deletion of a single allele of the pex11 gene is ...of catalase remained unchanged in neurons from...

16
INTRODUCTION Peroxisomes are dynamic organelles that can arise de novo (membrane formation). They mature by the import of matrix proteins, proliferate and divide, depending on developmental and environmental conditions. This process is called peroxisomal biogenesis and is mediated by more than 30 different proteins, classified as ‘peroxins’. These proteins are encoded by PEX genes and were numbered according to their date of discovery (Lanyon- Hogg et al., 2010; Wolf et al., 2010). Among them, the PEX11 protein has been implicated in the regulation of peroxisome proliferation and division, because overexpression of PEX11 is sufficient to induce peroxisomal proliferation (Schrader et al., 1998), whereas its disruption reduces the total number of this organelle. It was suggested that PEX11 binds to the peroxisomal membrane of pre- existing organelles and mediates the formation of subdomains, followed by protrusion, extension, segmentation and constriction of the plasma membrane (Delille et al., 2010). In addition, the Pex11p protein has been shown to mediate the transport of medium-chain fatty acids across the peroxisomal membrane, a process that indirectly affects peroxisome number and size in Saccharomyces cerevisiae (van Roermund et al., 2000). Furthermore, the overexpression of PEX11 increases the expression of peroxisome-related genes, such as those encoding PEX5, catalase, PMP70 and PPAR, in Xenopus laevis (Fox et al., 2011). The importance of peroxisome biogenesis for cell homeostasis is clearly demonstrated by the severe clinical phenotype of peroxisomal biogenesis disorders (PBDs). PBDs are inherited in an autosomal recessive manner and are caused by mutations in at least 12 PEX genes (classified into 12 complementation groups) (Weller et al., 2003). The disease is characterized by disturbances in both developmental and metabolic homeostasis, predominantly in the liver, kidney and brain (Faust et al., 2005). The clinical phenotype varies widely, with Zellweger syndrome (ZS) at the most severe end of the spectrum (survival of less than 1 year) followed by neonatal adrenoleukodystrophy (NALD) and infantile Refsum’s disease (IRD) as milder forms in which the patients survive into the second decade (Gärtner, 2003). In addition, some features of the disease (renal cysts) are not found in the milder forms. The severity of the symptoms is proposed to depend on the nature of PEX mutation (Brosius and Gärtner, 2002), e.g. a premature termination codon in the protein will lead to a total loss of function, but in cases in which the mutant protein is misfolded or unable to interact with other peroxins (Geisbrecht et al., 1998; Tamura et al., 1998) or contains a residual import activity (Imamura et al., 2001; Tamura et al., 2001; Walter et al., 2001), peroxisomal Disease Models & Mechanisms 125 Disease Models & Mechanisms 5, 125-140 (2012) doi:10.1242/dmm.007708 1 Institute for Anatomy and Cell Biology II, Division of Medical Cell Biology, University of Giessen, 35385 Giessen, Germany *Authors for correspondence ([email protected]; [email protected]) Received 7 February 2011; Accepted 29 July 2011 © 2012. Published by The Company of Biologists Ltd This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial Share Alike License (http://creativecommons.org/licenses/by-nc-sa/3.0), which permits unrestricted non-commercial use, distribution and reproduction in any medium provided that the original work is properly cited and all further distributions of the work or adaptation are subject to the same Creative Commons License terms. SUMMARY Impaired neuronal migration and cell death are commonly observed in patients with peroxisomal biogenesis disorders (PBDs), and in mouse models of this diseases. In Pex11-deficient mice, we observed that the deletion of a single allele of the Pex11 gene (Pex11 +/– heterozygous mice) caused cell death in primary neuronal cultures prepared from the neocortex and cerebellum, although to a lesser extent as compared with the homozygous- null animals (Pex11 –/– mice). In corresponding brain sections, cell death was rare, but differences between the genotypes were similar to those found in vitro. Because PEX11 has been implicated in peroxisomal proliferation, we searched for alterations in peroxisomal abundance in the brain of heterozygous and homozygous Pex11-null mice compared with wild-type animals. Deletion of one allele of the Pex11 gene slightly increased the abundance of peroxisomes, whereas the deletion of both alleles caused a 30% reduction in peroxisome number. The size of the peroxisomal compartment did not correlate with neuronal death. Similar to cell death, neuronal development was delayed in Pex11 +/– mice, and to a further extent in Pex11 –/– mice, as measured by a reduced mRNA and protein level of synaptophysin and a reduced protein level of the mature isoform of MAP2. Moreover, a gradual increase in oxidative stress was found in brain sections and primary neuronal cultures from wild-type to heterozygous to homozygous Pex11-deficient mice. SOD2 was upregulated in neurons from Pex11 +/– mice, but not from Pex11 –/– animals, whereas the level of catalase remained unchanged in neurons from Pex11 +/– mice and was reduced in those from Pex11 –/– mice, suggesting a partial compensation of oxidative stress in the heterozygotes, but a failure thereof in the homozygous Pex11 –/– brain. In conclusion, we report the alterations in the brain caused by the deletion of a single allele of the Pex11 gene. Our data might lead to the reconsideration of the clinical treatment of PBDs and the common way of using knockout mouse models for studying autosomal recessive diseases. Deletion of a single allele of the Pex11 gene is sufficient to cause oxidative stress, delayed differentiation and neuronal death in mouse brain Barbara Ahlemeyer 1, *, Magdalena Gottwald 1 and Eveline Baumgart-Vogt 1, * RESEARCH ARTICLE Disease Models & Mechanisms DMM

Upload: others

Post on 02-Feb-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Deletion of a single allele of the Pex11 gene is ...of catalase remained unchanged in neurons from Pex11 +/– mice and was reduced in those from Pex11 –/– mice, suggesting a partial

INTRODUCTIONPeroxisomes are dynamic organelles that can arise de novo(membrane formation). They mature by the import of matrixproteins, proliferate and divide, depending on developmental andenvironmental conditions. This process is called peroxisomalbiogenesis and is mediated by more than 30 different proteins,classified as ‘peroxins’. These proteins are encoded by PEX genesand were numbered according to their date of discovery (Lanyon-Hogg et al., 2010; Wolf et al., 2010). Among them, the PEX11 proteinhas been implicated in the regulation of peroxisome proliferationand division, because overexpression of PEX11 is sufficient toinduce peroxisomal proliferation (Schrader et al., 1998), whereasits disruption reduces the total number of this organelle. It wassuggested that PEX11 binds to the peroxisomal membrane of pre-existing organelles and mediates the formation of subdomains,followed by protrusion, extension, segmentation and constrictionof the plasma membrane (Delille et al., 2010). In addition, the

Pex11p protein has been shown to mediate the transport ofmedium-chain fatty acids across the peroxisomal membrane, aprocess that indirectly affects peroxisome number and size inSaccharomyces cerevisiae (van Roermund et al., 2000). Furthermore,the overexpression of PEX11 increases the expression ofperoxisome-related genes, such as those encoding PEX5, catalase,PMP70 and PPAR, in Xenopus laevis (Fox et al., 2011).

The importance of peroxisome biogenesis for cell homeostasisis clearly demonstrated by the severe clinical phenotype ofperoxisomal biogenesis disorders (PBDs). PBDs are inherited in anautosomal recessive manner and are caused by mutations in at least12 PEX genes (classified into 12 complementation groups) (Welleret al., 2003). The disease is characterized by disturbances in bothdevelopmental and metabolic homeostasis, predominantly in theliver, kidney and brain (Faust et al., 2005). The clinical phenotypevaries widely, with Zellweger syndrome (ZS) at the most severeend of the spectrum (survival of less than 1 year) followed byneonatal adrenoleukodystrophy (NALD) and infantile Refsum’sdisease (IRD) as milder forms in which the patients survive intothe second decade (Gärtner, 2003). In addition, some features ofthe disease (renal cysts) are not found in the milder forms. Theseverity of the symptoms is proposed to depend on the nature ofPEX mutation (Brosius and Gärtner, 2002), e.g. a prematuretermination codon in the protein will lead to a total loss offunction, but in cases in which the mutant protein is misfolded orunable to interact with other peroxins (Geisbrecht et al., 1998;Tamura et al., 1998) or contains a residual import activity (Imamuraet al., 2001; Tamura et al., 2001; Walter et al., 2001), peroxisomal

Disease Models & Mechanisms 125

Disease Models & Mechanisms 5, 125-140 (2012) doi:10.1242/dmm.007708

1Institute for Anatomy and Cell Biology II, Division of Medical Cell Biology,University of Giessen, 35385 Giessen, Germany*Authors for correspondence ([email protected];[email protected])

Received 7 February 2011; Accepted 29 July 2011

© 2012. Published by The Company of Biologists LtdThis is an Open Access article distributed under the terms of the Creative Commons AttributionNon-Commercial Share Alike License (http://creativecommons.org/licenses/by-nc-sa/3.0), whichpermits unrestricted non-commercial use, distribution and reproduction in any medium providedthat the original work is properly cited and all further distributions of the work or adaptation aresubject to the same Creative Commons License terms.

SUMMARY

Impaired neuronal migration and cell death are commonly observed in patients with peroxisomal biogenesis disorders (PBDs), and in mouse modelsof this diseases. In Pex11-deficient mice, we observed that the deletion of a single allele of the Pex11 gene (Pex11+/– heterozygous mice) causedcell death in primary neuronal cultures prepared from the neocortex and cerebellum, although to a lesser extent as compared with the homozygous-null animals (Pex11–/– mice). In corresponding brain sections, cell death was rare, but differences between the genotypes were similar to thosefound in vitro. Because PEX11 has been implicated in peroxisomal proliferation, we searched for alterations in peroxisomal abundance in the brainof heterozygous and homozygous Pex11-null mice compared with wild-type animals. Deletion of one allele of the Pex11 gene slightly increasedthe abundance of peroxisomes, whereas the deletion of both alleles caused a 30% reduction in peroxisome number. The size of the peroxisomalcompartment did not correlate with neuronal death. Similar to cell death, neuronal development was delayed in Pex11+/– mice, and to a furtherextent in Pex11–/– mice, as measured by a reduced mRNA and protein level of synaptophysin and a reduced protein level of the mature isoform ofMAP2. Moreover, a gradual increase in oxidative stress was found in brain sections and primary neuronal cultures from wild-type to heterozygousto homozygous Pex11-deficient mice. SOD2 was upregulated in neurons from Pex11+/– mice, but not from Pex11–/– animals, whereas the levelof catalase remained unchanged in neurons from Pex11+/– mice and was reduced in those from Pex11–/– mice, suggesting a partial compensationof oxidative stress in the heterozygotes, but a failure thereof in the homozygous Pex11–/– brain. In conclusion, we report the alterations in the braincaused by the deletion of a single allele of the Pex11 gene. Our data might lead to the reconsideration of the clinical treatment of PBDs and thecommon way of using knockout mouse models for studying autosomal recessive diseases.

Deletion of a single allele of the Pex11 gene is sufficientto cause oxidative stress, delayed differentiation andneuronal death in mouse brainBarbara Ahlemeyer1,*, Magdalena Gottwald1 and Eveline Baumgart-Vogt1,*

RESEARCH ARTICLED

iseas

e M

odel

s & M

echa

nism

s

DM

M

Page 2: Deletion of a single allele of the Pex11 gene is ...of catalase remained unchanged in neurons from Pex11 +/– mice and was reduced in those from Pex11 –/– mice, suggesting a partial

function is reduced, but not absent. Furthermore, it has beensuggested that the full phenotypic range is even wider than wecurrently appreciate and that diagnosis can easily overlook patientswith milder presentations (Weller et al., 2003). Steinberg et al.developed a PEX gene screen for a systematic screening of thePEX1, PEX26, PEX6, PEX12, PEX10 and PEX2 genes and haveidentified 25 novel PEX gene mutations and 91 as-yet-unclassifiedPBDs of the Zellweger syndrome spectrum (ZSS) (Steinberg et al.,2004). They conclude that approximately 20% of patients with aPEX1 or PEX6 defect are not identified owing to uncommonmutations. In addition, of all the mutations identified by the PEXgene screen, 36% of the patients were heterozygous (Steinberg etal., 2004). In a heterozygous PEX12+/– patient, the authors wereunable to find a second mutation to explain the severe PBDphenotype (ZSS). However, such screening methods might differwith respect to efficacy and correctness, and a detailed analysis ofthe gene defect would be needed in this case. Another patient, whohad sensorineural deafness and retinitis pigmentosa, was firstmisdiagnosed and was only later – when his son was diagnosedwith PBD – found to have one defective PEX6 allele with twomissense mutations and a second splice site mutation on anotherPEX gene (Raas-Rothschild et al., 2002). We hypothesize thatdisruption of a single allele of a PEX gene might be sufficient tocause milder neurological symptoms. Using the embryonic day 19(E19) Pex11-deficient mouse, we searched for differences betweenhomozygous (Pex11–/–), heterozygous (Pex11+/–) and wild-type(Pex11+/+) animals. We especially focused on alterations in thebrain, because it is this organ and sensory organs (eye, ear) thatseem to be prone to damage in ZS patients; for example, defectsin the layer formation of the cerebral and cerebellar cortices,hypomyelination and neurodegeneration have been observed(Powers and Moser, 1998).

RESULTSDeletion of a single allele of the Pex11gene causes neuronaldeathImpaired neuronal migration and focal areas of enhanced neuronalapoptosis are the typical pathological alterations in the medialneocortex of Pex11–/– mice and other ZS mouse models (Baes etal., 1997; Baes et al., 2002; Faust and Hatten, 1997; Li et al., 2002a;Li et al., 2002b; Maxwell et al., 2003). Because this group of diseasesis inherited in an autosomal recessive manner, it was generallythought that the phenotype of heterozygous animals is identical tothe wild-type ones, so studies on ZS included only wild-type andhomozygous animals. When we prepared primary neuronalcultures from the neocortex and cerebellum of Pex11+/+, Pex11+/–

and Pex11–/– mice, we measured a higher basal level of TUNEL(terminal desoxynucleotidyl transferase-mediated dUTP nick endlabeling)-positive cells in cultures from heterozygous micecompared with those from wild-type mice – neuronal damage waseven more enhanced in those from the homozygous-null animals(Fig. 1A,C). Cell death in cortical cultures was further characterizedby immunofluorescence preparations to reveal caspase-3 activation(Fig. 1B,C) and by propidium iodide staining to detect membraneleakage (Fig. 1C). Nuclear staining using Hoechst 33342 wasperformed in parallel. Evaluation of different parameters in the samecells revealed that 85% of all neurons with an apoptotic nuclearmorphology were positively stained for TUNEL, but only a

proportion of these contained activated caspase-3 (38%) or wereleaky for propidium iodide (37%; Fig. 1C). The caspase inhibitorzVAD.fmk (100 M) partially protected neurons against death inall three genotypes (Fig. 1E). Thus, cell death in Pex11-deficientneurons showed typical features of apoptosis, a partial involvementof caspase-3 activation and membrane leakage (also known to occurin later stages of apoptosis). Differences in cell death between allthree Pex11 genotypes were highly reproducible, as shown for fourdifferent litters in Fig. 1D. In addition, we determined neuronaldeath in parallel in cultures from two different areas of theneocortex – the lateral and medial part – as well as from thecerebellum of the same animal (Fig. 1F), and at different time pointsin culture (Fig. 1G).

Next, we examined cell death in the brain of these mice to verifythe results that we obtained in primary neuronal cultures.Heterozygous animals are the same size and weight as wild-typemice and no differences are observed macroscopically (Li et al.,2002b). Careful analysis of the medial neocortex and thecerebellum revealed a higher number of TUNEL-positive neuronsin heterozygotes compared with wild-type animals – cell deathwas again more pronounced in homozygous animals (Fig. 2, Table1). Similar results were obtained when we compared activecaspase-3 immunoreactivity in the respective brain areas (Table1), suggesting apoptotic cell death of the neurons. We would liketo emphasize that cell death in brain tissue was rare comparedwith that observed in neuronal cultures (apoptotic neurons wereremoved in vivo), and thus differences between heterozygous andwild-type animals might have been overlooked in previous studies.Analysis of brain sections confirmed our previous observation ofneuronal death being present in animals with one deficientPex11 allele and to a further extent in those with defects in bothalleles.

Neuronal death does not correlate with peroxisome abundancePEX11 has been implicated in peroxisome proliferation, and areduced abundance of peroxisomes can be found in culturedmouse Pex11–/– fibroblasts, as shown in our previous studies (Liet al., 2002b). No information, however, was given for alterationsin heterozygous Pex11 mice. In brain sections from E19 fetuses,the abundance (number of peroxisomes/area) was reduced by 50%in the medial and lateral neocortex (Fig. 3A,D) and by 30% in thecerebellum of homozygous animals (Fig. 3B,F) compared with wild-type littermates. By contrast, no change and a slight increase of15% was noted in the cerebellum and medial neocortex ofheterozygous mice, respectively (Fig. 3A,D). Semi-quantificationof PEX14 immunoreactivity in heterozygous Pex11 mice revealedincreased (Fig. 3C, neocortex) or unchanged (Fig. 3E, cerebellum)levels, whereas, in neurons from homozygous Pex11 mice, thePEX14 level was only slightly reduced (Fig. 3C,E). Similarly to asfound in vivo, the abundance of peroxisomes in cultured neurons(95-98% of all cells in culture) and astrocytes (2-5% of all cells inculture) was reduced by half when prepared from Pex11–/– micecompared with those from their heterozygous and wild-typelittermates, as analyzed by PEX14 immunofluorescence (Fig. 4A,B)and western blot analysis (Fig. 4C). Thus, in cortical neurons, theabundance of peroxisomes was reduced in the same manner as theoverall protein level of PEX14, suggesting no change in the size ofthe peroxisomes as found for the cortical tissue. A comparison of

dmm.biologists.org126

Damage in Pex11+/– and Pex11–/– brainRESEARCH ARTICLED

iseas

e M

odel

s & M

echa

nism

s

DM

M

Page 3: Deletion of a single allele of the Pex11 gene is ...of catalase remained unchanged in neurons from Pex11 +/– mice and was reduced in those from Pex11 –/– mice, suggesting a partial

Disease Models & Mechanisms 127

Damage in Pex11+/– and Pex11–/– brain RESEARCH ARTICLE

Fig. 1. Deletion of one allele of the Pex11 gene caused an increase in neuronal death in primary cultures of the mouse neocortex and cerebellum.(A)Double fluorescence staining of cortical cultures from one litter, including all three Pex11 genotypes, using the TUNEL assay (red; Aa,Ac,Ae) and Hoechst33342 (blue; Ab,Ad,Af). Positive control cultures were treated with DNase (Ag,Ah). (B)Double fluorescence staining of neuronal cultures from Pex11+/+ andPex11–/– mice using an antibody against the active form of caspase-3 (green) and the neuronal marker MAP2 (red) to characterize neuronal death.(C)Quantitative characterization of neuronal death using Hoechst 33342 combined with either propidium iodide (PI) or active caspase-3, or TUNEL staining.(D)Quantification of neuronal death using Hoechst 33342 in cortical cultures of four different litters showing the reproducibility of the results. (E)The broad-spectrum caspase-inhibitor z-VAD (100M) partially reduced neuronal death independent of the genotype. (F,G)Increased neuronal death was found in culturesderived from the lateral and medial neocortex and cerebellum (F) and as well as at different time points during cultivation (G) of heterozygous Pex11mice and –to a further extent – in those from homozygous mice. (C-G)Neuronal death was determined according to the nuclear morphology in three areas in each of fourdifferent cultures (800-1200 cells) for each genotype derived from the same (C,E-G) or from different (D) litters. Mean values ± s.d. are shown; (C-F) differencesbetween the indicated groups: ***P<0.001; (G) differences in comparison to cultures from wild-type Pex11 mice: ***P<0.001; difference between cultures fromheterozygous and homozygous Pex11 mice: #P<0.05.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 4: Deletion of a single allele of the Pex11 gene is ...of catalase remained unchanged in neurons from Pex11 +/– mice and was reduced in those from Pex11 –/– mice, suggesting a partial

the cell volume – both in primary neuronal cultures and in brainsections of the neocortex – revealed no difference between the threePex11 genotypes. However, a detailed analysis of changes in thesize and form of peroxisomes in Pex11-deficient neurons requireselectron microscopy and will be the subject of future studies. Forthe investigation of adaptive responses to the impaired ability ofperoxisome division, we analyzed the expression of other PEX11protein family members as well as of FIS-1 and DLP1 genes(Kobayashi et al., 2007). Interestingly, an increase in Pex11mRNAwas noted in cultures from Pex11–/–, but not in those fromPex11+/–, mice (Fig. 4D, Table 2). We suggest that this increasecould reflect an adaptive response to the peroxisome division defect.However, in comparison to the high expression level of Pex11mRNA in the brain, the expression level of Pex11 is 64-fold lowerand the amount of protein is below detectable levels (Li et al.,2002a). Therefore, upregulation of Pex11 might not adequatelysubstitute for the Pex11 defect, but prevent the total absence ofperoxisomes. In addition, we cannot exclude that the presence ofperoxisomes in the Pex11 knockout mouse is due to a residualfunction of the exon-4-encoded PEX11 protein. The mRNAlevels of Fis-1 and Dlp1, whose proteins mediate peroxisomalfission, were not affected (Fig. 4E).

Deletion of a single allele of the Pex11 gene causes a delay inneuronal differentiationThe neuronal marker protein microtubule-associated protein 2(MAP2) is only expressed in neurons that have reached their final

destination, and its level is progressively increased during the firstpostnatal week (Ahlemeyer et al., 2007; Ferreira et al., 1987). Thus,MAP2 was undetectable in the E19 brain, but we have analyzedthe expression of synaptophysin, a marker for early synaptogenesisthat is present at a sufficiently high level at birth (Li et al., 2010;Shimohama et al., 1998). In comparison to the neocortex andcerebellum of wild-type animals, synaptophysin immunoreactivitywas reduced in heterozygous Pex11 animals and this decrease waseven more pronounced in the homozygous ones (Fig. 5A,B). Thedelay in neural development can be reproduced in cortical neuronsfrom Pex11+/– and Pex11–/– mice, demonstrated by the reduced

dmm.biologists.org128

Damage in Pex11+/– and Pex11–/– brainRESEARCH ARTICLE

Fig. 2. Deletion of one allele of the Pex11 gene caused an increase in the abundance of TUNEL-positive cells in the neocortex and the cerebellum. TheTUNEL assay (to detect apoptotic cells; Aa,Ac,Ae,Ba,Bc,Be) and Hoechst stain (to visualize the nuclei of all cells; Ab,Ad,Af,Bb,Bd,Bf) were applied to brain sectionsof the migration zone in the neocortex (‘Cx’, A) and of the cerebellum (‘Cb’, B) of all three Pex11 genotypes. TUNEL-positive cells and the corresponding nucleishowing typical features of apoptosis such as condensation or fragmentation are encircled. The inset (in Af) shows a higher magnification of the encircled area(arrowhead); arrows indicate pyknotic nuclei.

Table 1. The abundance of TUNEL- and active-caspase-3-positive

cells, and the signal intensity of synaptophysin, 8-OHdG, catalase

and SOD2 immunoreactivity in the neocortex and cerebellum from

wild-type, heterozygous and homozygous Pex11 mice

Genotype

+/+ +/– –/– +/+ +/– –/–

Neocortex Cerebellum

TUNEL + +++ + ++ +++

Active caspase-3 + +++ + +++

Synaptophysin +++ ++ + +++ +

8-OHdG + ++ ++ + + ++

Catalase ++ ++ + ++ ++ ++

SOD2 + +++ + +++

+++, very high; ++, high; +, moderate; , weak.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 5: Deletion of a single allele of the Pex11 gene is ...of catalase remained unchanged in neurons from Pex11 +/– mice and was reduced in those from Pex11 –/– mice, suggesting a partial

formation and branching of the neural network after 7 days inculture (Fig. 5C) and by reduced mRNA levels of synaptophysin(Fig. 5Da, Table 2) as well as by the reduced protein levels of MAP2and synaptophysin (Fig. 5Db). Interestingly, the higher molecularweight form of MAP2 (MAP2a/b at 280 kDa), which normallyincreases in level in the postnatal period at day 10 (Przyborski andCambray-Deakin, 1995), was not detectable in cultures fromPex11+/– or Pex11–/– mice, but was clearly present in the moremature wild-type cultures (Fig. 5Db). The level of the low molecularweight form of MAP2 (MAP2c), known to be expressed at earlydevelopmental stages but to decrease postnatally after 2 weeks(Crandall and Fischer, 1989), was not different between thegenotypes. Moreover, MAP2a/b is largely located in dendrites andaxons, whereas MAP2c is widely located in every neuronalcompartment (Tucker et al., 1988).

Increased astrogliosis has been shown in adult CNP-Cre/Pex5loxP- and nestin-Cre/PEX13loxP-knockout mice(Kassmann et al., 2007; Müller et al., 2011). However, at early stages

of neuronal development such as E19, glial fibrillary acidic protein(GFAP)-immunopositive reactions were only found in themembrana glia limitans and the subventricular zone (Ahlemeyeret al., 2007), and primary neuronal cultures contained only veryfew clustered astrocytes (less than 2%) with the same GFAP mRNAand protein level (Fig. 5E) independent of the Pex11 genotype.

Deletion of a single allele of the Pex11 gene causes oxidativestressA deficiency in peroxisomal function has been suggested to causeoxidative stress (Baumgart et al., 2001; Bonekamp et al., 2009). InE19 sections of the mouse neocortex and cerebellum, an elevated8-hydroxy-2�-deoxyguanosine (8-OHdG) immunoreactivity wasfound in heterozygotes with a further increase in homozygousanimals (Fig. 6A,B, Table 1). Consistent findings were obtained byDot blot analyses of 8-OHdG immunoreactivity in nuclear extractsfrom the neocortex of Pex11+/– mice (Fig. 6C). With respect toantioxidant enzymes, we found a selective increase in manganese

Disease Models & Mechanisms 129

Damage in Pex11+/– and Pex11–/– brain RESEARCH ARTICLE

Fig. 3. The abundance of PEX14-positive peroxisomes differs between the brains of wild-type, heterozygous and homozygous Pex11mice. Analysis ofimmunofluorescence images of PEX14 revealed a reduced abundance (number of PEX14-positive peroxisomes/100m2) and immunoreactivity for this proteinin the neocortex (‘Cx’; Aa,Ac,C,D) and cerebellum (‘Cb’; Ba,Bc,E,F) of homozygous mice, whereas a higher PEX14 immunoreactivity and only a slight increase inabundance was found in the medial neocortex of heterozygous Pex11 mice (compare Aa with Ab; C,D). The abundance of PEX14-positive peroxisomes in thecerebellum was not different between heterozygous and homozygous Pex11 mice (Bb,Bc,F). Mean values ± s.d. are shown; differences either in comparison tothe respective brain area of the wild-type Pex11 mice or between the indicated groups: *P<0.05, **P<0.01, ***P<0.001.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 6: Deletion of a single allele of the Pex11 gene is ...of catalase remained unchanged in neurons from Pex11 +/– mice and was reduced in those from Pex11 –/– mice, suggesting a partial

superoxide dismutase (superoxide dismutase 2; SOD2)immunoreactivity in the neocortex and cerebellum of heterozygousPex11 mice (Fig. 6D, Table 1), and a decrease in catalaseimmunoreactivity in the neocortex of homozygous Pex11 mice(Fig. 6E, Table 1). Primary neuronal cultures allow a directmeasurement of the cellular reactive oxygen species (ROS) levels.

Compared with neurons from wild-type mice, ROS levels in thosefrom heterozygous and homozygous Pex11 animals were indeedincreased at day 2, 4, 7, 10 and 14 in culture, in the latter ones toa higher extent (Fig. 7A,C,D). Data from four different litters atday 7 in culture is shown in Fig. 7E. We suggest oxidative stress asthe mediator of cell death because tocopherol, when added 4 hoursafter seeding, not only reduced the increased ROS levels (Fig. 7C),but also reduced cell death (Fig. 7B) in cultured neurons from bothheterozygous and homozygous Pex11 animals to the level seen inthe wild-type controls. Similar to our in vivo findings, the mRNAand protein levels of SOD2 were selectively higher in cultures fromheterozygotes and those of catalase were lower in cultures fromhomozygous mice (Fig. 7F,G,H, Table 2) when compared with thosefrom wild-type littermates. Further studies are needed to find outwhy neurons from Pex11 knockout mice are compromised inupregulating antioxidative survival pathways.

DISCUSSIONTwo aspects regarding PBDs can be drawn from our experimentsusing brain tissue and cultured neurons from E19 Pex11-deficientmice: (1) this study provides direct evidence for oxidative stress asa result of Pex11 deficiency together with a defective regulationof the antioxidant response in the brain of Pex11–/– comparedwith Pex11+/– mice; and (2) the observation of a neural phenotypein Pex11 heterozygous mice, although with less severity than inthe homozygous animals.

Oxidative stress in Pex11+/– and Pex11–/– brains, and a defectiveregulation of the antioxidative response in Pex11–/– brainTo date, only limited data of the mechanism or role of oxidativestress in the neuropathology observed in ZS is available, althoughit is an observed phenomenon in other disorders of the neonatalhuman brain (Robertson et al., 2009; Taylor et al., 1999). Previousmorphological studies in general Pex5 knockout mice revealedultra-structural alterations of mitochondria, indicative of oxidativestress, together with reduced activities of mitochondrial respiratorychain complexes and an increase in SOD2 in cardiomyocytes andhepatocytes, but the brain was not examined (Baumgart et al.,2001). Similar mitochondrial alterations have been observed in theliver of mice lacking Pex13 (Maxwell et al., 2003) and Pex2 (Keaneet al., 2007) as well as in patients with Hsd17b4 gene mutations(Ferdinandusse et al., 2003). However, hepatocyte-specific Pex5

dmm.biologists.org130

Damage in Pex11+/– and Pex11–/– brainRESEARCH ARTICLE

Fig. 4. Deletion of both, but not of one, alleles of the Pex11gene reducedthe abundance of PEX14-positive peroxisomes in cultured corticalneurons. (A)Double immunofluorescence of PEX14 and the neuronal markerMAP2 (Aa,Ac,Ae) or the astrocyte marker GFAP (Ab,Ad,Af) in cultures fromPex11+/+ (Aa,Ab), Pex11+/– (Ac,Ad) and Pex11–/– (Ae,Af) mice.(B,C)Quantitative analysis of the peroxisomal abundance was performed bycounting the number of peroxisomes/100m2 in individual neurons in fourareas in each of six different cultures (150 cells) of each genotype (B) and bywestern blotting of PEX14 in homogenates of neuronal cultures from thedifferent Pex11 genotypes (C). (D,E)Quantitative (D) and semi-quantitative (E)RT-PCR analysis of Pex14 and of genes known to be involved in peroxisomalproliferation. In B and E, mean values ± s.d. are shown; differences incomparison to brain cells derived from wild-type Pex11 mice or between theindicated groups: *P<0.05, **P<0.01, ***P<0.001.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 7: Deletion of a single allele of the Pex11 gene is ...of catalase remained unchanged in neurons from Pex11 +/– mice and was reduced in those from Pex11 –/– mice, suggesting a partial

knockout mice do not show oxidative damage of proteins or lipids(Dirkx et al., 2005). By contrast, but similar to our findings, ABCD3knockdown in primary fibroblast cultures was found to increasethe production of superoxide in combination with an upregulationof SOD2 and a decrease in catalase levels (Di Benedetto et al., 2009).Oxidative stress has also been discussed as a pathogenic factor inX-linked adrenoleukodystrophy (X-ALD), because patients exhibitincreased levels of very long chain fatty acids and developinflammatory demyelination and neurodegeneration similar to thatfound in patients with PBDs (Berger and Gärtner, 2006). In X-ALD(ABCD1)-deficient adrenal cortical cells, mitochondria arestructurally abnormal (McGuinness et al., 2003) and human, butnot mouse, brain shows evidence of oxidative stress such as anincrease in SOD2 and oxidative damage (Powers et al., 2005). Ourresults on primary neuronal cultures and brain tissue from Pex11-deficient mice provide direct evidence for oxidative stress and DNAdamage, and suggest a defective mitochondrial antioxidativeresponse in PBDs.

Deletion of a single allele of the Pex11 gene is sufficient to inducea neural phenotypeMutations in PEX genes in human PBDs and their respectiveknockout mouse models are inherited in an autosomal recessivemanner. However, we here describe a phenotype in the brain ofanimals carrying just one defective Pex11 allele. The severity of

the symptoms is less in Pex11 heterozygous than in homozygousmice. This is of relevance: (1) for mouse models and in individualswith PBDs (although the loss of one Pex11 allele does notnecessarily follow the same pattern as the other PBD models); (2)for mouse models of recessive diseases in general, becausesometimes heterozygous animals were used as controls owing tothe limited availability of wild-type animals in a litter and werecompared with the knockout ones; and (3) because our data mightcontribute to the discussion about heterozygous advantages anddisadvantages.

As mentioned in the Introduction, the genotype-phenotyperelationship and especially the severity of symptoms in humanpatients suffering from PBDs is very complex, whereas all PBDmouse models exhibit severe symptoms with varying biochemicalparameters. The discrepancy between mice and humans mightbe because, in mice, one [Pex2 (Faust and Hatten, 1997) and Pex7(Brites et al., 2003)] to three [Pex5 (Baes et al., 1997), Pex11 (Liet al., 2002b) and Pex13 (Maxwell et al., 2003)] exons of therespective genes were deleted, whereas humans with PBDs oftenexhibit only a single point mutation in a PEX gene, with severeor less severe consequences for the function of the correspondingprotein. In addition, we know from our mouse models that theseverity of the symptoms depends on the genetic background ofthe animals. For example, Pex mutations generated in C57Bl/6Jmice lead to a severe outcome and the mutant mice die directly

Disease Models & Mechanisms 131

Damage in Pex11+/– and Pex11–/– brain RESEARCH ARTICLE

Table 2. The sequence and efficiency coefficient of forward and reverse primers and a comparative analysis of the mRNA expression of

defined genes after quantitative RT-PCR

Target

gene Accession number Sequence (3 to 5 ) E

Fold change

het/wt PFold change

hz/wt PF ACTGGCCGTAAATGGTTCAGAPex11 NM_011068.1

R CGGTTGAGGTTGGCTAATGTC

1.90 1.28±0.21 n.s. 0.95±0.19 n.s.

F CGCCTATTGATGGAACAAGAGACTPex11 NM_0011069.3

R TCCAGGTCCCACAGTTTCTACTC

1.98 0.47±0.11 ** n.d. ***

F GACTCTGCTTGGTGGTGGAPex11 NM_026951.2

R GCTCCGCAGCTTCTGTCTAA

1.83 0.95±0.15 n.s. 1.51±0.31 *

F TGATTGCCACAGCAGTGAAGPex14 NM_019781.2

R GCCAGGTCAATCTCTTCGTC

1.94 0.94±0.18 n.s. 1.02±0.14 n.s.

F GACCTGGGCAATGTGACTGCTGSod1 NM_011434.1

R GCACCAGTGTACGGCCAATGATG

1.98 0.86±0.17 n.s. 0.83±0.14 n.s.

F ACTGAAGTTCAATGGTGGGGSod2 NM_013671.3

R GCTTGATAGCCTCCAGCAAC

1.94 1.77±0.36 * 0.86±0.12 n.s.

F GGAGGCGGGAACCCAATAGCatalase

(Cat)NM_009804.2

R GTGTGCCATCTCGTCAGTGAA

1.91 0.93±0.21 n.s. 0.66±0.06 *

F GCCAGCCTCAGAACAAACAGMtap2 NM_001039934

R AAGGTCTTGGGAGGGAAGAAC

1.98 1.16±0.22 n.s. 0.96±0.14 n.s.

F AAGGAACTGAGGGACCCTGTSyp NM_009305.2

R AGCCTGTCTCCTTGAACACG

1.93 1.05±0.11 n.s. 0.75±0.06 *

F TGGCAAAGTGGAGATTGTTGCCGapdh NM_008084

R AAGATGGTGATGGGCTTCCCG

1.98 ufn – ufn –

The total RNA of cortical neurons, derived from the neocortex of E19 Pex11 homozygous and heterozygous animals and grown for 7 days in culture, was isolated, reversely

transcribed into cDNA and subjected to qPCR (SYBR Green method). The assay was run in triplicates for one sample with a total number of three samples (three litters) for each

Pex11 genotype. qRT-PCR analysis was performed by the ct method using the Pfaffl equation. The ratio values (fold change) of the mRNA of a defined gene between

heterozygous (het) and wild type (wt) or between homozygous (hz) and wild type normalized to GAPDH [used for normalization (ufn)] are shown. Significance of the differences

were evaluated by ANOVA-1 and post-hoc Scheffé test with *P<0.05; **P<0.01; ***P<0.001; n.s., not significant; E, efficiency coefficient; F, forward; R, reverse.

α

β

γ

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 8: Deletion of a single allele of the Pex11 gene is ...of catalase remained unchanged in neurons from Pex11 +/– mice and was reduced in those from Pex11 –/– mice, suggesting a partial

after birth (Baes et al., 1997; Brites et al., 2003; Faust and Hatten,1997; Li et al., 2002a; Li et al., 2002b; Maxwell et al., 2003), butthe survival could be prolonged up to postnatal day 18 (P18) whenthe mutation was placed on a Swiss Webster � 128 SvEv geneticbackground (Faust, 2003). In X-ALD, a peroxisomal single-enzyme defect, the opposite was observed: all three Abcd1knockout mouse models (Forss-Petter et al., 1997; Kobayashi etal., 1997; Lu et al., 1997) did not develop a detectable phenotypeup to 6 months, in contrast to humans with X-ALD, who showsevere symptoms in the adrenal gland and brain. However, olderX-ALD mice (starting from 15 months of age) exhibited slowernerve conduction as well as myelin and axonal abnormalities inthe spinal cord and the sciatic nerve (Dumser et al., 2007; Pujolet al., 2002). Interestingly, X-ALD is inherited in an X-linkedmanner and thus the severity of the symptoms is sex dependent.Males who inherit the mutation will be variably affected with anunpredictable phenotypic expression and prognosis. Femaleswho inherit the mutation are carriers, but also 50% are usually

mildly affected (Berger and Gärtner, 2006). Thus, even when bothalleles of genes are mutated, the severity of symptoms can varywidely depending on the type of mutation, the individual’s geneticbackground, sex and the species.

Interestingly, we previously observed a similar phenomenon ofa heterozygous phenotype in cerebellar cultures prepared fromPex13 knockout mice (Müller et al., 2011). However, thisobservation was not analyzed in more detail. It is likely that thebrain is especially sensitive to damaging conditions, and one mayask whether heterozygous Pex11 mice might also show defects inother organ systems. In general, heterozygous Pex11 animals areindistinguishable from and are the same size and weight as theirwild-type littermates, and are fertile (Li et al., 2002b). We did notsee obvious histopathological alterations or defects in the lung(development of the alveolar compartment) and bone (desmal andenchondral ossification), but the mRNA and protein levels of anumber of genes were different in heterozygous compared withwild-type animals (e.g. peroxins, peroxisomal membrane and

dmm.biologists.org132

Damage in Pex11+/– and Pex11–/– brainRESEARCH ARTICLE

Fig. 5. Deletion of one allele of the Pex11gene caused impaired neuronal development and network formation without changes in the amount andactivation state of astrocytes in primary cortical cultures. (A,B)Double immunofluorescence staining for PEX14 and synaptophysin (Synapto) in theneocortex (‘Cx’; A) and cerebellum (‘Cb’; B) of wild-type, heterozygous and homozygous Pex11mice. Note the decrease in synaptophysin immunoreactivitycaused by the deletion of the Pex11 gene. (C)Phase-contrast images showing reduced network formation of cortical cultures from Pex11–/– (Cb) comparedwith those from Pex11+/+ (Ca) mice. (D)Reduction of the presynaptic vesicle protein synaptophysin at the mRNA (Da) and protein (Db) level, and of the dendriticneuronal marker MAP2 at the protein level (mature isoform; Db). (E)GFAP mRNA (Ea) and protein (Eb) levels of cortical cultures were not different when preparedfrom wild-type, heterozygous or homozygous Pex11 mice.D

iseas

e M

odel

s & M

echa

nism

s

DM

M

Page 9: Deletion of a single allele of the Pex11 gene is ...of catalase remained unchanged in neurons from Pex11 +/– mice and was reduced in those from Pex11 –/– mice, suggesting a partial

matrix proteins, antioxidant enzymes, and transcriptions factors;data not shown). Thus, even though there was no obviousphenotype, heterozygotes are different and not comparable to wild-type animals, at least on the molecular level.

Next, we investigated whether there are other examples in theliterature describing alterations in the brain of heterozygousanimals or in patients with autosomal recessive diseases. In theearly-onset autosomal recessive form of Parkinson’s disease (PD),PARK2 is mutated. Sun et al. showed that individuals with

homozygous, but also heterozygous, mutated PARK2 showedsymptoms that differed in the onset as well as in severity comparedwith healthy PD individuals with an intact PARK2 gene (Sun etal., 2006). Similarly, clinical studies on PINK1 gene mutations,which predispose individuals to PD, showed that, although noneof the young heterozygous affected family members were awareof symptoms, they already exhibited unilaterally reduced orabsent arm movement or rigidity (Hedrich et al., 2006; vanNuenen et al., 2009). For amyotrophic lateral sclerosis, the D90A

Disease Models & Mechanisms 133

Damage in Pex11+/– and Pex11–/– brain RESEARCH ARTICLE

Fig. 6. Experimental evidence for oxidative stress in the brain of heterozygous Pex11mice. (A-C)Detection of 8-OHdG was performed byimmunofluorescence staining of the neocortex (‘Cx’; A) and cerebellum (‘Cb’; B) as well as by a Dot blot analysis using nuclear extracts of the neocortex (C) fromwild-type, heterozygous and homozygous Pex11 mice. In C, immunoblotting of histone H3 and Ponceau S staining was performed to show the relative amountof the dotted material. (D,E)Immunofluorescence staining for SOD2 (D) and catalase (E) of the neocortex of wild-type, heterozygous and homozygous Pex11mice. Please note an increase in 8-OHdG and SOD2 in animals with deletion of a single Pex11 allele.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 10: Deletion of a single allele of the Pex11 gene is ...of catalase remained unchanged in neurons from Pex11 +/– mice and was reduced in those from Pex11 –/– mice, suggesting a partial

dmm.biologists.org134

Damage in Pex11+/– and Pex11–/– brainRESEARCH ARTICLE

Fig. 7. Deletion of one allele of the Pex11 gene caused oxidative stress and changes in the level of antioxidant enzymes in primary neuronal culturesfrom the neocortex. (A-D)Representative micrographs of ethidine fluorescence in neurons from Pex11+/+, Pex11+/– and Pex11–/– mice in the absence(Aa,Ab,Ac) or presence (Ad) of 10M -tocopherol (toco). (B-E)Quantification of the mean fluorescence intensity (MFI) of ethidine in single cells in a corticalculture from all three genotypes from four distinct litters (E), at different time points during the cultivation (D) as well as in one litter in the absence or presenceof tocopherol (C) together with evaluation of the cell death (B) were shown. For C-E, 200-350 cells were analyzed in ten areas in each of three cultures for eachgenotype derived from the same litter. (F)Representative micrographs showing a selective increase in SOD2 immunoreactivity in neurons from Pex11+/– mice(Fb) compared with those from Pex11+/+ (Fa) and Pex11–/– (Fc) mice. (G,H)RT-PCR (G) and western blot analysis (H) of different antioxidant enzymes, showingincreased mRNA (G) and protein (Ha) levels of SOD2 in neurons from Pex11+/– mice and a decreased protein level of catalase in neurons from Pex11–/– mice. ForHa, neurons were treated with vehicle (vh) or tocopherol (toco). Semi-quantification of catalase and SOD2 protein levels normalized to -tubulin in vehicle-treated neuronal cultures from Pex11+/+, Pex11+/– and Pex11–/– mice of four different litters is given in Hb. (B-E,Hb) Mean values ± s.d. are shown. Differenceseither between the indicated groups or in comparison to cultures from wild-type Pex11mice: *P<0.05, **P<0.01, ***P<0.001 (B,C,E,Hb); difference betweencultures from heterozygous and homozygous Pex11 mice: ### P<0.001 (D).

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 11: Deletion of a single allele of the Pex11 gene is ...of catalase remained unchanged in neurons from Pex11 +/– mice and was reduced in those from Pex11 –/– mice, suggesting a partial

mutation of the copper-zinc superoxide dismutase (superoxidedismutase 1; SOD1) gene is inherited in an autosomal recessivemanner, but slight motor symptoms have also been found inheterozygous relatives (Mezei et al., 1999). In ataxia telangiectasia,a typical autosomal recessive disease, heterozygous individualshave an increased radiosensitivity and risk of cancer as well aschanges in the baseline expression of many genes compared withnormal control individuals (Watts et al., 2002). Thus, one mightspeculate that the less severe symptoms in heterozygotes are oftenoverlooked and that these patients were incorrectly consideredas asymptomatic. Hedrich et al. hypothesized that re-evaluationof the role of single gene mutations might have a major implicationfor gene counseling. However, also the opposite might be possible:the existence of the so-called ‘heterozygote advantage’. In thistheory, the heterozygote phenotype was supposed to have a higherfitness because this selection should be one of the mechanismsto maintain polymorphism in evolution (Gillespie, 2004; Otto andYong, 2002). As examples for the heterozygote advantage, one canmention sickle cell anemia, in which the carriers are resistant tomalaria infection (Williams, 2006), as well as hematochromatosis,in which the low iron content renders macrophages resistant topathogens (Weinberg, 2008), or cystic fibrosis in which modelmice are resistant to cholera (Gabriel et al., 1994). However, forthe latter disease, the results could not be reproduced in humans.Another phenomenon is so-called over-dominance, in which thephenotype of the heterozygote lies outside the range ofhomozygous individuals (Gillespie, 2004).

In summary, our results indicate that, when analyzing knockoutmice as models for autosomal recessive diseases, all three genotypes(at best from one litter) should be compared with each other inorder to fully understand the pathophysiology of the gene defect.Heterozygous animals – even when they seem to be asymptomatic– might differ in the expression of many genes and the investigationthereof could help to understand (and to mimic) endogenousadaptation to gene defects.

METHODSChemicals and reagentsNeurobasal medium, B27 supplement, glutamine, penicillin-streptomycin, (di)hydroethidine, DNase I (Cat. 180668-015),SuperScript II First-Strand Synthesis System (Cat. 1806-022),RNaseOut (Cat. 10777-019) and TOTO-3 iodide were purchasedfrom Invitrogen (Karlsruhe, Germany). Poly-L-lysine, papain,trypsin inhibitor, dimethylsulfoxide (DMSO), Hoechst 33342,Triton X-100, Tween 20, bovine serum albumin (BSA), propidiumiodide, (±)-tocopherol, Ponceau S and z-Val-Ala-Asp fluoromethylketone (z-VAD.fmk) were obtained from Sigma-Aldrich(Deisenhofen, Germany). The ApopTag Red In Situ DetectionApoptosis kit (S7165) was purchased from Chemicon (Schwalbach,Germany). Details on all primary antibodies used in this study,containing immunogen, host and source are given in Table 3. Thefollowing secondary antibodies were used for indirectimmunofluorescence: goat anti-rabbit IgG Alexa Fluor 488 (1:300;Invitrogen), goat anti-chicken Alexa Fluor 633 (1:300; Invitrogen)and goat anti-mouse IgG Texas Red (1:100; Vector Laboratories,Burlingham, CA), and for western blot analysis: goat anti-rabbitand goat anti-mouse IgG coupled to alkaline phosphatase (1:20,000; Sigma-Aldrich).

AnimalsAnimals had free access to food and water and were kept understandardized environmental conditions (12-hour light-dark cycle,23±1°C and 55±1% relative humidity). Experiments with laboratorymice were approved by the Government Commission of AnimalCare, Germany. For all experiments, heterozygous Pex11C57BL/6Jmice (genetic background >F8 generation) were mated overnight.In some series of experiments all E19 fetuses from one litter wereperfused with paraformaldehyde to obtain tissue sections, or themedial neocortices were homogenized to obtain nuclear extracts. Inother series of experiments, all E19 fetuses were used for thepreparation of primary neocortical and cerebellar cultures.

Genotyping of mouse tail DNAMouse tails were digested overnight and 0.2 g of the isolated DNAwas used for PCR analysis the following day as previously described(Li et al., 2002b). The following primers were used for the genotyping:primer 8: 5�-GTCTAGGACAGGTTCTGTGTTC-3�, primer 9: 5�-GTTTCCCCATCTTTCCCTTGAG-3� and neo primer 5�-ATATTGCTAAGAGCTTGGCGGCGGC-3�. The wild-type Pex11allele was amplified using primers 8 and 9 (590 bp), whereas therecombinant knockout allele was amplified by primer 8 and neoprimer (980 bp). The PCR reaction was performed using TaqDNApolymerase (Eppendorf, Hamburg, Germany) in a Bio-Rad iCycler(Bio-Rad, München, Germany) using the following parameters:denaturation at 95°C, 5 minutes; followed by 35 cycles of denaturationat 95°C, 1 minute, annealing at 58°C for 1 minute, extension at 72°Cfor 1 minute; and a final extension at 72°C for 10 minutes.

Perfusion fixation of mice and processing of brain tissuePregnant dames of heterozygous Pex11 C57BL/6J mouse matingsincluding all E19 fetuses were anesthetized with isoflurane andsubsequent intraperitoneal injection of a mixture ofketamine/xylazine. After a short rinse with 0.9% NaCl to removeblood cells, perfusion fixation was carried out for 5 minutes viathe left ventricle of the heart using freshly prepared 4%depolymerized paraformaldehyde in Ca2+- and Mg2+-freephosphate-buffered saline (PBS). Whole brains were carefullydissected out of the skull and additionally immersion fixedovernight in the same fixative. The following morning, the completebrains were transferred to an automated vacuum infiltration tissueprocessor (Leica TP 1020) and processed for paraffin embedding.The following dehydration and infiltration steps were used: (1) 70%ethanol, 80% ethanol, 90% ethanol, 4� 100% ethanol, 4� xylene,each step 1.5 hours, and (2) 2� paraffin (Paraplast plus containing0.8% DMSO), each step 2 hours. After vertical embedding, 2-mparaffin sections of total brain (coronal orientation) were cut on aLeica RM 2135 microtome and mounted on Superfrost Plus slides.We examined brain sections containing the medial neocortex(comparable to Bregma 0.98 mm of the adult mouse) andcerebellum (comparable to Bregma –5.8 mm of the adult mouse)of each genotype from the same litter.

Indirect immunofluorescence and TUNEL stain on paraffin-embedded brain sectionsFor optimal retrieval of peroxisomal antigens and accessibility ofepitopes, deparaffinized and rehydrated brain sections weresubjected to digestion with 0.01% trypsin for 10 minutes at 37°C

Disease Models & Mechanisms 135

Damage in Pex11+/– and Pex11–/– brain RESEARCH ARTICLED

iseas

e M

odel

s & M

echa

nism

s

DM

M

Page 12: Deletion of a single allele of the Pex11 gene is ...of catalase remained unchanged in neurons from Pex11 +/– mice and was reduced in those from Pex11 –/– mice, suggesting a partial

followed by microwaving in 10 mM citrate buffer at pH 6.0 for 3�5minutes at 800 W in a conventional household microwave oven(Baumgart et al., 2003; Grabenbauer et al., 2001). Nonspecificbinding sites were blocked with 4% BSA and 0.05% Tween 20 inPBS for 2 hours at room temperature and sections were incubatedwith primary antibodies (for details see Table 3) overnight at 4°C.On the following morning, the sections were rinsed carefully withPBS and thereafter incubated with the secondary antibodies for 2hours at room temperature (for details see Table 3). Nuclei werelabeled with TOTO-3 iodide (20 M). Negative controls wereperformed in parallel by omitting the primary antibody. Thenumber of PEX14-positive peroxisomes/100 m2 was determinedin five areas in each of the three different brains with wild-type,Pex11+/– and Pex11–/– genotypes. We counted the number ofperoxisomes within the cytoplasm excluding the nuclear region invimentin-positive cells (at embryonic stage E19, only vimentin, butnot MAP2, is expressed in neurons) (Ahlemeyer et al., 2007). Therespective cytoplasmic area was quantified using the Leica ConfocalSoftware program. Peroxisomes ranging from 0.03-0.2 m2 weredetectable at highest resolution. Values are expressed as the numberof peroxisomes/100 m2. In addition, semi-quantification of thePEX14 signal intensities in more than 25 areas with comparablecell densities of the different brains of each genotype wasperformed. Quantification of the signal intensities and area wasdone using the Leica Confocal Software program.

TUNEL stain was performed according to the manufacturer’sinstruction. Briefly, in de-paraffinized, rehydrated and trypsin-digested brain sections, the double-stranded and single-strandedDNA was labeled with digoxigenin-conjugated desoxynucleotidesat their 3�OH ends by terminal deoxynucleotidyl transferasereaction. Thereafter, sections were incubated with rhodamine-conjugated anti-digoxigenin antibody and Hoechst 33342(counterstaining) for direct analysis under the fluorescentmicroscope. DNase-I-treated brain sections were used as positivecontrols; negative controls were performed omitting the terminaldeoxynucleotidyl transferase reaction. The same protocol wasapplied to primary neuronal cultures.

Preparation of primary cultures of the medial neocortex andcerebellum of E19 miceThe fetuses of a heterozygous Pex11 C57BL/6J mating wereremoved by Cesarean section from the uterus of the pregnant dameat E19. Primary cultures were prepared from the medial neocorticesor the cerebellum of all fetuses from the same litter as describedpreviously (Ahlemeyer and Baumgart-Vogt, 2005). We dissectedand processed the cortices or cerebellum of individual miceseparately and in parallel, because immediate preparation prior toPCR genotyping was necessary for obtaining optimal neuronalcultures. Cortical and cerebellar neurons were seeded at a densityof 3�105 cells and 2�106 cells onto 35-mm poly-L-lysine-coatedPetri dishes, respectively. In one series of experiments, neuronaldeath and ROS level were determined at different time pointsduring culture. All other experiments were performed on day 7 inculture. At that time, primary neocortical and cerebellar culturescontained approximately 95-98% and 90% neurons, respectively.The low amount of astrocytes is mainly due to neuronal growth-promoting culture conditions such as poly-L-lysine coating and theuse of neurobasal medium as well as to the fact that cultures wereprepared from E19 fetuses. Drug treatment and some techniqueswere applied solely to primary neuronal cultures either becausesome of these techniques need living cells (ROS detection,propidium iodide stain) or because of the advantage to detectchanges in neurons only [reverse transcriptase (RT)-PCR, westernblot analysis], whereas tissue homogenates contain all other celltypes, including blood vessels, meninges and matrix proteins.

Indirect immunofluorescence on primary neuronal culturesCells on coverslips were rinsed with PBS and fixed with 4%paraformaldehyde in PBS for 20 minutes at room temperature. Afterfixation, cells were washed three times with PBS. Subsequently, theywere incubated for 10 minutes in PBS containing 1% glycine and0.3% Triton X-100 for permeabilization, and for an additional periodof 10 minutes in PBS containing 1% glycine. After washing withPBS, cells were incubated for 30 minutes in PBS containing 1% BSAand 0.05% Tween 20 for blocking of nonspecific protein-binding

dmm.biologists.org136

Damage in Pex11+/– and Pex11–/– brainRESEARCH ARTICLE

Table 3. Primary antibodies used in this study

Target Host Source; catalog number Dilution WB

Dilution IC,

brain sections

Dilution IC,

neuronal cultures

-tubulin Ms Sigma-Aldrich, Deisenhofen, Germany; T5168 1:5000 – –

Cleaved caspase-3 Rb Cell Signaling, Biozol, Eching, Germany; #9664 – 1:500 1:100

Catalase Rb Gift from Denis I. Crane, Griffith University, Brisbane, Australia 1:1000 1:2000 –

GFAP Ms Chemicon, Hofheim, Germany; MAB 3402 1:20,000 – –

Histone H3 Rb Cell Signaling, Biozol, Eching, Germany; #9715 1:1000 – –

MAP2 Ch Novus Biologicals, Littleton, CO; NB 330-213 – – 1:500

MAP2 Ms Sigma-Aldrich, Deisenhofen, Germany; M4403 1:500 – –

8-OHdG Ms Cosmo Bio Co., Tokyo, Japan; N45.1 1:200 1:100 –

PEX14 Rb Gift from Denis I. Crane, Griffith University, Brisbane, Australia 1:500 1:2000 1:1000

SOD1 Rb RDI Systems, Flanders, NJ; RDI-RTSODabr 1:10,000 – –

SOD2 Rb RDI Systems, Flanders, NJ; RDI-RTSODMabr 1:10,000 – –

SOD2 Rb Abcam, Cambridge, UK; ab13533 – 1:10,000 1:2000

Synaptophysin Rb DakoCytomation, Glostrup, Denmark; M0076 1:500 1:400 –

WB, western blotting; IC, immunofluorescence staining; Ms, mouse; Rb, rabbit; Ch, chicken.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 13: Deletion of a single allele of the Pex11 gene is ...of catalase remained unchanged in neurons from Pex11 +/– mice and was reduced in those from Pex11 –/– mice, suggesting a partial

sites. Indirect immunofluorescence was performed as previouslydescribed (Ahlemeyer et al., 2007) by one or two subsequentincubations with different sets of primary antibodies plus secondaryantibody incubations separated by extensive washing steps (fordetails see Table 3). Images of immunofluorescence preparationswere taken with a confocal laser scanning microscope (Leica TCSSP2, Leica, Bensheim, Germany). The number of PEX14-positiveperoxisomes/100 m2 was determined as described above in fourareas in each of the six different Petri dishes in each of the threePex11 genotypes. Values are expressed as number ofperoxisomes/100 m2.

Characterization of neuronal damageNeuronal damage was characterized by four different approachesusing either Hoechst 33342 (nuclear stain), the membrane-impermeable dye propidium iodide (to detect membrane damage),an antibody against the active (cleaved) form of caspase-3 or theTUNEL assay. In three different sets of experiments, cells werestained with either propidium iodide or anti-active-caspase-3antibodies or TUNEL combined with Hoechst 33342 (Fig. 1A-K). The numbers of propidium-iodide-, active-caspase-3- orTUNEL-positive cells were counted in relation to the number ofneurons with apoptotic nuclear morphology as detected byHoechst 33342 in the same areas within the same cultures. In allexperiments, three distinct areas in each of the four different Petridishes (corresponding to 800-1200 cells) were determined for eachPex11 genotype all derived from the same litter. Positive andnegative controls for TUNEL staining were performed asdescribed above for the brain sections. Images were taken with

a Leica DM RD fluorescence microscope equipped with a LeicaDC480 camera (Leica, Bensheim, Germany). Data are presentedas percentage of neuronal death (cells with an apoptotic nuclearmorphology or propidium-iodide-, activated-caspase-3- orTUNEL-positive cells).

Drug treatmentTo get further insight into the pathomechanism of neuronal deathin peroxisome deficiency, cortical cultures were treated with theantioxidant -tocopherol (10 M) or the broad spectrum caspaseinhibitor z-VAD.fmk (100 M), which were both added 4 hoursafter seeding to half of the Petri dishes prepared from one animal.All drugs remained continuously in the culture medium up to day7. Control cultures received vehicle only.

RT-PCR analysesTotal RNA from cultured cortical neurons of distinct Pex11genotypes was prepared using the RNAeasy Mini kit (Qiagen,Hilden, Germany). cDNA was synthesized from DNase-I-treatedtotal RNA using the SuperScript II First-Strand Synthesis Systemplus RNaseOut. For semi-quantitative analysis, specific primers foreach gene were designed using the PRIMER3 program(http://www.ncbi.nlm.nih.gov/tools/primer-blast) and synthesizedby Eurofins/MWG/Operon (Ebersberg, Germany; Table 4). PCRreaction was performed containing 50 ng cDNA, 100 nmol forwardand reverse primers and 5PRIME TaqDNA polymerase in a finalvolume of 25 l 5PRIME Master Mix (5PRIME, Hamburg,Germany), with the following parameters: denaturation at 95°C for2 minutes; followed by 32-45 cycles of denaturation at 95°C for 30

Disease Models & Mechanisms 137

Damage in Pex11+/– and Pex11–/– brain RESEARCH ARTICLE

Table 4. Primers used in this study for semiquantitaive RT-PCR

Target gene Accession number Sequence (3 to 5 ) °C Cyc

F GTCCAGTGCGCTGTAGATGTGAAACatalase (Cat) NM_009804.2

R GAGCAGCGGAGTTACAGGTTAGC

65 32

F GAGGAGTGGTATCGGTCTAAGTTTGGfap NM_010277.31

R GCCGCTCTAGGGACTCGTT

65 39

F CCGGCTCAAGGAATATGAAAFis1 NM_001163243.1

R CCATGCCTACCAGTCCATCT

60 35

F CGGTGGTGCT AGG ATT TGT TDnm1l NM_0152816.2

R GCACCATTTCATTTGTCACG

60 35

F AACATTCTGCTGGGGGCGGAMtap2 NM_001039934.1

R TGCTTAGCAAGCGCCGCAGT

60 36

F AACTCTACAAGAGGTACCTGPex14 NM_019781.2

R TGCACCTGAGTCACTGTCTGG

60 35

F AGCGGTGAACCAGTTGTGTTGTSod1 NM_011434.1

R CCACACAGGGAATGTTTACTGC

65 33

F AAGTAGGTAGGGCCTGTCCGATGSod2 NM_013671.3

R CTAAGGGACCCAGACCCAACAAG

60 31

F CTGGGCCAAAGGCCTGTCCGSyp NM_009305.2

R CCTGCCCATAGCCCGCATCG

65 38

F AAAGCGGGTGGTAAACTCCA28S rRNA NR_003279.1

R GGTTTCACGCCCTCTTGAAC

60 35

F, forward primer; R, reverse primer; ° C, annealing temperature; Cyc, number of cycles.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 14: Deletion of a single allele of the Pex11 gene is ...of catalase remained unchanged in neurons from Pex11 +/– mice and was reduced in those from Pex11 –/– mice, suggesting a partial

seconds, annealing at 50-65°C for 1 minute, extension at 72°C for1 minute; and a final extension at 72°C, 7 minutes (1 cycle) in aBio-Rad iCycler. Reaction products were the separated on 2%agarose gels, stained with ethidium bromide, and photographedusing the Gel-Doc 2000 documentation system from BioRad.Quantitative PCR analysis was performed using the real-time SYBRGreen method on a BioRad thermal iQ5 cycler using the iQ SYBRGreen Supermix from BioRad and specific primers. Primersequences were selected using the Massachusetts General Hospital(MGH) Primer bank (http://pga.mgh.harvard.edu/primerbank);PCR products were first visualized on a 2.5% SYBR Gold agarosegel to confirm the correct band size. The efficiency coefficient ofeach primer was determined by a tenfold dilution series in thethermal iQ5 cycler; the values are shown together with the primersequences in Table 2. Each reaction was performed in triplicate foreach template using the mean value of the wells for final calculation.Normalization for cDNA quantity was done using GAPDH controlprimers for each template. The relative expression of a defined gene (ratio between either heterozygous versus wild-type orhomozygous versus wild-type of templates from the same litter)was calculated using the Pfaffl equation (Pfaffl et al., 2001). Meanratio values from three different litters are shown in Fig. 4D andTable 2.

Determination of the cellular ROS levelThe cell-permeable probe (di)hydroethidine is preferentiallyoxidized by superoxide to its fluorescent product, ethidine. Ethidineis retained intracellularly, thus allowing quantitative estimations ofthe cellular ROS level (Bindokas et al., 1996; Ahlemeyer et al., 2001).Dihydroethidine was prepared as a stock of 5 mM in DMSO andwas added to the culture medium to a final concentration of 5 M.After incubation for 20 minutes, cortical neurons from distinctPex11 genotypes, grown on coverslips, were washed with PBS andfixed with 4% paraformaldehyde in PBS for 20 minutes. Thecoverslips were mounted on slices for measuring cellular ethidinefluorescence under a confocal laser scanning microscope (LeicaTCS SP2, Leica, Germany). Images were taken with a 40�fluorescence objective (HCxPL Apo CS 40� 1.25 Oil) using the519 nm laser line (argon/krypton laser) under fixed settings withrespect to laser energy, signal detection (gain, offset, pinhole size)for each series of experiments. Ten regions in each of the threedifferent Petri dishes for each genotype (corresponding to 2500-3000 cells) were chosen, scanned in the single scan mode and thefluorescent images were saved and digitalized in a 512�512 pixelformat. Ethidine fluorescence was quantified individually in all cellsusing the Leica Confocal software program (Leica, Bensheim,Germany). The values are expressed as mean fluorescence intensity(MFI) of ethidine per cell.

Western blot analysisNeuronal cultures from different Pex11 genotypes (3�105 cells)were homogenized in 50 l of a homogenization buffer containing50 mM MOPS, pH 7.4, 250 mM sucrose, 1 mM EDTA, 0.1% ethanol(v/v) and 10% protease inhibitor mix M (Serva, Heidelberg,Germany) using a 2 ml Potter-Elvehjem homogenizer. Thehomogenate was centrifuged at 400 g for 10 minutes at 4°C toremove nuclei and non-homogenized cell debris. The proteinamounts of the nuclear fraction and supernatants were determinedaccording to Bradford (Bradford, 1976) using BSA as standard.Protein samples (4-8 g) were separated on 12-15% SDS-polyacrylamide gels (depending on the expected band size of theantigen to be detected) and transferred onto a polyvinylidenedifluoride membrane by semi-dry blotting (Transcell Blot SD,BioRad, München, Germany). In one series of experiments (Fig.6C), nuclear extracts were spotted onto the membranes for thepreparation of a Dot blot to semi-quantify 8-OHdGimmunoreactivity. Nonspecific protein binding on the membraneswas blocked using Tris-buffered saline with 0.05% Tween 20(TBST) and 10% non-fat dry milk (blocking buffer). The blots wereincubated for 1 hour at room temperature with the primaryantibodies in TBST with 5% non-fat dry milk, washed intensivelyand incubated for 1 hour at room temperature with the secondaryantibodies in TBST (for details see Table 3). Alkaline phosphataseactivity was detected using Immun-Star AP substrate from BioRad(München, Germany). The blots were exposed to Kodak Bio-MaxMR-1 films (Sigma-Aldrich, Deisenhofen, Germany), scanned andthe integrated optical densities of the signals were analyzedsemiquantitatively using QuantityOne (BioRad, München,Germany). We calculated the ratio of the integrated optical densitiesof each tested protein versus tubulin. Data are given as relativepercentage of a distinct protein in comparison to that of the wild-type cultures, which was set to 100%.

dmm.biologists.org138

Damage in Pex11+/– and Pex11–/– brainRESEARCH ARTICLE

TRANSLATIONAL IMPACT

Clinical issuePeroxisomal biogenesis disorders (PBDs) are inherited in an autosomalrecessive manner and can be caused by mutations in PEX genes, of whichthere are at least 12 in humans. PEX genes encode peroxin proteins, which arerequired for the normal biogenesis or maintenance of peroxisomes(intracellular organelles with a range of essential biochemical functions). PBDsare characterized by disturbances in developmental and metabolichomeostasis, predominantly affecting the liver, kidney and brain. The clinicalphenotype varies widely: Zellweger syndrome is the most severe form (survivalrate is less than 1 year), followed by neonatal adrenoleukodystrophy and amilder form called infantile Refsum’s disease, where patients survive into thesecond decade. Obtaining further insight into the molecular pathologenesisand pathologies of these devastating disorders is of high interest.

ResultsThis study addresses the effects in mice of homozygous versus heterozygousdeletion of Pex11, a gene previously implicated in the regulation ofperoxisomal proliferation. In their analyses of primary neuronal cultures and ofbrain samples from Pex11+/+, Pex11 +/– and Pex11 –/– mice, the authors findthat the proportion of cell death in heterozygous mice is higher than in wild-type mice, but less than in homozygous knockout mice. Moreover, the extentof cell death correlates with a decrease in peroxisome number inhomozygotes, but not in heterozygotes. Heterozygotes also show delayedneuronal differentiation and increased levels of oxidative stress, which areboth, however, more pronounced in homozygotes. Importantly, the authorscarefully analyze brain alterations not only in homozygous and wild-type mice,but also in heterozygous animals, in which a phenotype has not previouslybeen reported.

Implications and future directionsThe finding that loss of function of a single Pex11 allele can causeneurological symptoms in mice might have important implications for thediagnosis and future research of PBDs – as well as other recessive disorders –in humans. In addition, these new data on the role of oxidative stress in thepathology of PBDs might help to develop new antioxidant-based strategies fortreating the disorders in humans.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 15: Deletion of a single allele of the Pex11 gene is ...of catalase remained unchanged in neurons from Pex11 +/– mice and was reduced in those from Pex11 –/– mice, suggesting a partial

Statistical analysisIn some series of experiments, data were derived from three orfour litters (Fig. 1D, Fig. 4B, Fig. 7E,Hb, Table 2) and statisticaldifferences between the genotypes were evaluated by ANOVA-1followed by post-hoc Scheffé test. Data from four litters were furtheranalyzed by Levene’s test followed by general linear model analysis(univariant mode) and Scheffé, Tukey and Sidak multicomparisonpost-hoc analysis showing that data evaluation is independent ofthe litter. In addition, the normality of the values was ensured bya Shapiro-Wilk test and we excluded residual influencingparameters by a Q-Q-Plot analysis. Therefore, when data werederived from several culture dishes and/or areas, but from the samelitter, the univariant (Fig. 1C,F, Fig. 3C-F, Fig. 4B) or multivariant(Fig. 1G, Fig. 7D) mode of the general linear model for repeatedexperiments followed by post-hoc Scheffé test was used to evaluatestatistical differences between the genotypes. In case of a drugtreatment (Fig. 1E, Fig. 7B,C), differences between non-treated andtreated groups were analyzed by nonparametrical test for multiplerelated samples followed by Friedman test.ACKNOWLEDGEMENTSWe thank Gabriele Thiele, Andrea Textor and Elke Richter for excellent technicalassistance, and Johannes Herrmann from the Hochschulrechenzentrum of theJustus-Liebig-University of Giessen for his advice in the statistical evaluation of ourdata.

FUNDINGThis research received no specific grant from any funding agency in the public,commercial or not-for-profit sectors.

COMPETING INTERESTSThe authors declare that they do not have any competing or financial interests.

AUTHOR CONTRIBUTIONSE.B.-V. and B.A. conceived and designed the experiments. E.B.-V. provided advice,gave suggestions and financial support. M.G. performed tissue sectioning andimmunofluorescence staining on brain sections, B.A. performed all otherexperiments and data analysis. B.A. and E.B.-V. wrote the paper.

REFERENCESAhlemeyer, B. and Baumgart-Vogt, E. (2005). Optimized protocols for the

simultaneous preparation of primary neuronal cultures of the neocortex,hippocampus and cerebellum from individual newborn (P0.5) C57Bl/6J mice. J.Neurosci. Methods 149, 110-120.

Ahlemeyer, B., Bauerbach, E., Plath, M., Steuber, M., Heers, C., Tegtmeier, F. andKrieglstein, J. (2001). Retinoic acid reduces apoptosis and oxidative stress bypreservation of SOD protein level. Free Radic. Biol. Med. 30, 1067-1077.

Ahlemeyer, B., Neubert, I., Kovacs, W. and Baumgart-Vogt, E. (2007). Differentialexpression of peroxisomal matrix and membrane proteins during postnataldevelopment of mouse brain. J. Comp. Neurol. 505, 1-17.

Baes, M., Gressens, P., Baumgart, E., Carmeliet, P., Casteels, M., Fransen, M.,Evrard, P., Fahimi, D., Declercq, P. E., Collen, D. et al. (1997). A mouse model forZellweger syndrome. Nat. Genet. 17, 49-57.

Baes, M., Gressens, P., Huyghe, S., De, N. K., Qi, C., Jia, Y., Mannaerts, G. P., Evrard,P., Van Veldhoven, P. P., Declercq, P. E. and Reddy, J. K. (2002). The neuronalmigration defect in mice with Zellweger syndrome (Pex5 knockout) is not caused bythe inactivity of peroxisomal -oxidation. J. Neuropathol. Exp. Neurol. 61, 368-374.

Baumgart, E., Vanhorebeek, I., Grabenbauer, M., Borgers, M., Declercq, P. E.,Fahimi, H. D. and Baes, M. (2001). Mitochondrial alterations caused by defectiveperoxisomal biogenesis in a mouse model for Zellweger syndrome (Pex5 knockoutmouse). Am. J. Pathol. 159, 1477-1494.

Baumgart, E., Fahimi, D. H., Steininger, H. and Grabenbauer, M. (2003). A review ofmorphological techniques for detection of peroxisomal (and mitochondrial) proteinsand their corresponding mRNAs during ontogenesis in mice: application to the PEX5knockout mouse with Zellweger syndrome. Microsc. Res. Tech. 61, 121-138.

Berger, J. and Gärtner, J. (2006). X-linked adrenoleukodystrophy: clinical, biochemicaland pathogenetic aspects. Biochim. Biophys. Acta 1763, 1721-1732.

Bindokas, V. P., Jordan, J., Lee, C. C. and Miller, R. J. (1996). Superoxide productionin rat hippocampal neurons: selective imaging with hydroethidine. J. Neurosci. 16,1324-1336.

Bonekamp, N. A., Völkl, A., Fahimi, H. D. and Schrader, M. (2009). Reactive oxygenspecies and peroxisomes: struggling for balance. Biofactors 35, 346-355.

Bradford, M. M. (1976). A rapid and sensitive method for the quantification ofmicrogram quantities of protein utilizing the principle of protein dye binding. Anal.Biochem. 72, 248-254.

Brites, P., Motley, A. M., Gressens, P., Mooyer, P. A., Ploegaert, I., Everts, V., Evrard,P., Carmeliet, P., Dewerchin, M., Schoonjans, L. et al. (2003). Impaired neuronalmigration and enchondral ossification in Pex7 knockout mice: a model for rhizomelicchondrodysplasia punctata. Hum. Mol. Genet. 12, 2255-2267.

Brosius, U. and Gärtner, J. (2002). Cellular and molecular aspects of Zellwegersyndrome and other peroxisome biogenesis disorders. Cell. Mol. Life Sci. 59, 1058-1069.

Crandall, J. E. and Fischer, I. (1989). Developmental regulation of microtubule-associated protein 2 expression in regions of mouse brain. J. Neurochem. 53, 1910-1917.

Delille, H. K., Agricola, B., Guimaraes, S. C., Borta, H., Luers, G. H., Fransen, M. andSchrader, M. (2010). Pex11pbeta-mediated growth and division of mammalianperoxisomes follows a maturation pathway. J. Cell Sci. 123, 2750-2762.

Di Benedetto, R., Denti, M. A., Salvati, S., Attorri, L. and Di Biase, A. (2009). PMP70knock-down generates oxidative stress and pro-inflammatory cytokine productionin C6 glial cells. Neurochem. Int. 54, 37-42.

Dirkx, R., Vanhorebeek, I., Martens, K., Schad, A., Grabenbauer, M., Fahimi, D.,Declercq, P., Van Veldhoven, P. P. and Baes, M. (2005). Absence of peroxisomes inmouse hepatocytes causes mitochondrial and ER abnormalities. Hepatology 41, 868-878.

Dumser, M., Bauer, J., Lassmann, H., Berger, J. and Forss-Petter, S. (2007). Lack ofadrenoleukodystrophy protein enhances oligodendrocyte disturbance and microgliaactivation in mice with combined Abcd1/Mag deficiency. Acta Neuropathol. 114,573-586.

Faust, P. L. (2003). Abnormal cerebellar histogenesis in PEX2 Zellweger mice reflectsmultiple neuronal defects induced by peroxisome deficiency. J. Comp. Neurol. 461,394-413.

Faust, P. L. and Hatten, M. E. (1997). Targeted deletion of the PEX2 peroxisomeassembly gene in mice provides a model for Zellweger syndrome, a human neuronalmigration disorder. J. Cell Biol. 139, 1293-1305.

Faust, P. L., Banka, D., Siriratsivawong, R., Ng, V. G. and Wikander, T. M. (2005).Peroxisome biogenesis disorders: the role of peroxisomes and metabolic dysfunctionin developing brain. J. Inherit. Metab. Dis. 28, 369-383.

Ferdinandusse, S., Finckh, B., de Hingh, Y. C., Stroomer, L. E., Denis, S.,Kohlschütter, A. and Wanders, R. J. (2003). Evidence for increased oxidative stress in peroxisomal D-bifunctional protein deficiency. Mol. Genet. Metab. 79, 281-287.

Ferreira, A., Busciglio, J. and Caceres, A. (1987). An immunocytochemical analysis ofthe ontogeny of the microtubule-associated proteins MAP-2 and Tau in the nervoussystem of the rat. Dev. Brain Res. 34, 9-31.

Forss-Petter, S., Werner, H., Berger, J., Lassmann, H., Molzer, B., Schwab, M. H.,Bernheimer, H., Zimmermann, F. and Nave, K. A. (1997). Targeted inactivation ofthe X-linked adrenoleukodystrophy gene in mice. J. Neurosci. Res. 50, 829-843.

Fox, M. A., Walsh, L. A., Nieuwesteeg, M. and Damjanovski, S. (2011). PEX11binduces peroxisomal gene expression and alters peroxisome number during earlyXenopus laevis development. BMC Dev. Biol. 11, 24-34.

Gabriel, S. E., Brigman, K. N., Koller, B. H., Boucher, R. C. and Stutts, M. J. (1994).Cystic fibrosis heterozygote resistance to cholera toxin in the cystic fibrosis mousemodel. Science 5182, 107-109.

Gärtner, J. (2003). Is there a phenotype/genotype correlation in peroxisomebiogenesis disorders (PBDs)? Adv. Exp. Med. Biol. 544, 59-65.

Geisbrecht, B. V., Collins, C. S., Reuber, B. E. and Gould, S. J. (1998). Disruption of aPEX1-PEX6 interaction is the most common cause of the neurologic disordersZellweger syndrome, neonatal adrenoleukodystrophy, and infantile Refsum disease.Proc. Natl. Acad. Sci. USA 95, 8630-8635.

Gillespie, J. (2004). Population Genetics: a Concise Guide, 2nd edn. Baltimore: JohnsHopkins University Press.

Grabenbauer, M., Fahimi, H. D. and Baumgart-Vogt, E. (2001). Detection ofperoxisomal proteins and their mRNAs in serial sections of fetal and newborn mouseorgans. J. Histochem. Cytochem. 49, 155-164.

Hedrich, K., Hagenah, J., Djarmati, A., Hiller, A., Lohnau, T., Lasek, K., Grünewald,A., Hilker, R., Steinlechner, S., Boston, H. et al. (2006). Clinical spectrum ofhomozygous and heterozygous PINK1 mutations in a large German family withParkinson disease: role of a single hit? Arch. Neurol. 63, 833-838.

Imamura, A., Shimozawa, N., Suzuki, Y., Zhang, Z., Tsukamoto, T., Orii, T., Osumi, T.and Kondo, N. (2001). Temperature sensitive acyl-CoA oxidase import in group Aperoxisome biogenesis disorders. J. Med. Genet. 38, 871-874.

Kassmann, C. M., Lappe-Siefke, C., Baes, M., Brügger, B., Mildner, A., Werner, H. B.,Natt, O., Michaelis, T., Prinz, M., Frahm, J. et al. (2007). Axonal loss and

Disease Models & Mechanisms 139

Damage in Pex11+/– and Pex11–/– brain RESEARCH ARTICLED

iseas

e M

odel

s & M

echa

nism

s

DM

M

Page 16: Deletion of a single allele of the Pex11 gene is ...of catalase remained unchanged in neurons from Pex11 +/– mice and was reduced in those from Pex11 –/– mice, suggesting a partial

neuroinflammation caused by peroxisome-deficient oligodendrocytes. Nat. Genet.39, 969-976.

Keane, M. H., Overmars, H., Wikander, T. M., Ferdinandusse, S., Duran, M.,Wanders, R. J. and Faust, P. L. (2007). Bile acid treatment alters hepatic disease andbile acid transport in peroxisome-deficient PEX2 Zellweger mice. Hepatology 45,982-997.

Kobayashi, S., Tanaka, A. and Fujiki, Y. (2007). Fis1, DLP1 and Pex11p coordinatelyregulated peroxisome morphogenesis. Exp. Cell Res. 313, 1675-1686.

Kobayashi, T., Shinnoh, N., Kondo, A. and Yamada, T. (1997). Adrenoleukodystrophyprotein-deficient mice represent abnormality of very long chain fatty acidmetabolism. Biochem. Biophys. Res. Commun. 232, 631-636.

Lanyon-Hogg, T., Warriner, S. L. and Baker, A. (2010). Getting a camel through theeye of a needle: the import of folded proteins by peroxisomes. Biol. Cell 102, 245-263.

Li, L., Tasic, B., Micheva, K. D., Ivanov, V. M., Spletter, M. L., Smith, S. J. and Luo, L.(2010). Visualizing the distribution of synapses from individual neurons in the mousebrain. PLoS ONE 5, e11503.

Li, X., Baumgart, E., Dong, G. X., Morrell, J. C., Jimenez-Sanchez, G., Valle, D.,Smith, K. D. and Gould, S. J. (2002a). PEX11 is required for peroxisomeproliferation in response to 4-phenylbutyrate but is dispensable for peroxisomeproliferator-activated receptor alpha-mediated peroxisome proliferation. Mol. Cell.Biol. 22, 8226-8240.

Li, X., Baumgart, E., Morrell, J. C., Jimenez-Sanchez, G., Valle, D. and Gould, S. J.(2002b). PEX11 deficiency is lethal and impairs neuronal migration but does notabrogate peroxisome function. Mol. Cell. Biol. 22, 4358-4365.

Lu, J. F., Lawler, A. M., Watkins, P. A., Powers, J. M., Moser, A. B., Moser, H. W. andSmith, K. D. (1997). A mouse model for X-linked adrenoleukodystrophy. Proc. Natl.Acad. Sci. USA 94, 9366-9371.

Maxwell, M., Bjorkman, J., Nguyen, T., Sharp, P., Finnie, J., Paterson, C., Tonks, I.,Paton, B. C., Kay, G. F. and Crane, D. I. (2003). Pex13 inactivation in the mousedisrupts peroxisome biogenesis and leads to a Zellweger syndrome phenotype. Mol.Cell. Biol. 23, 5947-5957.

McGuinness, M. C., Lu, J. F., Zhang, H. P., Dong, G. X., Heinzer, A. K., Watkins, P. A.,Powers, J. and Smith, K. D. (2003). Role of ALDP (ABCD1) and mitochondria in X-linked adrenoleukodystrophy. Mol. Cell. Biol. 23, 744-753.

Mezei, M., Andersen, P. M., Stewart, H., Weber, M. and Eisen, A. (1999). Motorsystem abnormalities in heterozygous relatives of a D90A homozygous CuZn-SODALS patient of finnish extraction. Neurol. Sci. 169, 49-55.

Müller, C. C., Nguyen, T. H., Ahlemeyer, B., Meshram, M., Santrampurwala, N., Cao,S., Sharp, P., Fietz, P. B., Baumgart-Vogt, E. and Crane, D. I. (2011). PEX13deficiency in mouse brain as a model of Zellweger syndrome: abnormal cerebellumformation, reactive gliosis and oxidative stress. Dis. Model. Mech. 4, 104-119.

Otto, S. P. and Yong, P. (2002). The evolution of gene duplicates. Adv. Genet. 46, 451-483.

Pfaffl, M. W., Horgan, G. W. and Dempfle, L. (2001). A new mathematical model forrelative quantification in real-time RT-PCR. Nucleic Acids Res. 29, e45.

Powers, J. M. and Moser, H. W. (1998). Peroxisomal disorders: genotype, phenotype,major neuropathologic lesions, and pathogenesis. Brain Pathol. 8, 101-120.

Powers, J. M., Pei, Z., Heinzer, A. K., Deering, R., Moser, A. B., Moser, H. W.,Watkins, P. A. and Smith, K. D. (2005). Adrenoleukodystrophy: oxidative stress ofmice and men. J. Neuropathol. Exp. Neurol. 64, 1067-1079.

Przyborski, S. A. and Cambray-Deakin, M. A. (1995). Developmental regulation ofMAP2 variants during neuronal differentiation in vitro. Brain Res. Dev. Brain Res. 89,187-201.

Pujol, A., Hindelang, C., Callizot, N., Bartsch, U., Schachner, M. and Mandel, J. L.(2002). Late onset neurological phenotype of the X-ALD gene inactivation in mice: amouse model for adrenomyeloneuropathy. Hum. Mol. Genet. 11, 499-505.

Raas-Rothschild, A., Wanders, R. J., Mooijer, P. A., Gootjes, J., Waterham, H. R.,Gutman, A., Suzuki, Y., Shimozawa, N., Kondo, N., Eshel, G. et al. (2002). A PEX6-

defective peroxisomal biogenesis disorder with severe phenotype in an infant,versus mild phenotype resembling Usher syndrome in the affected parents. Am. J.

Hum. Genet. 70, 1062-1068.Robertson, C. L., Scafidi, S., McKenna, M. C. and Fiskum, G. (2009). Mitochondrial

mechanisms of cell death and neuroprotection in pediatric ischemic and traumaticbrain injury. Exp. Neurol. 218, 371-380.

Schrader, M., Reuber, B. E., Morrell, J. C., Jimenez-Sanchez, G., Obie, C., Stroh, T.A., Valle, D., Schroer, T. A. and Gould, S. J. (1998). Expression of PEX11betamediates peroxisome proliferation in the absence of extracellular stimuli. J. Biol.

Chem. 273, 29607-29614.Shimohama, S., Fujimoto, S., Sumida, Y., Akagawa, K., Shirao, T., Matsuoka, Y. and

Taniguchi, T. (1998). Differential expression of rat brain synaptic proteins indevelopment and aging. Biochem. Biophys. Res. Commun. 251, 394-398.

Steinberg, S., Chen, L., Wei, L., Moser, A., Moser, H., Cutting, G. and Braverman, N.(2004). The PEX Gene Screen: molecular diagnosis of peroxisome biogenesisdisorders in the Zellweger syndrome spectrum. Mol. Genet. Metab. 83, 252-263.

Sun, M., Latourelle, J. C., Wooten, G. F., Lew, M. F., Klein, C., Shill, H. A., Golbe, L. I.,Mark, M. H., Racette, B. A., Perlmutter, J. S. et al. (2006). Influence ofheterozygosity for parkin mutation on onset age in familial Parkinson disease: theGenePD study. Arch. Neurol. 63, 826-832.

Tamura, S., Okumoto, K., Toyama, R., Shimozawa, N., Tsukamoto, T., Suzuki, Y.,Osumi, T., Kondo, N. and Fujiki, Y. (1998). Human PEX1 cloned by functionalcomplementation on a CHO cell mutant is responsible for peroxisome-deficientZellweger syndrome of complementation group I. Proc. Natl. Acad. Sci. USA 95, 4350-4355.

Tamura, S., Matsumoto, N., Imamura, A., Shimozawa, N., Suzuki, Y., Kondo, N. andFujiki, Y. (2001). Phenotype-genotype relationships in peroxisome biogenesisdisorders of PEX1-defective complementation group 1 are defined by Pex1p-Pex6pinteraction. Biochem. J. 357, 417-426.

Taylor, D. L., Edwards, A. D. and Mehmet, H. (1999). Oxidative metabolism, apoptosisand perinatal brain injury. Brain Pathol. 9, 93-117.

Tucker, R. P., Binder, L. I., Viereck, C., Hemmings, B. A. and Matus, A. I. (1988). Thesequential appearance of low- and high-molecular-weight forms of MAP2 in thedeveloping cerebellum. J. Neurosci. 8, 4503-4512.

van Nuenen, B. F. L., Weiss, M. M., Bloem, B. R., Reetz, K., van Eimeren, T.,Lohmann, K., Hagenah, J., Pramstaller, P. P., Binkofski, F., Klein, C. et al. (2009).Heterozygous carriers of a Parkin or PINK1 mutation share a common functionalendophenotype. Neurology 72, 1041-1047.

van Roermund, C. W., Tabak, H. F., van Den Berg, M., Wanders, R. J. and Hettema,E. H. (2000). Pex11p plays a primary role in medium-chain fatty acid oxidation, aprocess that affects peroxisome number and size in Saccharomyces cerevisiae. J. Cell

Biol. 150, 489-498.Walter, C., Gootjes, J., Mooijer, P. A., Portsteffen, H., Klein, C., Waterham, H. R.,

Barth, P. G., Epplen, J. T., Kunau, W. H., Wanders, R. J. et al. (2001). Disorders ofperoxisome biogenesis due to mutations in PEX1: phenotypes and PEX1 proteinlevels. Am. J. Hum. Genet. 69, 35-48.

Watts, J. A., Morley, M., Burdick, J. T., Fiori, J. L., Ewens, W. J., Spielman, R. S. andCheung, V. G. (2002). Gene expression phenotype in heterozygous carriers of ataxiatelangiectasia. Am. J. Hum. Genet. 71, 791-800.

Weinberg, E. D. (2008). Survival advantage of the hemochromatosis C282Y mutation.Perspect. Biol. Med. 51, 98-102.

Weller, S., Gould, S. J. and Valle, D. (2003). Peroxisome biogenesis disorders. Annu.

Rev. Genomics Hum. Genet. 4, 165-211.Williams, T. N. (2006). Human red blood cell polymorphisms and malaria. Curr. Opin.

Microbiol. 9, 388-394.Wolf, J., Schliebs, W. and Erdmann, R. (2010). Peroxisomes as dynamic organelles:

peroxisomal matrix protein import. FEBS J. 16, 3268-3278.

dmm.biologists.org140

Damage in Pex11+/– and Pex11–/– brainRESEARCH ARTICLED

iseas

e M

odel

s & M

echa

nism

s

DM

M