role in regulating gene expression and not pro-tein function. An analysis of patients with a GAD1 ge-netic variant suggested that DNA sequence variation can effectively regulate mRNA expression levels in the post-mortem tissue of subjects with schizophrenia [36] . A sec-ond mechanism that may contribute to decreased GAD67 expression in schizophrenia is epigenetics. Genes can be suppressed when promoters or other regulatory sequenc-es are methylated causing changes in chromatin struc-ture that prevent transcription [39] . Methylation is car-ried out by enzymes such as DNMT1 which is overex-pressed in GABAergic interneurons of schizophrenic patients and correlated with decreased GAD67 mRNA in the same cells suggesting dysfunctional epigenetic regu-lation of the gene; however, a causal relationship between increased DNMT1 and GAD1 promoter methylation has not been established conclusively in postmortem stud-ies [39–43] . Finally, it has been suggested recently that GAD67 downregulation and other GABA-associated dysfunction measured in postmortem tissue collectively reflect general dysfunction of GABA system develop-ment via changes in cell cycle regulation [44] , impair-ments in interneuron maturation [45] , and migration de-fects [20, 46] . The fact that several different mechanisms can lead to decreases in GAD67 gene expression argues that diverse insults and influences can converge, giving rise to a common GABAergic dysfunction.

Modeling GABAergic Deficits in Animal Models

Based on the postmortem data, it is clear that altera-tions in the GABAergic system are hallmark features of schizophrenia. However, postmortem studies leave two important questions unanswered. First, what part does GAD67 play in the pathophysiology of schizophrenia,

Page 3: Modeling Interneuron Dysfunction in Schizophrenia

Schmidt/Mirnics

Dev Neurosci 2012;34:152–158154

and is this related to altered behavior? Second, do differ-ent classes of interneurons regulate different aspects of behavior? Since solutions cannot easily be determined in humans, animal models are developed to address these questions empirically. Animal models offer the ability to study the causal influence of genetic and environmental manipulations at different developmental stages on cel-lular, molecular, and behavioral processes and have pro-vided insight into potential mechanisms of GABA system dysfunction in human disorders.

GABAergic cell types are so diverse that creating a no-menclature for their defining characteristics continues to be a tedious task, but they can be broadly categorized based on morphological, molecular, and physiological properties [47] . Classification is important because subtypes of inter-neurons are involved in regulating different aspects of pyramidal cell function and are concentrated in brain regions that mediate different behaviors [25, 48–51] . For example, parvalbumin (PV), neuropeptide Y, and chole-cystokinin (CCK) are molecular markers of distinct inter-neuron populations [49] . PV-positive chandelier cells syn-apse on the axon initial segments and basket cells regulate the cell soma of pyramidal cells, regulating output and in-tegration across multiple cortical and subcortical areas [52–54] . In contrast, CCK-positive basket cells regulate in-hibition of pyramidal cell soma directly via synaptic trans-mission and indirectly via modulation of other interneu-rons primarily in limbic and frontal circuits [55–59] . Furthermore, neuropeptide-Y-positive neurogliaform and Martinotti interneurons release GABA via volume trans-mission [60] and inhibit pyramidal cell distal dendrites [61] in diffuse cortical regions, the striatum, hippocampus, amygdala, and hypothalamus [50, 52, 61–63] . All of these different cell types appear to be affected in schizophrenia [21, 25, 30, 32, 33, 46] , but their contribution to the disease symptomatology remains largely unknown. Therefore, dysfunction of particular classes of interneurons could generate diverse pathophysiology and behavior. For exam-ple, PV-positive chandelier cells appear to control � -oscil-lations in the brain, which are believed to be linked to working memory [25] while CCK-positive cells may play a role in integrating neuromodulatory information and fine-tune network activity underlying mood [59] .

To systematically tease out the cell-type-specific inter-neuronal contribution to behavioral processes, our labora-tory has recently developed several novel mouse models that downregulate GAD67 in specific classes of interneu-rons [64] . Using a synthetic miRNA, GAD67 was down-regulated in CCK-positive, neuropeptide-Y-positive, PV-positive and CNR1-positive interneurons using BAC-driv-

en, fluorescent constructs. In the targeted cell populations of the transgenic animals, GAD67 expression was abol-ished by 70–95%, but without a seizure phenotype. Pre-liminary data indicate that the tested interneuronal cell types regulate different, well-defined physiological and behavioral processes. Therefore, we argue that future ther-apeutic interventions directed toward restoring the func-tion of specific interneuronal cell types might be a promis-ing approach to developing conceptually novel, knowl-edge-based therapies of schizophrenia, geared toward alleviating the most debilitating symptoms of the disease.

There are two important caveats that must be ac-knowledged when comparing the development of GABA-ergic circuitry in human and mouse. First, the propor-tion of interneurons to pyramidal cells in the human cortex is much greater than in the mouse [65] . This dis-crepancy also raises the possibility that interneuron def-icits in human could have more robust effects on behavior than what is seen in the transgenic rodent models. Sec-ond, up to 65% of human cortical interneurons arise from an alternate source of progenitors in the neocortical ven-tricular and subventricular zone that may have developed during primate evolution [66] . Regardless of these cave-ats, rodent models to assess the differentiation and mi-gration of GABAergic interneurons from the ganglionic eminences [67–69] are very informative about the neuro-biological processes underlying the developmental as-pects of schizophrenia. For example, dopamine [70] , can-nabinoids [71] and multiple schizophrenia susceptibility genes [72, 73] play roles in regulating interneuron migra-tion and synapse formation during development in mice and may contribute to GABA system developmental dis-turbances in schizophrenia [46, 74–76] .

Complementing the genetic population-based studies and expression/epigenetic data from postmortem re-search, the use of animal models is also able to shed light on the mechanisms by which GAD67 alters normal brain function. Data from rodent models suggest that GAD67 expression can be reduced by chronic dopamine D 2 recep-tor stimulation [77, 78] or acute NMDA receptor antago-nism [79] in multiple brain regions. These data mirror the ability of chronic dopamine stimulation [80] and acute NMDA receptor antagonism [81, 82] to precipitate psycho-sis in humans. Thus, the NMDA hypofunction hypothe-sis, the dopamine hypothesis, and the GABA dysfunction hypothesis of schizophrenia could be integrated with GAD67 deficiency being a player in each [29] . Further-more, some antipsychotic drugs demethylate the GAD1 promoter which may enhance their therapeutic profile in some cases [83, 84] . According to one study, treating mice

Page 4: Modeling Interneuron Dysfunction in Schizophrenia

Modeling Interneuron Dysfunction in Schizophrenia

Dev Neurosci 2012;34:152–158 155

with nicotine suppressed DNMT1 expression, demethyl-ated the GAD1 promoter, and increased GAD67 protein levels which may explain in part the high incidence of cig-arette smoking among individuals with schizophrenia [85] . This study complements the human postmortem epi-genetic data and supports epigenetic modification as a po-tentially reversible mechanism of GAD67 deficiency. However, coincidence of GAD67 rescue with symptom-atic improvement in the short and long term remains un-clear [84, 86] . Importantly, neither haloperidol nor olan-zapine reduced GABA-associated gene expression in non-human primates ruling out medication effects as a pri-mary cause of GAD67 reduction in the human postmortem brain of subjects with schizophrenia [33] .

Interneuron deficits have also emerged as principle components of other animal models of schizophrenia. For example, one of the most studied animal models is the neonatal ventral hippocampal lesion (NVHL) rat model [87–89] . Excitotoxic lesion of the ventral hippo-campus 1 week following birth (P7–P8) leads to adoles-cent onset of changes in dopaminergic systems, cortical circuitry, and behavior associated with schizophrenia [87, 90] . GABA system dysfunction is a core component of the NVHL model with multiple research groups reporting decreased GAD67 [91] and increased GABA A receptor [92, 93] gene expression as well as decreased interneuron cell number in some regions [94] . One of the main find-ings in the NVHL model is altered dopaminergic regula-tion of prefrontal circuits. O’Donnell et al. [95] found that stimulating the ventral tegmental area increased prefron-tal cortex pyramidal cell firing in lesioned animals while decreasing firing in sham-treated rats which may be the result of dysfunctional ventral tegmental area inputs onto prefrontal cortex interneurons. Later work showing a loss of D 2 receptor modulation of prefrontal cortex interneu-rons in NVHL rats supports this interpretation [96] .

At the behavioral level, psychosis cannot be readily as-certained in model animals since hallucinations and delu-sions are not quantifiable traits. In contrast, neurobiology underlying motivation, reward, social behavior, decision-making, affective behavior, and learning and memory is remarkably similar in rodents, nonhuman primates, and humans [97–101] . Human studies report that amygdalo-cortical and corticolimbic circuit disturbances in schizo-phrenia are likely related to altered motivation and deci-sion-making [102, 103] , and that basolateral amygdala in-teractions with orbitofrontal cortices (which dynamically encode the value of stimuli [97, 104–107] ) are necessary for adaptive learning and cognition [108–110] . All these disturbances can be assessed and modeled in mice, but at

a detail and time course that far exceeds the resolution limits of human studies. Importantly, the negative and cognitive symptoms related to schizophrenia are relative-ly well-suited for modeling, and their consideration rep-resents a tremendous clinical need since they are consis-tent predictors of prognosis and are not well managed with current medications [111, 112] . In fact, many schizo-phrenia-related behavioral processes have been extensive-ly investigated in rodents [110, 113–115] , shedding light on the underlying molecular mechanisms. For example, such studies revealed that dopamine regulates interneuron ac-tivity in the amygdala and basolateral amygdala afferents increase connections with interneurons in the prefrontal cortex during development. Therefore, it is possible that the dopamine and GABA systems act synergistically to regulate decision-making, reward, social and goal-direct-ed behavior, suggesting that the GABAergic deficits con-tribute to the emergence of behavioral impairments seen in schizophrenic patients [63, 99, 116–129] . Arriving at these types of knowledge using human subject-based re-search is not feasible, clearly arguing that deciphering the pathophysiology of schizophrenia will require integration of human and animal model data sets.

When considering the utility of modeling complex psychiatric disorders in animals, it is important to define the goal of the research and the context of the analyses. Recapitulating schizophrenia in an animal model is an unrealistic goal. However, we can use animal models to discover fundamental molecular and genetic processes that drive behavior. Furthermore, we can perturb the crit-ical molecular-cellular systems, and understand their contribution to various behavioral domains, which in fact might be critical for understanding the symptoms of var-ious disorders. Since the negative and cognitive behav-ioral domains are much more translatable between pa-tients and rodent models and they represent a tremendous need for therapeutic development, it seems appropriate to promote the analysis of animal models in these areas. Fur-thermore, if it is determined that GAD67 expression in particular cell types regulates specific domains of behav-ior, therapeutic strategies that target various interneuron cell types should be promising avenues for a knowledge-based approach to the treatment of schizophrenia.

Conclusions

We conclude that GABA system dysfunction in schizo-phrenia is a common, well-reproduced, multicausal and physiologically relevant finding, and might be one of the

Page 5: Modeling Interneuron Dysfunction in Schizophrenia

Schmidt/Mirnics

Dev Neurosci 2012;34:152–158156

final common pathways of the disease pathophysiology. Since dopaminergic and glutamatergic system perturba-tions lead to GAD67 downregulation and changes in in-terneuron development, it is possible that GABAergic dysfunction is a critical component of all existing theo-ries of schizophrenia. While creating a ‘schizophrenic mouse’ is impossible, understanding the contribution of specific interneuron cell types to behavior will be essen-tial for deciphering the role GAD67 deficiency plays in

the pathophysiology and symptomatology of schizophre-nia. Once we understand the functional impact of these deficits, we can develop knowledge-based therapeutic targets for the treatment of this devastating disease; just as Kraepelin suggested nearly a century ago:

‘However little it may be possible to identify human with animal brain-functions and illnesses, yet, from the effects produced by par-ticular noxae in the brains of animals, conclusions can be drawn as to the issue of like processes in man’ [9] .

References

1 Kessler RC, et al: The prevalence and corre-lates of nonaffective psychosis in the Nation-al Comorbidity Survey Replication (NCS-R). Biol Psychiatry 2005; 58: 668–676.

2 Lewis DA, Levitt P: Schizophrenia as a disor-der of neurodevelopment. Annu Rev Neuro-sci 2002; 25: 409–432.

3 Weinberger DR: Implications of normal brain development for the pathogenesis of schizophrenia. Arch Gen Psychiatry 1987; 44: 660–669.

4 Crown WH, et al: Hospitalization and total medical costs for privately insured persons with schizophrenia. Adm Policy Ment Health 2001; 28: 335–351.

5 Hennekens CH, et al: Schizophrenia and in-creased risks of cardiovascular disease. Am Heart J 2005; 150: 1115–1121.

6 Saha S, Chant D, McGrath J: A systematic re-view of mortality in schizophrenia: is the dif-ferential mortality gap worsening over time? Arch Gen Psychiatry 2007; 64: 1123–1131.

7 Masand PS, et al: Metabolic and endocrine disturbances in psychiatric disorders: a mul-tidisciplinary approach to appropriate atypi-cal antipsychotic utilization. CNS Spectr 2005; 10(suppl 14):1–15.

8 World Health Organization: The Global Burden of Disease: 2004 Update. Geneva, World Health Organization, 2008.

9 Kraepelin E: Ziele und Wege der psychia-trischen Forschung. Arbeiten aus der Deutschen Forschungsanstalt für Psychiat-rie in München (Ends and Means of Psychi-atric Research; 1922 transl). J Ment Sci 1919; 68: 115–143.

10 Zilboorg G: Eugen Bleuler and present-day psychiatry. Am J Psychiatry 1957; 114: 289–298.

11 Watson JB: Psychology as the behaviorist views it. Psychol Rev 1913; 20: 158–177.

12 Arnold SE, et al: Absence of neurodegenera-tion and neural injury in the cerebral cortex in a sample of elderly patients with schizophre-nia. Arch Gen Psychiatry 1998; 55: 225–232.

13 Van Kammen DP: � -Aminobutyric acid (GABA) and the dopamine hypothesis of schizophrenia. Am J Psychiatry 1977; 134: 138–143.

14 Roberts E: Prospects for research on schizo-phrenia. An hypothesis suggesting that there is a defect in the GABA system in schizo-phrenia. Neurosci Res Program Bull 1972; 10: 468–482.

15 Spokes EG, et al: Distribution of GABA in post-mortem brain tissue from control, psy-chotic and Huntington’s chorea subjects. J Neurol Sci 1980; 48: 303–313.

16 Hanada S, et al: [ 3 H]muscimol binding sites increased in autopsied brains of chronic schizophrenics. Life Sci 1987; 40: 259–266.

17 Benes FM, et al: Increased GABA A receptor binding in superficial layers of cingulate cor-tex in schizophrenics. J Neurosci 1992; 12: 924–929.

18 Hashimoto T, et al: Alterations in GABA-re-lated transcriptome in the dorsolateral pre-frontal cortex of subjects with schizophre-nia. Mol Psychiatry 2008; 13: 147–161.

19 Simpson MD, et al: Reduced GABA uptake sites in the temporal lobe in schizophrenia. Neurosci Lett 1989; 107: 211–215.

20 Benes FM, et al: Deficits in small interneu-rons in prefrontal and cingulate cortices of schizophrenic and schizoaffective patients. Arch Gen Psychiatry 1991; 48: 996–1001.

21 Daviss SR, Lewis DA: Local circuit neurons of the prefrontal cortex in schizophrenia: se-lective increase in the density of calbindin-immunoreactive neurons. Psychiatry Res 1995; 59: 81–96.

22 Wang AY, et al: Bipolar disorder type 1 and schizophrenia are accompanied by de-creased density of parvalbumin- and soma-tostatin-positive interneurons in the para-hippocampal region. Acta Neuropathol 2011; 122: 615–626.

23 Akbarian S, et al: Gene expression for glu-tamic acid decarboxylase is reduced without loss of neurons in prefrontal cortex of schizo-phrenics. Arch Gen Psychiatry 1995; 52: 258–266.

24 Martin DL, Rimvall K: Regulation of gam-ma-aminobutyric acid synthesis in the brain. J Neurochem 1993; 60: 395–407.

25 Lewis DA, Hashimoto T, Volk DW: Cortical inhibitory neurons and schizophrenia. Nat Rev Neurosci 2005; 6: 312–324.

26 Knable MB, et al: Molecular abnormalities in the major psychiatric illnesses: Classifica-tion and Regression Tree (CRT) analysis of post-mortem prefrontal markers. Mol Psy-chiatry 2002; 7: 392–404.

27 Akbarian S, Huang HS: Molecular and cel-lular mechanisms of altered GAD1/GAD67 expression in schizophrenia and related dis-orders. Brain Res Rev 2006; 52: 293–304.

28 Costa E, et al: A GABAergic cortical deficit dominates schizophrenia pathophysiology. Crit Rev Neurobiol 2004; 16: 1–23.

29 Kalkman HO, Loetscher E: GAD(67): the link between the GABA-deficit hypothesis and the dopaminergic- and glutamatergic theories of psychosis. J Neural Transm 2003; 110: 803–812.

30 Mirnics K, et al: Molecular characterization of schizophrenia viewed by microarray anal-ysis of gene expression in prefrontal cortex. Neuron 2000; 28: 53–67.

31 Fatemi SH, et al: GABAergic dysfunction in schizophrenia and mood disorders as re-flected by decreased levels of glutamic acid decarboxylase 65 and 67 kDa and Reelin pro-teins in cerebellum. Schizophr Res 2005; 72: 109–122.

32 Hashimoto T, et al: Gene expression deficits in a subclass of GABA neurons in the pre-frontal cortex of subjects with schizophre-nia. J Neurosci 2003; 23: 6315–6326.

33 Hashimoto T, et al: Alterations in GABA-re-lated transcriptome in the dorsolateral pre-frontal cortex of subjects with schizophre-nia. Mol Psychiatry 2008; 13: 147–161.

34 Hashimoto T, et al: Conserved regional pat-terns of GABA-related transcript expres-sion in the neocortex of subjects with schizophrenia. Am J Psychiatry 2008; 165: 479–489.

35 Guidotti A, et al: Decrease in reelin and glu-tamic acid decarboxylase67 (GAD67) ex-pression in schizophrenia and bipolar disor-der: a postmortem brain study. Arch Gen Psychiatry 2000; 57: 1061–1069.

36 Straub RE, et al: Allelic variation in GAD1 (GAD67) is associated with schizophrenia and influences cortical function and gene ex-pression. Mol Psychiatry 2007; 12: 854–869.

Page 6: Modeling Interneuron Dysfunction in Schizophrenia

Modeling Interneuron Dysfunction in Schizophrenia

Dev Neurosci 2012;34:152–158 157

37 Du J, et al: Comprehensive analysis of poly-morphisms throughout GAD1 gene: a fami-ly-based association study in schizophrenia. J Neural Transm 2008; 115: 513–519.

38 Addington AM, et al: GAD1 (2q31.1), which encodes glutamic acid decarboxylase (GAD67), is associated with childhood-onset schizophrenia and cortical gray matter vol-ume loss. Mol Psychiatry 2005; 10: 581–588.

39 Veldic M, et al: DNA-methyltransferase 1 mRNA is selectively overexpressed in telen-cephalic GABAergic interneurons of schizo-phrenia brains. Proc Natl Acad Sci USA 2004; 101: 348–353.

40 Huang HS, Akbarian S: GAD1 mRNA ex-pression and DNA methylation in prefrontal cortex of subjects with schizophrenia. PLoS One 2007; 2:e809.

41 Ruzicka WB, et al: Selective epigenetic alter-ation of layer I GABAergic neurons isolated from prefrontal cortex of schizophrenia pa-tients using laser-assisted microdissection. Mol Psychiatry 2007; 12: 385–397.

42 Veldic M, et al: Epigenetic mechanisms ex-pressed in basal ganglia GABAergic neurons differentiate schizophrenia from bipolar dis-order. Schizophr Res 2007; 91: 51–61.

43 Veldic M, et al: In psychosis, cortical inter-neurons overexpress DNA-methyltransfer-ase 1. Proc Natl Acad Sci USA 2005; 102: 2152–2157.

44 Benes FM: Regulation of cell cycle and DNA repair in post-mitotic GABA neurons in psy-chotic disorders. Neuropharmacology 2011; 60: 1232–1242.

45 Hyde TM, et al: Expression of GABA signal-ing molecules KCC2, NKCC1, and GAD1 in cortical development and schizophrenia. J Neurosci 2011; 31: 11088–11095.

46 Ikeda K, et al: Distribution of neuropeptide Y interneurons in the dorsal prefrontal cor-tex of schizophrenia. Prog Neuropsycho-pharmacol Biol Psychiatry 2004; 28: 379–383.

47 Ascoli GA, et al: Petilla terminology: no-menclature of features of GABAergic inter-neurons of the cerebral cortex. Nat Rev Neu-rosci 2008; 9: 557–568.

48 Kawaguchi Y, Aosaki T, Kubota Y: Choliner-gic and GABAergic interneurons in the stri-atum. Nihon Shinkei Seishin Yakurigaku Zasshi 1997; 17: 87–90.

49 Kubota Y, et al: Selective coexpression of multiple chemical markers defines discrete populations of neocortical GABAergic neu-rons. Cereb Cortex 2011; 21: 1803–1817.

50 Tepper JM, Bolam JP: Functional diversity and specificity of neostriatal interneurons. Curr Opin Neurobiol 2004; 14: 685–692.

51 Sosulina L, Graebenitz S, Pape HC: GABAer-gic interneurons in the mouse lateral amyg-dala: a classification study. J Neurophysiol 2010; 104: 617–626.

52 Markram H, et al: Interneurons of the neo-cortical inhibitory system. Nat Rev Neurosci 2004; 5: 793–807.

53 Woodruff AR, Sah P: Networks of parvalbu-min-positive interneurons in the basolateral amygdala. J Neurosci 2007; 27: 553–563.

54 Woodruff AR, Anderson SA, Yuste R: The enigmatic function of chandelier cells. Front Neurosci 2010; 4: 201.

55 Meziane H, et al: Distribution of cholecys-tokinin immunoreactivity in the BALB/c mouse forebrain: an immunocytochemical study. J Chem Neuroanat 1997; 12: 191–209.

56 Hornung JP, De Tribolet N, Tork I: Morphol-ogy and distribution of neuropeptide-con-taining neurons in human cerebral cortex. Neuroscience 1992; 51: 363–375.

57 Geola FL, et al: Regional distribution of cho-lecystokinin-like immunoreactivity in the human brain. J Clin Endocrinol Metab 1981; 53: 270–275.

58 Foldy C, et al: Cell type-specific gating of perisomatic inhibition by cholecystokinin. Nat Neurosci 2007; 10: 1128–1130.

59 Freund TF: Interneuron diversity series: rhythm and mood in perisomatic inhibition. Trends Neurosci 2003; 26: 489–495.

60 Olah S, et al: Regulation of cortical microcir-cuits by unitary GABA-mediated volume transmission. Nature 2009; 461: 1278–1281.

61 Karagiannis A, et al: Classification of NPY-expressing neocortical interneurons. J Neu-rosci 2009; 29: 3642–3659.

62 Partridge JG, et al: Excitatory and inhibitory synapses in neuropeptide Y-expressing stria-tal interneurons. J Neurophysiol 2009; 102: 3038–3045.

63 Truitt WA, et al: Anxiety-like behavior is modulated by a discrete subpopulation of in-terneurons in the basolateral amygdala. Neuroscience 2009; 160: 284–294.

64 Garbett KA, et al: Novel animal models for studying complex brain disorders: BAC-driv-en miRNA-mediated in vivo silencing of gene expression. Mol Psychiatry 2010; 15: 987–995.

65 Jones EG: The origins of cortical interneu-rons: mouse versus monkey and human. Cereb Cortex 2009; 19: 1953–1956.

66 Letinic K, Zoncu R, Rakic P: Origin of GABAergic neurons in the human neocor-tex. Nature 2002; 417: 645–649.

67 Metin C, et al: Modes and mishaps of neuro-nal migration in the mammalian brain. J Neurosci 2008; 28: 11746–11752.

68 Flames N, et al: Delineation of multiple sub-pallial progenitor domains by the combina-torial expression of transcriptional codes. J Neurosci 2007; 27: 9682–9695.

69 Long JE, et al: Dlx1&2 and Mash1 transcrip-tion factors control MGE and CGE pattern-ing and differentiation through parallel and overlapping pathways. Cereb Cortex 2009; 19(suppl 1):96–106.

70 Crandall JE, et al: Dopamine receptor activa-tion modulates GABA neuron migration from the basal forebrain to the cerebral cor-tex. J Neurosci 2007; 27: 3813–3822.

71 Galve-Roperh I, et al: The endocannabinoid system and the regulation of neural develop-ment: potential implications in psychiatric disorders. Eur Arch Psychiatry Clin Neuro-sci 2009; 259: 371–382.

72 Harrison PJ, Weinberger DR: Schizophrenia genes, gene expression, and neuropathology: on the matter of their convergence. Mol Psy-chiatry 2005; 10: 40–68;image 5.

73 Neddens J, et al: Conserved interneuron-specific ErbB4 expression in frontal cortex of rodents, monkeys, and humans: implica-tions for schizophrenia. Biol Psychiatry 2011; 70: 636–645.

74 Marenco S, et al: Genetic association of ErbB4 and human cortical GABA levels in vivo. J Neurosci 2011; 31: 11628–11632.

75 Eastwood SL, Harrison PJ: Interstitial white matter neuron density in the dorsolateral prefrontal cortex and parahippocampal gy-rus in schizophrenia. Schizophr Res 2005; 79: 181–188.

76 Connor CM, Guo Y, Akbarian S: Cingulate white matter neurons in schizophrenia and bipolar disorder. Biol Psychiatry 2009; 66: 486–493.

77 Lindefors N: Dopaminergic regulation of glutamic acid decarboxylase mRNA expres-sion and GABA release in the striatum: a re-view. Prog Neuropsychopharmacol Biol Psy-chiatry 1993; 17: 887–903.

78 Laprade N, Soghomonian JJ: Differential regulation of mRNA levels encoding for the two isoforms of glutamate decarboxylase (GAD65 and GAD67) by dopamine recep-tors in the rat striatum. Brain Res Mol Brain Res 1995; 34: 65–74.

79 Qin ZH, Zhang SP, Weiss B: Dopaminergic and glutamatergic blocking drugs differen-tially regulate glutamic acid decarboxylase mRNA in mouse brain. Brain Res Mol Brain Res 1994; 21: 293–302.

80 Sato M, Numachi Y, Hamamura T: Relapse of paranoid psychotic state in methamphet-amine model of schizophrenia. Schizophr Bull 1992; 18: 115–122.

81 Javitt DC, Zukin SR: Recent advances in the phencyclidine model of schizophrenia. Am J Psychiatry 1991; 148: 1301–1308.

82 Krystal JH, et al: Subanesthetic effects ofthe noncompetitive NMDA antagonist, ke-tamine, in humans. Psychotomimetic, per-ceptual, cognitive, and neuroendocrine re-sponses. Arch Gen Psychiatry 1994; 51: 199–214.

83 Guidotti A, et al: Characterization of the ac-tion of antipsychotic subtypes on valpro-ate-induced chromatin remodeling. Trends Pharmacol Sci 2009; 30: 55–60.

84 Guidotti A, et al: Epigenetic GABAergic tar-gets in schizophrenia and bipolar disorder. Neuropharmacology 2011; 60: 1007–1016.

85 Satta R, et al: Nicotine decreases DNA meth-yltransferase 1 expression and glutamic acid decarboxylase 67 promoter methylation in GABAergic interneurons. Proc Natl Acad Sci USA 2008; 105: 16356–16361.

Page 7: Modeling Interneuron Dysfunction in Schizophrenia

Schmidt/Mirnics

Dev Neurosci 2012;34:152–158158

86 Thomsen MS, et al: � (7) Nicotinic acetylcho-line receptor activation prevents behavioral and molecular changes induced by repeated phencyclidine treatment. Neuropharmacol-ogy 2009; 56: 1001–1009.

87 O’Donnell P: Cortical disinhibition in the neonatal ventral hippocampal lesion model of schizophrenia: new vistas on possible therapeutic approaches. Pharmacol Ther 2012; 133: 19–25.

88 Tseng KY, Chambers RA, Lipska BK: The neonatal ventral hippocampal lesion as a heuristic neurodevelopmental model of schizophrenia. Behav Brain Res 2009; 204: 295–305.

89 Lipska BK, Weinberger DR: A neurodevelop-mental model of schizophrenia: neonatal disconnection of the hippocampus. Neuro-tox Res 2002; 4: 469–475.

90 Lipska BK, Jaskiw GE, Weinberger DR: Post-pubertal emergence of hyperresponsiveness to stress and to amphetamine after neonatal excitotoxic hippocampal damage: a potential animal model of schizophrenia. Neuropsy-chopharmacology 1993; 9: 67–75.

91 Lipska BK, et al: Gene expression in dopa-mine and GABA systems in an animal model of schizophrenia: effects of antipsychotic drugs. Eur J Neurosci 2003; 18: 391–402.

92 Mitchell CP, Grayson DR, Goldman MB: Neonatal lesions of the ventral hippocampal formation alter GABA-A receptor subunit mRNA expression in adult rat frontal pole. Biol Psychiatry 2005; 57: 49–55.

93 Endo K, et al: Alterations in GABA(A) recep-tor expression in neonatal ventral hippo-campal lesioned rats: comparison of prepu-bertal and postpubertal periods. Synapse 2007; 61: 357–366.

94 Francois J, et al: Selective reorganization of GABAergic transmission in neonatal ventral hippocampal-lesioned rats. Int J Neuropsy-chopharmacol 2009; 12: 1097–1110.

95 O’Donnell P, et al: Neonatal hippocampal damage alters electrophysiological proper-ties of prefrontal cortical neurons in adult rats. Cereb Cortex 2002; 12: 975–982.

96 Tseng KY, et al: A neonatal ventral hippo-campal lesion causes functional deficits in adult prefrontal cortical interneurons. J Neurosci 2008; 28: 12691–12699.

97 Morrison SE, Salzman CD: Re-valuing the amygdala. Curr Opin Neurobiol 2010; 20: 221–230.

98 Bissonette GB, et al: Double dissociation of the effects of medial and orbital prefrontal cortical lesions on attentional and affective shifts in mice. J Neurosci 2008; 28: 11124–11130.

99 Schultz W: Potential vulnerabilities of neu-ronal reward, risk, and decision mechanisms to addictive drugs. Neuron 2011; 69: 603–617.

100 Robbins TW: Chemical neuromodulation of frontal-executive functions in humans and other animals. Exp Brain Res 2000; 133: 130–138.

101 Seymour B, Dolan R: Emotion, decision making, and the amygdala. Neuron 2008; 58: 662–671.

102 Benes FM: Amygdalocortical circuitry in schizophrenia: from circuits to molecules. Neuropsychopharmacology 2009; 35: 239–257.

103 Waltz JA, et al: Abnormal responses to monetary outcomes in cortex, but not in the basal ganglia, in schizophrenia. Neuropsy-chopharmacology 2010; 35: 2427–2439.

104 Murray EA, Wise SP: Interactions between orbital prefrontal cortex and amygdala: ad-vanced cognition, learned responses and instinctive behaviors. Curr Opin Neurobiol 2010; 20: 212–220.

105 Gallagher M, McMahan RW, Schoenbaum G: Orbitofrontal cortex and representation of incentive value in associative learning. J Neurosci 1999; 19: 6610–6614.

106 Schoenbaum G, et al: Encoding predicted outcome and acquired value in orbitofron-tal cortex during cue sampling depends upon input from basolateral amygdala. Neuron 2003; 39: 855–867.

107 Saddoris MP, Gallagher M, Schoenbaum G: Rapid associative encoding in basolateral amygdala depends on connections with or-bitofrontal cortex. Neuron 2005; 46: 321–331.

108 Kehagia AA, Murray GK, Robbins TW: Learning and cognitive flexibility: fronto-striatal function and monoaminergic mod-ulation. Curr Opin Neurobiol 2010; 20: 199–204.

109 Gruber AJ, et al: More is less: a disinhibited prefrontal cortex impairs cognitive flexi-bility. J Neurosci 2010; 30: 17102–17110.

110 Schoenbaum G, Esber GR: How do you (es-timate you will) like them apples? Integra-tion as a defining trait of orbitofrontal function. Curr Opin Neurobiol 2010; 20: 205–211.

111 Green MF: What are the functional con-sequences of neurocognitive deficits in schizophrenia? Am J Psychiatry 1996; 153: 321–330.

112 Kirkpatrick B, et al: The NIMH-MATRICS consensus statement on negative symp-toms. Schizophr Bull 2006; 32: 214–219.

113 Cardinal RN, et al: Emotion and motiva-tion: the role of the amygdala, ventral stria-tum, and prefrontal cortex. Neurosci Biobehav Rev 2002; 26: 321–352.

114 Donaldson ZR, Young LJ: Oxytocin, vaso-pressin, and the neurogenetics of sociality. Science 2008; 322: 900–904.

115 Dalley JW, Cardinal RN, Robbins TW: Pre-frontal executive and cognitive functions in rodents: neural and neurochemical sub-strates. Neurosci Biobehav Rev 2004; 28: 771–784.

116 Marowsky A, et al: A specialized subclass of interneurons mediates dopaminergic facil-itation of amygdala function. Neuron 2005; 48: 1025–1037.

117 Lisman JE, et al: Circuit-based framework for understanding neurotransmitter and risk gene interactions in schizophrenia. Trends Neurosci 2008; 31: 234–242.

118 Diaz MR, et al: Dopamine D 3 -like receptors modulate anxiety-like behavior and regu-late GABAergic transmission in the rat lat-eral/basolateral amygdala. Neuropsycho-pharmacology 2011; 36: 1090–1103.

119 Zweifel LS, et al: Activation of dopamine neurons is critical for aversive conditioning and prevention of generalized anxiety. Nat Neurosci 2011; 14: 620–626.

120 de Oliveira AR, et al: Conditioned fear is modulated by D(2) receptor pathway con-necting the ventral tegmental area and ba-solateral amygdala. Neurobiol Learn Mem 2011; 95: 37–45.

121 Boulougouris V, Dalley JW, Robbins TW: Effects of orbitofrontal, infralimbic and prelimbic cortical lesions on serial spatial reversal learning in the rat. Behav Brain Res 2007; 179: 219–228.

122 Gendreau PL, et al: D 2 -like dopamine re-ceptor mediation of social-emotional reac-tivity in a mouse model of anxiety: strain and experience effects. Neuropsychophar-macology 1998; 18: 210–221.

123 Wall PM, et al: Infralimbic D 2 receptor in-fluences on anxiety-like behavior and ac-tive memory/attention in CD-1 mice. Prog Neuropsychopharmacol Biol Psychiatry 2003; 27: 395–410.

124 Truitt WA, et al: From anxiety to autism: spectrum of abnormal social behaviors modeled by progressive disruption of in-hibitory neuronal function in the basolat-eral amygdala in Wistar rats. Psychophar-macology (Berl) 2007; 191: 107–118.

125 Likhtik E, et al: Amygdala intercalated neu-rons are required for expression of fear ex-tinction. Nature 2008; 454: 642–645.

126 de la Mora MP, et al: Role of dopamine re-ceptor mechanisms in the amygdaloid modulation of fear and anxiety: structural and functional analysis. Prog Neurobiol 2010; 90: 198–216.

127 Takahashi YK, et al: Expectancy-related changes in firing of dopamine neurons de-pend on orbitofrontal cortex. Nat Neurosci 2011; 14: 1590–1597.

128 Cunningham MG, Bhattacharyya S, Benes FM: Increasing interaction of amygdalar afferents with GABAergic interneurons be-tween birth and adulthood. Cereb Cortex 2008; 18: 1529–1535.

129 Leeson VC, et al: Discrimination learning, reversal, and set-shifting in first-episode schizophrenia: stability over six years and specific associations with medication type and disorganization syndrome. Biol Psy-chiatry 2009; 66: 586–593.