Striatal neuronal death mediated by astrocytes from the Gcdh ...

12
ORIGINAL ARTICLE Striatal neuronal death mediated by astrocytes from the Gcdh-/- mouse model of glutaric acidemia type I Silvia Olivera-Bravo ,1 , César A. J. Ribeiro ,2 , Eugenia Isasi 1 , Emiliano Trías 1 , Guilhian Leipnitz 2 , Pablo Díaz-Amarilla 1 , Michael Woontner 3 , Cheryl Beck 3 , Stephen I. Goodman 3 , Diogo Souza 2 , Moacir Wajner 2,4 and Luis Barbeito 5, * 1 Neurobiología Celular y Molecular, IIBCE, Montevideo, CIP 11600, Uruguay, 2 Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre CEP 900035-003 RS, Brazil, 3 School of Medicine, University of Colorado Denver, Aurora, CO 80045, USA, 4 Serviço de Genética Médica do Hospital de Clínicas de Porto Alegre, Porto Alegre CEP 900035-003 RS, Brazil and 5 Institut Pasteur Montevideo, Mataojo 2020, Montevideo CIP 11400, Uruguay *To whom correspondence should be addressed. Tel: + 5982 5220910; Fax: +5982 5220910; Email: [email protected] Abstract Glutaric acidemia type I (GA-I) is an inherited neurometabolic childhood disorder caused by defective activity of glutaryl CoA dehydrogenase (GCDH) which disturb lysine (Lys) and tryptophan catabolism leading to neurotoxic accumulation of glutaric acid (GA) and related metabolites. However, it remains unknown whether GA toxicity is due to direct effects on vulnerable neurons or mediated by GA-intoxicated astrocytes that fail to support neuron function and survival. As damaged astrocytes can also contribute to sustain high GA levels, we explored the ability of Gcdh -/- mouse astrocytes to produce GA and induce neuronal death when challenged with Lys. Upon Lys treatment, Gcdh-/- astrocytes synthetized and released GA and 3-hydroxyglutaric acid (3HGA). Lys and GA treatments also increased oxidative stress and proliferation in Gcdh-/- astrocytes, both prevented by antioxidants. Pretreatment with Lys also caused Gcdh-/- astrocytes to induce extensive death of striatal and cortical neurons when compared with milder effect in WT astrocytes. Antioxidants abrogated the neuronal death induced by astrocytes exposed to Lys or GA. In contrast, Lys or GA direct exposure on Gcdh-/- or WT striatal neurons cultured in the absence of astrocytes was not toxic, indicating that neuronal death is mediated byastrocytes. In summary, GCDH-defective astrocytes actively contribute to produce and accumulate GA and 3HGA when Lys catabolism is stressed. In turn, astrocytic GA production induces a neurotoxic phenotype that kills striatal and cortical neurons byan oxidative stress-dependent mechanism. Targeting astrocytes in GA-I may prompt the development of new antioxidant-based therapeutical approaches. Introduction Glutaric acidemia type I (GA-I, MIM# 231670) is an inherited neu- rometabolic and degenerative disease of early childhood caused by lack of function mutations in the mitochondrial enzyme glu- taryl-CoA dehydrogenase (GCDH, MIM# 608801, E.C. 1.3.99.7). Re- duced GCDH activity alters -tryptophan and -lysine (Lys) catabolism (14), resulting in accumulation of glutaric (GA) and 3-hydroxyglutaric (3HGA) acids in brain and body uids. In- creased concentration of GA-I metabolites is thought to trigger the clinical features of GA-I, characterized by acute encephalo- pathic crises, and the installation of subsequent chronic motor and neurological sequels (4,5). GA-I pathological features include acute loss of striatal neurons, progressive cortical neurodegen- eration and white matter diffuse alterations (1,6). Co-rst authors. Received: December 24, 2014. Revised: March 27, 2015. Accepted: May 5, 2015 © The Author 2015. Published by Oxford University Press. All rights reserved. For Permissions, please email: [email protected] Human Molecular Genetics, 2015, Vol. 24, No. 16 45044515 doi: 10.1093/hmg/ddv175 Advance Access Publication Date: 12 May 2015 Original Article 4504 Downloaded from https://academic.oup.com/hmg/article-abstract/24/16/4504/744753 by guest on 12 April 2018

Transcript of Striatal neuronal death mediated by astrocytes from the Gcdh ...

Page 1: Striatal neuronal death mediated by astrocytes from the Gcdh ...

OR I G INA L ART I C L E

Striatal neuronal death mediated by astrocytes from theGcdh−/− mouse model of glutaric acidemia type ISilvia Olivera-Bravo†,1, César A. J. Ribeiro†,2, Eugenia Isasi1, Emiliano Trías1,Guilhian Leipnitz2, Pablo Díaz-Amarilla1, Michael Woontner3, Cheryl Beck3,Stephen I. Goodman3, Diogo Souza2, Moacir Wajner2,4 and Luis Barbeito5,*1Neurobiología Celular y Molecular, IIBCE, Montevideo, CIP 11600, Uruguay, 2Departamento de Bioquímica,Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre CEP 900035-003RS, Brazil, 3School of Medicine, University of Colorado Denver, Aurora, CO 80045, USA, 4Serviço de GenéticaMédica do Hospital de Clínicas de Porto Alegre, Porto Alegre CEP 900035-003 RS, Brazil and 5Institut PasteurMontevideo, Mataojo 2020, Montevideo CIP 11400, Uruguay

*To whom correspondence should be addressed. Tel: + 5982 5220910; Fax: +5982 5220910; Email: [email protected]

AbstractGlutaric acidemia type I (GA-I) is an inherited neurometabolic childhood disorder caused by defective activity of glutaryl CoAdehydrogenase (GCDH) which disturb lysine (Lys) and tryptophan catabolism leading to neurotoxic accumulation of glutaricacid (GA) and related metabolites. However, it remains unknown whether GA toxicity is due to direct effects on vulnerableneurons ormediated by GA-intoxicated astrocytes that fail to support neuron function and survival. As damaged astrocytes canalso contribute to sustain high GA levels, we explored the ability of Gcdh −/− mouse astrocytes to produce GA and induceneuronal death when challenged with Lys. Upon Lys treatment, Gcdh−/− astrocytes synthetized and released GA and3-hydroxyglutaric acid (3HGA). Lys and GA treatments also increased oxidative stress and proliferation in Gcdh−/− astrocytes,both prevented by antioxidants. Pretreatmentwith Lys also caused Gcdh−/− astrocytes to induce extensive death of striatal andcortical neurons when compared with milder effect in WT astrocytes. Antioxidants abrogated the neuronal death induced byastrocytes exposed to Lys or GA. In contrast, Lys or GA direct exposure on Gcdh−/− or WT striatal neurons cultured in theabsence of astrocytes was not toxic, indicating that neuronal death is mediated by astrocytes. In summary, GCDH-defectiveastrocytes actively contribute to produce and accumulate GA and 3HGAwhen Lys catabolism is stressed. In turn, astrocytic GAproduction induces a neurotoxic phenotype that kills striatal and cortical neurons by an oxidative stress-dependentmechanism. Targeting astrocytes in GA-I may prompt the development of new antioxidant-based therapeutical approaches.

IntroductionGlutaric acidemia type I (GA-I, MIM# 231670) is an inherited neu-rometabolic and degenerative disease of early childhood causedby lack of function mutations in the mitochondrial enzyme glu-taryl-CoA dehydrogenase (GCDH, MIM# 608801, E.C. 1.3.99.7). Re-duced GCDH activity alters -tryptophan and -lysine (Lys)catabolism (1–4), resulting in accumulation of glutaric (GA) and

3-hydroxyglutaric (3HGA) acids in brain and body fluids. In-creased concentration of GA-I metabolites is thought to triggerthe clinical features of GA-I, characterized by acute ‘encephalo-pathic crises’, and the installation of subsequent chronic motorand neurological sequels (4,5). GA-I pathological features includeacute loss of striatal neurons, progressive cortical neurodegen-eration and white matter diffuse alterations (1,6).

† Co-first authors.Received: December 24, 2014. Revised: March 27, 2015. Accepted: May 5, 2015

© The Author 2015. Published by Oxford University Press. All rights reserved. For Permissions, please email: [email protected]

Human Molecular Genetics, 2015, Vol. 24, No. 16 4504–4515

doi: 10.1093/hmg/ddv175Advance Access Publication Date: 12 May 2015Original Article

4504

Downloaded from https://academic.oup.com/hmg/article-abstract/24/16/4504/744753by gueston 12 April 2018

Page 2: Striatal neuronal death mediated by astrocytes from the Gcdh ...

In an effort to understand the pathophysiological mechan-isms of neuronal death in GA-I, a Gcdh−/− mouse model of thedisease, which lacks GCDH activity and consequently accumu-lates high levels of GA-I metabolites in tissues and fluids (7,8),was developed. Interestingly, Gcdh−/− mice do not show spon-taneous striatal neurodegeneration or relevant neurologicalsymptoms, unless they are fed with a high Lys diet to stimulatethe production of GA-I metabolites (7–9). Thus, while neurologicaldamage in GA-I seems to be dependent on GA and 3HGA-mediated neurotoxicity (2,10), the vulnerability of striatal neu-rons to GA-I metabolites appears to be less marked. For instance,cultured neurons from either mouse (11,12) or rats (13) appearnon-responsive to pathophysiological concentrations of GA.Moreover, Gcdh−/− neuronal cultures did not show affected sur-vival in basal conditions (14) implying that GCDH absence mightnot be sufficient to elicit neuron death.

Growing evidence suggest that astrocytes may play a pivotalrole in GA-I pathology (13–18). Astrocytes play key homeostaticand metabolic roles in the CNS and have the ability to uptakeGA (14). Upon GA and 3HGA pretreatment, cultured astrocytesreact with a phenotypic change that includes increased prolifer-ation, expression of S100β and mitochondrial dysfunction(13,15,16). In turn, such reactive astrocytes mediate the death ofstriatal neurons (13), suggesting a mechanism by which GA-da-maged astrocytes follow a profound and long-lasting functionalchange that kills vulnerable neurons. In accordance, Jafari et al.(17) reported that GA-induced astrocyte damage results indecreased ability of astrocytes to produce glutamine, leading tohyperammonemia and depletion of neuronal glutamate reser-voirs and thus, promoting neuronal and oligodendrocyte death.Other report has showed that Gcdh−/− astrocytes treated withGA or 3HGA have a reduced [14C]-succinate efflux (14), furthersuggesting a defective metabolic supply to neurons. Thus, GA-Iaccumulated metabolites may cause astrocytes to be less sup-portive to maintain synaptic and trophic activities necessaryfor the healthy brain function.

In the present study, we show evidence that Gcdh−/− astro-cytes produce and accumulate GA and 3HGA in conditions of in-creased Lys catabolism. We also show that astrocytes isolatedfrom WT and Gcdh−/− mouse pups induce neuronal death in acell-culture system where neurons are maintained on top ofastrocytic feeder layers that were previously challenged withLys or GA. Furthermore, these effects were prevented by antioxi-dant pretreatment in Gcdh−/− astrocytes. Conversely, neitherLys nor GA at the concentrations employed produced significanteffects on neuronal survival.

ResultsAstrocytes from Gcdh−/− mice generate GA and 3HGAwhen exposed to Lys

Although the neuronal expression of GCDH is consensually re-cognized (6,14,18,19), we assessed whether this enzymatic path-way is part of the astrocyte signaling repertoire to maintain CNShomeostasis (19–21). The analysis of GCDH expression by using apolyclonal anti-GCDH in astrocyte cultures from WT miceevidenced a punctate pattern suggesting amitochondrial expres-sion of this enzymatic protein. In co-cultures, GCDH immunor-eactivity in neurons was much higher than that of astrocytes(Fig. 1A). The ability of WT and Gcdh−/− astrocytes isolatedfrom the cerebral cortex to produce GA and 3HGA was then as-sessed by using gas chromatography coupled to mass spectrom-etry (GC/MS). GC/MS analyses show that Gcdh−/− astrocytes

secreted low levels of GA-I metabolites in basal conditions andthat levels increased several timeswhen astrocyteswere exposedto Lys (Fig. 1B). Quantitative results indicate that in basal condi-tions Gcdh −/− astrocytes released 11 timesmore GA (1.94 versus0.17 µ) and 2.5-fold more 3HGA (0.22 versus 0.10 µ) than WTastrocytes (Table 1). In the presence of Lys, Gcdh−/− astrocytesreleased 15 times more GA (64 versus 1.94 µ) and two timesmore 3HGA (0.49 versus 0.22 µ) than WT astrocytes (Table 1).Interestingly, for both astrocyte backgrounds, Lys exposition in-creased around 30–40 times the amount of GA released and 2–3times that of 3HGA (Table 1). Variable amounts of GA (rangingfrom 3.68 to 5.51 µ and 1.4 to 22.6 µ), and 3HGA (rangingfrom 0.28 to 0.39 µ and 0.37 to 0.57 µ), were also detected incell homogenates fromWTandGcdh−/− astrocytes, respectively;thus indicating that part of the GA and 3HGA synthetized by as-trocytes remained inside the cells.

Astrocytes exposed to Lys undergo oxidative stress andincreased proliferation

Neurotoxic levels of GA have been showed to promote sub-lethaltoxicity in astrocytes, characterized by mitochondrial dysfunc-tion, increased expression of S100β and exacerbated proliferation(13,15,16). Similarly, a single exposure to 10 m Lys induced asignificant degree of sub-lethal damage in Gcdh−/− corticalastrocytes when compared with those maintained in low Lysconcentrations. Ten millimolar Lys elicited oxidative stress asestimated by carboxy-H2DCFDA (DCF), diminution of glutathione(GSH) levels and increased levels of Thiobarbituric acid reactivesubstances (TBARS) (Fig. 2A andB), whichwas associatedwith in-creased number of S100β expressing astrocytes and proliferatingcells labelled with 5-bromo-3′-deoxyuridine (BrdU). In compari-son, WT astrocytes that express functional GCDH also reactedto Lys overload with a comparable increased cell proliferation,S100β expression and oxidative stress, although the amplitudeof the response was less prominent than Gcdh−/− astrocytes(Fig. 2A and B). Lys-induced astrocyte damage was not observedat lower concentrations (Supplementary Material, Table S1).

Exposure of Gcdh−/− astrocytes to 5 m GA induced similareffects that Lys, suggesting Lys toxicity could be at least partiallymediated by the production of cytotoxic levels of GA.

Astrocytes exposed to Lys caused death of striatalneurons

To determine whether Lys exposure to Gcdh−/−astrocytes couldmodulate their ability to support neuronal growth in co-culture,astrocyteswere exposed to 10 m Lys for 24 h and, after washing,neuronswere seeded for 3 days on the top of astrocytemonolayerfor evaluation of survival andmorphology. For comparison, simi-lar experiments were performed using 5 m GA instead of Lys.WT and Gcdh−/− astrocyte feeder layers equally supported thesurvival of striatal neurons in the absence of Lys or GA challenge(Fig. 3A). Exposure to Lys or GA caused astrocytes to become toxicfor striatal neurons causing a 35–45% decreased survival (Fig. 3B)and decreased body size and simpler pattern of primary den-drites in either condition (Table 2). Astrocyte-mediated neuronaltoxicity induced by Lys was not significantly different betweenWT and Gcdh−/−astrocytes. Similarly, striatal neurons fromboth WT and Gcdh−/− mouse embryos were equally vulnerableto astrocytes challenged with Lys, although Gcdh−/− neuronsappeared withmore limited growth potential andmore sensitiveto astrocyte-induced damage (Fig. 3B).

Human Molecular Genetics, 2015, Vol. 24, No. 16 | 4505

Downloaded from https://academic.oup.com/hmg/article-abstract/24/16/4504/744753by gueston 12 April 2018

Page 3: Striatal neuronal death mediated by astrocytes from the Gcdh ...

Figure 1. Astrocytes express GCDH in cultures and co-cultures with neurons. (A) GCDH immunoreactivity in WT astrocyte cultures shows a perinuclear punctate specific

expression. The same pattern was maintained in astrocytes co-cultured with WT neurons in spite of the astrocytic signal (short arrow) was much lower than that of

neurons (long arrow). In co-cultures of Gcdh−/−astrocytes (KO) with WT neurons, the high neuronal GCDH expression (long arrows) coexists with a very low no

specific binding in astrocytes (short arrow). Calibration bars: 25 μm. (B) CG/MS chromatograms showing the presence of detectable levels of GA (arrows) in the culture

medium of WT and Gcdh−/− astrocytes under basal conditions or exposed to 10 m Lys during 72 h.

4506 | Human Molecular Genetics, 2015, Vol. 24, No. 16

Downloaded from https://academic.oup.com/hmg/article-abstract/24/16/4504/744753by gueston 12 April 2018

Page 4: Striatal neuronal death mediated by astrocytes from the Gcdh ...

Cortical but not hippocampal neurons are also vulnerableto Lys-stimulated astrocytes

The vulnerability of other neuronal populations to the toxicityexerted by astrocytes challenged by Lys was also tested. Neuronsfrom cerebral cortex or hippocampus seeded on the top of non-stimulated WT or Gcdh−/− astrocytes developed normally, both

in number and size (Fig. 4A). Astrocytes stimulated with Lys orGA caused a 25–35% decrease in survival of cortical neurons(Fig. 4B), as well as a reduction in body size and primary neurites(Table 3). Comparedwith cortical neurons, hippocampal neuronsshowed a less marked and non-significant vulnerability to astro-cytes stimulated with Lys or GA (Fig. 4A and B). As described forstriatal neurons, cortical and hippocampal Gcdh−/− neurons

Table 1. Glutaric and 3-hydroxyglutaric acids concentrations (µM) in culture medium and cell homogenates after exposure of WT and Gcdh −/−astrocytes to 10 m Lys for 72 h

WT astrocytes Gcdh −/− astrocytesVehicle 10 m Lys Vehicle 10 m Lys

Glutaric acidCulture medium 0.17 (0.13–0.19) 4.17 (3.50–4.87) 1.94 (1.64–2.39) 64.0 (48.7–83.3)Cell homogenate 0.60 (0.24–1.61) 4.85 (3.68–5.51) 1.51 (1.35–1.73) 19.5 (14.1–22.6)

3-Hydroxyglutaric acidCulture medium 0.10 (0.10–0.11) 0.30 (0.24–0.39) 0.22 (0.18–0.28) 0.49 (0.37–0.64)Cell homogenate 0.12 (0.10–0.17) 0.33 (0.28–0.39) 0.21 (0.18–0.24) 0.43 (0.37–0.57)

Data are expressed as median and range of the values (n = 3).

Figure 2.Astrocytes increase proliferation and oxidative stress in response to Lys and GA. (A) The exposure ofWTand Gcdh−/− astrocytes to 10 m Lys or 5 mGA during

24 h caused an increased BrdUpositive proliferation (cyan); amoderate increase in S100ß (green) immunoreactivityaccompanied bya decrease inGFAP signal (red), aswell

as an increased green carboxy-H2DCFDA (DCF)fluorescence. Nucleiwere stainedwithDAPI (blue). Calibration bars: 50 μmfor upper andmidpictures and 40 μmfor bottom

image, respectively. (B) Quantitation of proliferation rate and S100ß and GFAP positive cells upon Lys and GA treatments. All values were referred as a percentage of total

cells positive toDAPI. (C) Quantitation of the oxidative stress induced by Lys or GAaftermeasuringDCFemission, lipid peroxidation byassessing TBA-RS levels (B) andGSH

levels. Data are mean ± SEM of three separate experiments. *P < 0.05 indicates statistical difference related to WT astrocytes under basal conditions.

Human Molecular Genetics, 2015, Vol. 24, No. 16 | 4507

Downloaded from https://academic.oup.com/hmg/article-abstract/24/16/4504/744753by gueston 12 April 2018

Page 5: Striatal neuronal death mediated by astrocytes from the Gcdh ...

appeared more vulnerable than those from WT animals, al-though the difference was not statistically significant (Fig. 4B).

Antioxidants prevented Lys- and GA-astrocyte damageand dependent neuronal death

Because Lys or GA potently induced oxidative stress, weevaluated whether antioxidant exposure or induction of anti-oxidant/cytoprotective defenses could prevent Lys and GA in-duced damage to astrocytes, and further protect co-culturedneurons. Lipid peroxidation and/or decrease in GSH levels eli-cited by Lys or GA in Gcdh−/− astrocytes were prevented by theantioxidants Melatonin (MEL) (22,23) and Trolox (24); as well asby tert-butyl-hydroquinone (tBHQ), an inducer of the protectiveNuclear Factor Erythroid 2-related Factor 2 (Nrf2)/antioxidant re-sponse element (ARE) pathway (25–27) (Supplementary Material,Fig. S1).

Remarkably, Figure 5 shows that both strategies applied on as-trocytes before Lys or GA challenge and co-culturing with striatalneurons prevented most of the neuronal death induced by thedirect exposure of Gcdh−/− astrocytes to Lys or GA (Fig. 5). Similarpreservation of neuron survival was observed with antioxidantstreatments on WT astrocytes (Supplementary Material, Fig. S2).MEL showed a slightly minor protection than tBHQ (Fig. 5,Supplementary Material, Fig. S2) which is likely related to theinduction of the neuroprotective Nrf2/ARE pathway as shownin Supplementary Material, Figure S1 for Gcdh−/− astrocytes.

Lys exposure does not induce cell death in Gcdh−/−striatal neurons

Sincemetabolic overload with Lys could be cytotoxic for neuronsvia a direct effect (28,29) or by producing increased levels of me-tabolites such as GA, we determined the effect of a 72 h exposure

Figure 3. Vulnerability of GCDH−/− striatal neurons to astrocytes pretreated with Lys or GA. (A) Representative panoramic light images and β-tubulin (green)

immunofluorescences of all of the background combinations in astrocyte–neuron co-cultures 4 days after plating neurons on astrocytes pretreated with vehicle (V),

10 m Lys or 5 m GA during 24 h. Note the significant decreased number and simpler morphology of neurons seeded on top of Lys or GA pretreated astrocytes. In all

conditions, Gcdh−/− neurons appeared more affected. Scale bar = 75 μm for light images and 40 μm for fluorescent pictures, respectively. (B) Quantitation of neuronal

survival denoting significant effects of Lys and GA. All values were related to WT neurons seeded on top of control WT astrocytes. *P < 0.05 indicates statistical

signification related to WT astrocytes.

4508 | Human Molecular Genetics, 2015, Vol. 24, No. 16

Downloaded from https://academic.oup.com/hmg/article-abstract/24/16/4504/744753by gueston 12 April 2018

Page 6: Striatal neuronal death mediated by astrocytes from the Gcdh ...

of 3–4 days in vitro (DIV) striatal embryonic neuronal cultures to10 m Lys or 5 m GA. Neither Lys nor GA caused alterationsin neurons survival or growth (Fig. 6A). Gcdh−/− striatal neuronsexposed to Lys showed amoderate but non-significant decreasedsurvival (Fig. 6B). Remarkably, Lys or GA did not cause significanteffects on the survival of E18 cortical and hippocampal neurons.

DiscussionRecent evidences indicate that glial cells play important patho-genic roles in neurodegeneration through non-cell autonomousmechanisms (20,30,31). In different genetic models, specific po-pulations of vulnerable neurons can degenerate when astrocytesbecome dysfunctional, affecting the clearance of extracellularglutamate and/or the release of neurotrophic factors or com-pounds fueling neuron metabolism (19,32,33). In accordance,the reduced activity of GCDH or the direct exposure of astrocytesto GA or 3HGA has been found to induce astrocyte dysfunctionand subsequent astrocyte-mediated neuronal death (13,15,16).In the present study, we have used Gcdh−/− astrocytes to furtherinvestigate the contribution of astrocytes to striatal neuronaldeath in a cell-culture system where neurons were maintainedon the top of the astrocytic feeder layer. We found evidencethat GCDH is expressed in cortical astrocytes and that Gcdh−/−astrocytes maintained in a high Lys medium can accumulateand release GA and 3HGA in a much higher extent when com-paredwithWTastrocytes exposed to Lys. Moreover, Lys exposurestrongly stimulated oxidative stress and proliferation in WT andGcdh−/− astrocytes, and caused them to become neurotoxic forco-cultured neurons. Remarkably, exposure of isolated neuronalcultures to Lys or GA did not induce any apparent death, suggest-ing astrocytes as the key pathogenic cell-type mediating neuronloss in GA-I.

We demonstrated for the first time thatWTastrocytes expresslow levels of the enzyme GCDH displaying a perinuclear punctu-ate cellular distribution reminiscent of mitochondria. This resultsuggests that astrocytes can detoxify to a certain extent the GAproduced locally during Lys or tryptophan catabolism. Remark-ably, incubation of WT or Gcdh−/− astrocytes with high Lys con-centration stimulated the production and accumulation of GAand also 3HGA in the culture medium, being several fold greater(especially for GA) in Gcdh−/− than WT astrocytes. These datasupport the view that astrocytic GCDH has the capacity to handle

GA accumulation, although the metabolic pathway can be satu-rated by high Lys concentration. Moreover, while most GA (and3HGA) produced by astrocytes appears to diffuse extracellularly,a substantial amount remains inside the cells, potentially reach-ing cytotoxic concentrations responsible for mitochondrial dys-function or switch of signaling favoring cell proliferation(13,15). We also showed here that exposure of either Gcdh−/− orWT astrocytes to high Lys or GA provoked a reactive response,characterized by oxidative stress, increased proliferation rate ofaround 50 and 70% for WT and Gcdh−/− astrocytes, respectively,as well as increased number of S100β positive cells. The reactiveresponse was more pronounced in Gcdh−/− astrocytes, asexpected for a reduced ability to handle intracellular GA. Takentogether, our results indicate that astrocytes can be a source ofGA in high catabolic states and that in turn, GA accumulationcan impact or damage keyastrocytic functions. Instead of trigger-ing necrosis or apoptosis, astrocytes respond to GA by increasingproliferation and phenotypic changes that might be relevant tounderstand GA-I pathophysiology.

Remarkably, WT and Gcdh−/− astrocytes became neurotoxicfor striatal and cortical neurons when incubated with high Lysor GA concentrations. Data obtained indicate that oxidativestress plays a leading role on Lys- and GA-dependent astrocytetoxicity. Remarkably, the solely preservation of astrocyte againstoxidative damagewas enough to protect against neuronal death.Astrocyte preservation was achieved either by direct scavengerantioxidant properties or by induction of own antioxidantresponses such as the Nrf2/ARE pathway. Activation of theastrocytic Nrf2/ARE pathway and downstream cascades werepreviously shown as neuroprotective in different damagingconditions (25–27).

On the other hand, the absence of GCDH had little effectenhancing the astrocyte-mediated neuronal death. This resultcould be explained by the fact that the Lys and GA concentra-tion used in WT astrocytes largely surpassed the capacity tometabolize GA, thus triggering similar response in both astro-cyte types. Gcdh−/− and WT astrocytes exposed to Lys notonly induced neuronal death but also reduced neuronal sizeand neurite growth, suggesting insufficient trophic or meta-bolic support preceding neuronal death. This is in agreementwith previous reports showing that Gcdh−/− astrocytes ex-posed to GA have a defective export of tricarboxylic acidcycle intermediates (14). WT astrocytes exposed to GA showeda decreased ability to produce glutamine (17), both pathwayspotentially leading to neuronal metabolic starvation and alsoto death by hyperammonemia (17). Striatal neurons were themost affected to Lys or GA treated astrocytes, when comparedto cortical and hippocampal neurons. This is in accordancewith the higher vulnerability of striatal neurons in GA-I pa-tients (3,6,9,34). In this context, the worse detrimental neuron-al effects were found in the experimental condition combiningGcdh−/− neuron and astrocyte cultures, suggesting a synergis-tic detrimental effect of reduced GCDH activity in interactingneural cells.

Regarding the understanding of GA-I pathophysiology, ourdata suggest that GA and 3HGA can be produced locally in thestriatum in conditions of GCDH genetic defects. While GCDHlevels seem to be higher in neurons than astrocytes, the latercan critically contribute to GA detoxification in normal condi-tions. On the other hand, astrocytes probably have the ability touptake GA (14), therefore contributing to buffering the compoundor its effects up to certain levels. IntactWTastrocytes can protectagainst GA neurotoxic effects (35–37), reducing the concentra-tions of glutamate in the synaptic cleft or releasing glutathione

Table 2. Effects of Lys (10 m) and GA (5 m) astrocyte pretreatmenton body size and number of primary dendrites of co-cultured striatalneurons

WT astrocytes Gcdh−/− astrocytesVehicle Lys GA Vehicle Lys GA

WT neuronsBody size 100 ± 6 77 ± 4 78 ± 6 88 ± 6 77 ± 8* 78 ± 5*First-orderdendrite

100 ± 7 74 ± 8* 74 ± 7* 100 ± 6 74 ± 8* 76 ± 7*

Gcdh−/− neuronsBody size 96 ± 6 73 ± 4* 71 ± 4* 80 ± 5 69 ± 6* 67 ± 4*First-orderdendrite

100 ± 7 72 ± 6* 92 ± 9 90 ± 6 87 ± 11 88 ± 5

Values were obtained as the percentage of body size and number of first-order

dendrites shown by WT neurons seeded on top of control WT astrocytes

(Vehicle). Data represented are mean ± SEM obtained from three separated

experiments performed by triplicate or quintuplicate.

*P < 0.05, relative to WT neurons seeded on top of control WT astrocytes.

Human Molecular Genetics, 2015, Vol. 24, No. 16 | 4509

Downloaded from https://academic.oup.com/hmg/article-abstract/24/16/4504/744753by gueston 12 April 2018

Page 7: Striatal neuronal death mediated by astrocytes from the Gcdh ...

to reduce oxidative stress (19,20). However, uponmetabolic dam-age with GA, astrocytes proliferate and lose their homeostaticnormal functions (20) adopting a phenotype less supportive ordirectly toxic to neurons. Astrocyte dysfunction elicited by Lysor GA may also impair blood brain barrier homeostasis as wellas oligodendrocyte generation (16,38), leading to the additionalneurological damage characteristic of GA-I patients (39–41).

Taken together, our data strongly suggest that dysfunctionalGcdh−/− astrocytes play a crucial role in GA-I neuropathology,and that preserving protective astrocyte phenotype is criticalto maintain neuron survival. Data showing that Lys and GA-dependent astrocyte neurotoxicity is mediated by oxidative

stress and abrogated by antioxidants; strongly suggest that inhib-ition of oxidative stressmay be avaluable therapeutic strategy forGA-I patients.

Materials and MethodsEthical statement

This study was carried out in strict accordance with the Nationallaw and Guide for the Care and Use of Laboratory Animals of theNational Institutes of Health (USA). All efforts weremade tomin-imize suffering, discomfort, stress and number of animals neces-sary to produce reliable scientific data.

Figure 4. Differential vulnerability of cortical and hippocampal neurons to astrocytes pretreated with Lys or GA. (A) Representative images of β-tubulin positive 4 DIV

cortical and hippocampal neurons plated on top of WT or Gcdh−/−astrocytes pretreated during 24 h with vehicle (V), 10 m Lys or 5 m GA. Note the significant

effects on the morphology of cortical neurons when seeded on top of Lys or GA-pretreated astrocytes in clear contrast with the very healthy appearance of

hippocampal neurons in all conditions. (B) Quantitation of survival of cortical and hippocampal neurons co-cultured on WT or Gcdh−/− astrocyte feeder layers. Note

as the viability of cortical neurons significantly decreased when co-cultured on top or WT or Gcdh−/− astrocytes pretreated with Lys or GA; whereas that of

hippocampal neurons was significantly unaffected. Values were referred as the percentage of the viability determined in WT neurons co-cultured on control WT

astrocytes. Data are media ± SEM from three separated experiments. *P < 0.05 indicates statistical difference related to WT astrocytes.

4510 | Human Molecular Genetics, 2015, Vol. 24, No. 16

Downloaded from https://academic.oup.com/hmg/article-abstract/24/16/4504/744753by gueston 12 April 2018

Page 8: Striatal neuronal death mediated by astrocytes from the Gcdh ...

Chemicals

Dulbecco’s modified Eagle’s medium (DMEM), Neurobasal me-dium, B27, glutamine, fetal bovine serum (FBS), penicillin/strep-tomycin, trypsin, DCF, poly--lysine and DAPI were purchased

from Invitrogen (Carlsbad, CA, USA). Lys, GA, BrdU, β-tubulin,tBHQ, MEL, Trolox, GFAP and S100β antibodies, as well as allother chemicals of analytical grade were obtained from Sigma(St Louis, MO, USA). Anti-GCDH antibody was kindly providedby Jonna Westover (Utah State University, USA). The anti-BrdUantibody was purchased to Dako (Carpinteria, CA, USA). Theanti heme-oxigenase 1 (HO-1) antibody was purchased to Stress-gen (San Diego, CA, USA). HRP or fluorescent secondary anti-bodies were obtained from Invitrogen Molecular Probes.Deuterium-labeled internal standards for GA (d4-GA) and HGA(d5-HGA) were obtained from MDS Isotopes, Montreal, Canada.

Animals

Gcdh−/− andWTmice of 129SvEv backgroundwere generated bycrossing heterozygotes at the Unidade Experimental Animal ofthe Hospital de Clínicas de Porto Alegre (Porto Alegre, Brazil).Pregnant females were kept in plastic cages on a 12:12 h light/dark cycle (lights on 07.00–19.00 h) with constant temperature(22 ± 1°C), and water and 20% w/w protein commercial chow adlibitum. Cultures of astrocytes were made from 1–2 days oldrat pups from seven whole litters. Neurons were obtainedfrom E17.5–18 embryos after anesthetizing and sacrificing sevenfemales. To avoid cross contamination betweenWTandGcdh−/−animals, in all conditions, animals were processed separately

Figure 5.Antioxidants abrogated neuronal death induced byastrocyte pretreatmentwith Lys or GA. (A) Light images showing the protection that 1 µMEL and 20 µ tBHQ

caused on the number and gross morphology of striatal neurons co-cultured on top of Gcdh−/− astrocyte feeder layers challenged with 10 m Lys or 5 m GA. Also note

that upon Lys and GA astrocyte challenge, the frequency of dead neurons (black arrows) on top of astrocytemonolayers seemed increased. Magnification: ×40. Calibration

bar = 25 μm. (B) Quantitation of survival ofWTand Gcdh−/− neurons upon the treatment of Gcdh−/− astrocytes within the different experimental conditions. Valueswere

related to the number of surviving neurons seeded on top of astrocytes treated with vehicle (V). *P < 0.05 means statistical differences related to each respective control.

Table 3. Effects of Lys (10 m) and GA (5 m) astrocyte pretreatmenton body size and number of primary dendrites of co-cultured corticalneurons

WT astrocytes Gcdh−/− astrocytesVehicle Lys GA Vehicle Lys GA

WT neuronsBody size 100 ± 12 80 ± 12 85 ± 12 100 ± 8 63 ± 9* 54 ± 10*First-orderdendrite

100 ± 9 95 ± 8 76 ± 8* 100 ± 12 72 ± 15 81 ± 9

Gcdh−/− neuronsBody size 93 ± 5 75 ± 8* 80 ± 10 78 ± 12 60 ± 11* 54 ± 9*First-orderdendrite

83 ± 12 64 ± 10* 68 ± 10* 75 ± 13 65 ± 15* 64 ± 13*

Values were obtained as the percentage of body size and number of first-order

dendrites shown by WT neurons seeded on top of control WT astrocytes

(Vehicle). Data represented are the mean ± SEM obtained from three separated

experiments performed by triplicate or quintuplicate.

*P < 0.05, relative to WT neurons seeded on top of control WT astrocytes.

Human Molecular Genetics, 2015, Vol. 24, No. 16 | 4511

Downloaded from https://academic.oup.com/hmg/article-abstract/24/16/4504/744753by gueston 12 April 2018

Page 9: Striatal neuronal death mediated by astrocytes from the Gcdh ...

and cultures made independently and with different dissectionmaterials. All experiments were performed at least three inde-pendent times in duplicates or triplicates.

Isolated neuronal cultures

Neurons from the striatum, fronto-parietal cortex or CA1-CA3hippocampal regions were prepared from E17-18 embryos ac-cording toVentimiglia et al. (42)withminormodifications. Briefly,pregnant females were euthanized and both uterus horns re-moved. Five to seven embryos were obtained in aseptic condi-tions and washed in sterile phosphate buffered saline solution(PBS). Then each correspondent brain region was dissected, im-mersed in fresh Neurobasal medium containing 2% B27 and1 m glutamine, cleaned, minced and mechanically dissociatedto obtain isolated cells. Around 300 000 cells (3.2–3.4 × 104 cells/cm2 density) were seeded onto 35 mm or 25000 cells on 24 multi-well plates pre-covered with 0.1 mg/ml poly--lysine and pre-washed with sterile water. At 3–4 DIV, neurons were treatedwith 10 m Lys or 5 m GA for 72 h, then fixed with 4% parafor-maldehyde (PFA) and imaged by light microscopy or immunos-tained against β-tubulin to evaluate viability and morphologicalparameters.

Astrocyte cultures and treatments

Primary astrocyte cultures were prepared from whole cortices of1- to 2-day-old Gcdh−/− and WT mice. Briefly, cortices from 4–5pups were placed in sterile PBS, cleaned from meninges, cut insmall pieces and incubated in 0.05% trypsin-EDTA for 25 min at

37°C. Trypsin was then blocked with astrocyte culture medium(DMEM supplemented with 10% FBS, 3.6 g/l HEPES, 1.2 g/lNaHCO3, 100 IU/ml penicillin and 100 μg/ml streptomycin), tissuehomogenized by repeated pipetting, and then spun 10 min at1000 rpm (CL-2, Sigma Centrifuge). The resulting pellet was re-suspended in DMEM-10% FBS and cells plated in culture bottlesat a density of 2 × 104 cells/cm2 in astrocyte culture medium.Whole media was changed every day. Once in confluence, cul-tures were enriched in astrocytes by shaking for 48 h at 250 rpmand 37°C. This procedure allows obtaining astrocyte monolayersat least 98%GFAP positive anddevoid of OX42microglial cells andGalC oligodendrocytes. After a week of enrichment, astrocyteswere trypsinized for 3–7 min and then plated on sterile 35 mmPetri dishes or 24 multiwell plates until confluence (around 1week after plating). Before each treatment, astrocytes were incu-batedwith DMEM-2% FBS during 24 h and then exposed to 10 m

Lys or 5 m GA for another 24 h (13,15). Appropriate aliquots of a500–1000 m Lys or GA stock solution were prepared in 10 m

PBS, and the pH adjusted to 7.4 immediately prior to use. Insome experiments, the antioxidants MEL (1 µ) and Trolox(soluble α-tocopherol, 10 µ) or tBHQ (20 µ), prepared inDMSO (vehicle); were used to pre-incubate Gcdh−/− astrocytesfor 1 (MEL, Trolox) or 6 h (tBHQ); respectively. Then, 10 m

Lys or 5 m GA was added to culture medium. After 24 h, pro-liferation and oxidation parameters were analyzed. In otherbatch of experiments, WT and Gcdh−/− astrocytes were pre-treated with 1 µ MEL or 20 µ tBHQ for 1 or 6 h before adding10 m Lys or 5 m GA. 24 h later, co-cultures with striatal WTor Gcdh−/− neurons were performed.

Astrocyte proliferation assay

Proliferation rate was assessed by determining the percentage ofcells that incorporated BrdU related to total DAPI positive cells.This compoundwas added at the beginning of each experimentalsituation (10 µ, 24 h) and then was recognized by immunocyto-chemistry (15). Briefly, cultured astrocytes were fixed in ice-cold4% PFA (20 min, room temperature, RT), permeabilized with0.1% Triton X-100 (20 min, RT) and denatured with 2 N HCl(37°C, 30 min). After extensive washes with 10 m, borate buffer,pH 8, the non-specific bindingwas blockedwith 5% bovine serumalbumin (BSA, 60 min, RT). After that, cells were incubated with a1:800 dilution of anti-BrdU antibody in 5% BSA (overnight, wetchamber, 4°C), washed again and finally incubated with 1:500anti-mouse Alexa 488 secondary antibody (25°C, 90 min). Cellsweremountedwith glycerol containing 1 µg/ml DAPI and imagedin an Olympus FV300 laser scanning confocal microscope. Astro-cytes that show green signal in at least 70% of the whole nucleuswere considered BrdU positive.

Co-cultures of neurons on astrocyte feeder layers

Confluent astrocyte monolayers were incubated with DMEM-2%FBS during 24 h and then exposed to 10 m Lys or 5 m GA foranother 24 h. After that, the medium was completely removedand fresh astrocyte culture medium was added to discard anyminimal contact of Lys or GA with neurons. After 6 h, themedium was completely removed and each neuron suspensionadded to astrocyte monolayers. Fifty or 300 μl of a 3 × 105

neurons/ml dilution were seeded on top of confluent astrocytemonolayers grown in 24 multiwell plates or 35 mm Petri dishes,respectively. After 30 min of rest to facilitate neuron attachmentto astrocyte feeder layer, the volume was completed to 400 or1500 µl with Neurobasal medium-2% B27 and 2% FBS. Three to

Figure 6.Absence of direct effects of Lys and GA on 3–4 DIV embryonic E18 striatal

neurons obtained from WT and Gcdh−/− mice. (A) Bright field images showing

absence of significant morphological effects of Gcdh−/− striatal neurons

submitted to 10 m Lys or 5 m GA during 72 h. Scale bar = 20 μm. (B)Quantitation of survival of WT and Gcdh−/− neurons after Lys and GA

treatments. Values were related to vehicle-treated neurons (V). No significant

decreases were found, except a marginally significant decrease (#, P < 0.1)

inGcdh−/− striatal neurons submitted to Lys. Data are mean ± SEM of 2–3

separated experiments.

4512 | Human Molecular Genetics, 2015, Vol. 24, No. 16

Downloaded from https://academic.oup.com/hmg/article-abstract/24/16/4504/744753by gueston 12 April 2018

Page 10: Striatal neuronal death mediated by astrocytes from the Gcdh ...

four days later, co-cultureswere fixedwith 4% PFA and submittedto light imaging and neuronal immunostaining.

Immunocytochemistry

This approach was performed in neuronal and astrocyte culturesas well as co-cultures. The procedures were similar in all cases(15). Briefly, cells were fixed with 4% PFA, permeabilized with0.1% Triton X-100 (20 min, RT), blocked with 5% BSA (60 min,RT) and incubated (wet chamber, overnight, 4°C) with one ortwo of the following antibodies: 1:400 anti-GFAP, 1:500 anti-S100β or1:500 anti-β-tubulin. Cells were then washed and incu-batedwith correspondent 1:500 dilutions of secondaryantibodiesconjugated to Alexa Fluor 488 or 546 (90 min, RT). After threewashes with PBS, cells were mounted on glass slides with 50%glycerol-PBS containing 1 µg/ml DAPI. All images were takenwith a confocal Olympus FV300 microscope maintaining equalparameters in all conditions analyzed.

Cell counting and statistical analysis

Thenumberof viable neuronswas estimated by immunostainingagainst β-tubulin or bright field imaging. All cells positive to β-tubulin were counted regardless of its appearance in at least75% of each whole area seeded. From light images, neuronscountedwere thosewith a clearmorphology and bearing cell pro-cesses with a length at least equal to the body size. All samplesunderwent same parallel procedures. Measurement of size andnumber of neurites was done by using the free ImageJ (NIH,USA) software. At least 150 neurons were analyzed per experi-mental condition.

Organic acid analysis

Astrocytes from WT and Gcdh−/− mice were cultivated in thepresence of 10 m Lys for 72 h, the culture media collected andorganic acid analysis was carried out according to Sweetmanet al. (43) with slight modifications (44). First, culture mediawere acidified to pH 1.5–2.0 and mixed with 100 µl hexadecane(internal standard). Ammonium chloride was then added andthe organic acids were extracted twice with ethylacetate andthe organic phases were pooled. Five hundred milligrams of so-dium sulfate was added to each preparation, the mixture stayedfor at least 60 min at RT, then passed through 0.2 µm filters andevaporated in N2 at 60°C. A 100 μl of ethanol was added, homoge-nized, centrifuged for 10 min and finally evaporated in N2.Derivatization to allow the analysis of organic acids as trimethyl-sylil compounds was performed by adding 27.5 µl of BSTFA(bis-(trimethylsylil) trifluoracetamide) and 1% TMCS (trimethyl-chlorosilane), and allowed to react (60 min, 60°C). Half microliterof each derivatized sample was then injected into Varian Saturn2000 GC/MS equipment with a CP-Sil 8 CB capillary column(length 30 m, internal diameter 0.25 mm, film 0.25 µm), an opensplit injector and helium as the carrier gas. The GC/MS tempera-tures were as follows: injector 250°C, column 90°C to 280°C withan increment of 3°C permin, transfer line 280°C, ion source 150°Cand mass analyzer 35°C. The total run time was 75 min. Finally,the mass spectrometer was programmed from m/z 10–650 atthe rate of 0.6 Hz.

Quantitative measurement of GA and 3HGA in culture me-dium and astrocyte homogenates was performed by gas chroma-tography–mass spectroscopy using stable-isotope dilution (6).Deuterium-labeled internal standards for GA (d4-GA, 0.05 mg/ml) and HGA (d5-HGA, 0.05 mg/ml) were added at 0.05 ml each

to 1.0 ml cultured medium or 0.3 ml cell homogenates. Sulfosa-licylic acid at 0.15 ml of 9.33% or 70 mgwas added to the samples.Organic acids were extracted twice with 3 ml diethyl ether and1.5 ml ethyl acetate. Combined organic phases were dried at 30°C under nitrogen and derivatized with 0.05 ml BTSFA/1% TMCS[N,O-Bis(trimethylsilyl)trifluoroacetamide with trimethylchloro-silane] for 20 min at 80°C. Injected volume of 0.001 ml was ana-lyzed using an Agilent Technologies 6890N Gas Chromatographequipped with a 5973N Mass Selective Detector. The mass spec-trometer monitored ions at 265/261 (with check of ratio at 237/233) for GA and 188/185 (check ratio at 262/259) for HGA in separ-ate runs.

Assessment of oxidative levels in living astrocytes

Carboxy-H2DCFDA (DCF) probe in astrocytesOxidative activity was measured in controls and Lys or GA-trea-tedWT and Gcdh−/− astrocytes with the cell-permeant carboxy-H2DCFDA probe (Invitrogen). According to the manufacturer’sinstructions, living cells were washed and incubated with 5 µcarboxy-H2DCFDA in 10 m PBS containing 20 m glucose (1 h,37°C). Then, 1 µg/ml DAPI was added, cells rinsed and each fluor-escence emission immediately imaged or measured after excita-tions of 405 and 488 nm in a Varioskan spectrophotometer,respectively. All data were expressed as percent of respectivecontrol values obtained for WT and Gcdh−/− astrocytes.

Malondialdehyde (TBA-RS) and reduced glutathione (GSH) concentra-tions in astrocyte homogenatesConfluent fresh astrocytemonolayers seeded on two 35 mmPetridishes were scraped in 20 m sodium phosphate buffer, pH 7.4,containing 140 m KCl, and then were centrifuged at 750 g(10 min, 4°C) to discard nuclei and cell debris (45). The pelletwas discarded and the supernatant, a suspension of preservedorganelles, including mitochondria, was used to measure lipidperoxidation and reduced glutathione concentrations. The pro-tein content was determined by the method of Lowry et al. (46),using BSA as the standard.

MDA levels were determined according Yagi (47) with slightmodifications. Briefly, 200 µl of 10% trichloroacetic acid and300 µl of 0.67% thiobarbituric acid in 7.1% sodium sulfate wereadded to 100 µl of cell supernatants and incubated for 2 h in aboiling water bath. The mixture was allowed to cool in runningtap water for 5 min. The resulting pink-stained complex was ex-tracted with 300 µl of butanol. Fluorescence of the organic phasewas read with 515 nm excitation and 553 nm emission wave-lengths.MDA levels, determined in triplicate for each experimen-tal condition, were calculated as nmol MDA/mg protein using acalibration curve determined with 1,1,3,3-tetramethoxypropane.

GSH concentrations were measured according to Browneand Armstrong (48) with minor modifications. Supernatants(0.3–0.5 mg of protein/mL) were first deproteinized with meta-phosphoric acid, centrifuged at 7000g for 10 min and immediate-ly used for GSH quantification. One hundred and eighty-fivemicroliters of 100 m sodium phosphate buffer, pH 8.0, contain-ing 5 m ethylenediaminetetraacetic acid, and 15 µl of o-phthal-dialdehyde (1 mg/mL) were added to 30 µl of supernatantpreviously deproteinized. This mixture was incubated at RT ina dark room for 15 min. Fluorescence was measured usingexcitation and emissionwavelengths of 350 and 420 nm, respect-ively. The calibration curve was prepared with standard GSH(0.001–1 m) and the concentrations, determined in triplicatefor each experimental condition, and referred as nmol GSH/mgprotein.

Human Molecular Genetics, 2015, Vol. 24, No. 16 | 4513

Downloaded from https://academic.oup.com/hmg/article-abstract/24/16/4504/744753by gueston 12 April 2018

Page 11: Striatal neuronal death mediated by astrocytes from the Gcdh ...

Western blotting assays

To assess the Nrf2/ARE dependent expression of HO-1 (23–25);confluent Gcdh−/− astrocyte cultures were pretreated with 1 µMEL or 20 µM tBHQ, then challenged to 10 m Lys. 24 h later, as-trocytes of all conditions were scrapped in cell lysis buffer, soni-cated and the protein concentration determined by thebicinchoninic acid procedure. Denatured samples were seededand a typical SDS-PAGE electrophoresis and blotting was per-formed (23). Proteins that were transferred to a PVDF membranewere incubated overnight with a 1:1000 dilution of anti-HO-1 orwith 1:4000 of anti-βactin antibody that was used as a proteinloading control. After 1 h of incubationwith HRP-conjugated sec-ondary antibodies, the PVDF membrane was incubated with acommercial ECL kit (Pierce, Rockford, IL, USA) and bands wereanalyzed with the Image J (NIH, USA) gel analyzer tool. Datawere referred as the percentage of βactin expression in each cor-responding sample.

Statistical analysis

All values shown are the mean ± SEM of at least 3–5 independentexperiments performed in triplicates. Data analysis was per-formed using standard statistical packages (SigmaStat 2.0 andOrigin 8.1). To determine statistical difference among groupswe use a two-factor analysis of variance (ANOVA). P < 0.05 wasconsidered statistically significant, and P < 0.1 marginallysignificant.

Supplementary MaterialSupplementary Material is available at HMG online.

Conflict of Interest statement. None declared.

FundingThis work was supported by Conselho Nacional de Desenvolvi-mento Científico e Tecnológico – CNPQ (grant # 490297/2011-0,Edital Uruguai - CNPq/DICYT; grant # 470236/2012-4, Edital Uni-versal 14/2012); Fundação de Apoio à Pesquisa do Estado do RioGrande do Sul – FAPERGS; FOCEM (MERCOSUR Structural Conver-gence Fund), COF 03/11; and the colaboration of the Brazilian-Uruguay Bilateral Cooperation Program (DICYT-URUGUAY),PEDECIBA Biología and National Agency for Research and Innov-ation (ANII), FMV 3_201117213, FCE _1_2011_1_7342.

References1. Goodman, S. and Frerman, F. (2001) Organic acidemias due to

defects in lysine oxidation: 2-ketoadipic acidemia and gluta-ric acidemia. In Scriver, C.R., Beaudet, A.L., Sly,W.S. andValle,D. (eds), The Metabolic and Molecular Basis of Disease. McGraw-Hill, pp. 2195–2204.

2. Kolker, S., Ahlemeyer, B., Krieglstein, J. and Hoffmann, G.F.(2000) Maturation-dependent neurotoxicity of 3-hydroxyglu-taric and glutaric acids in vitro: a new pathophysiologic ap-proach to glutaryl-CoA dehydrogenase deficiency. Pediatr.Res., 47, 495–503.

3. Strauss, K.A. and Morton, D.H. (2003) Type I glutaric aciduria,part 2: a model of acute striatal necrosis. Am. J. Med. Genet. CSemin. Med. Genet., 121C, 53–70.

4. Funk, C.B., Prasad, A.N., Frosk, P., Sauer, S., Kolker, S., Green-berg, C.R. and Del Bigio, M.R. (2005) Neuropathological,

biochemical and molecular findings in a glutaric acidemiatype 1 cohort. Brain, 128, 711–722.

5. Hoffmann, G.F., Athanassopoulos, S., Burlina, A.B., Duran,M., de Klerk, J.B., Lehnert, W., Leonard, J.V., Monavari, A.A.,Muller, E., Muntau, A.C. et al. (1996) Clinical course, earlydiagnosis, treatment, and prevention of disease in glutar-yl-CoA dehydrogenase deficiency. Neuropediatrics, 27,115–123.

6. Zinnanti, W.J., Lazovic, J., Housman, C., LaNoue, K., O’Calla-ghan, J.P., Simpson, I., Woontner, M., Goodman, S.I., Connor,J.R., Jacobs, R.E. et al. (2007)Mechanismof age-dependent sus-ceptibility and novel treatment strategy in glutaric acidemiatype I. J. Clin. Invest., 117, 3258–3270.

7. Koeller, D.M.,Woontner,M., Crnic, L.S., Kleinschmidt-DeMas-ters, B., Stephens, J., Hunt, E.L. and Goodman, S.I. (2002) Bio-chemical, pathologic and behavioral analysis of a mousemodel of glutaric acidemia type I. Hum. Mol. Genet., 11, 347–357.

8. Koeller, D.M., Sauer, S., Wajner, M., de Mello, C.F., Goodman,S.I., Woontner, M., Muhlhausen, C., Okun, J.G. and Kolker, S.(2004) Animal models for glutaryl-CoA dehydrogenase defi-ciency. J. Inherit. Metab. Dis., 27, 813–818.

9. Zinnanti, W.J., Lazovic, J., Wolpert, E.B., Antonetti, D.A.,Smith, M.B., Connor, J.R., Woontner, M., Goodman, S.I. andCheng, K.C. (2006) A diet-induced mouse model for glutaricaciduria type I. Brain, 129, 899–910.

10. Kolker, S., Ahlemeyer, B., Krieglstein, J. and Hoffmann, G.F.(1999) 3-Hydroxyglutaric and glutaric acids are neurotoxicthrough NMDA receptors in vitro. J. Inherit. Metab. Dis., 22,259–262.

11. Freudenberg, F., Lukacs, Z. and Ullrich, K. (2004) 3-Hydroxy-glutaric acid fails to affect the viability of primary neuronalrat cells. Neurobiol. Dis., 16, 581–584.

12. Lund, T.M., Christensen, E., Kristensen, A.S., Schousboe, A.and Lund, A.M. (2004) On the neurotoxicity of glutaric, 3-hy-droxyglutaric, and trans-glutaconic acids in glutaric acide-mia type 1. J. Neurosci. Res., 77, 143–147.

13. Olivera-Bravo, S., Fernandez, A., Sarlabos, M.N., Rosillo, J.C.,Casanova, G., Jimenez, M. and Barbeito, L. (2011) Neonatalastrocyte damage is sufficient to trigger progressive striataldegeneration in a rat model of glutaric acidemia-I. PLoSONE, 6, e20831.

14. Lamp, J., Keyser, B., Koeller, D.M., Ullrich, K., Braulke, T. andMuhlhausen, C. (2011) Glutaric aciduria type 1 metabolitesimpair the succinate transport from astrocytic to neuronalcells. J. Biol. Chem., 286, 17777–17784.

15. Olivera, S., Fernandez, A., Latini, A., Rosillo, J.C., Casanova, G.,Wajner, M., Cassina, P. and Barbeito, L. (2008) Astrocyticproliferation and mitochondrial dysfunction inducedby accumulated glutaric acidemia I (GAI) metabolites:possible implications for GAI pathogenesis. Neurobiol. Dis.,32, 528–534.

16. Olivera-Bravo, S., Isasi, E., Fernandez, A., Rosillo, J.C., Jimenez,M., Casanova, G., Sarlabos, M.N. and Barbeito, L. (2014) Whitematter injury induced by perinatal exposure to glutaric Acid.Neurotox. Res., 25, 381–391.

17. Jafari, P., Braissant, O., Zavadakova, P., Henry, H., Bonafe, L.and Ballhausen, D. (2013) Ammonium accumulation andcell death in a rat 3D brain cell model of glutaric aciduriatype I. PLoS ONE, 8, e53735.

18. Isasi, E., Barbeito, L. and Olivera-Bravo, S. (2014) Increasedblood-brain barrier permeability and alterations in perivas-cular astrocytes and pericytes induced by intracisternal glu-taric acid. Fluid Barrier CNS, 11, 15.

4514 | Human Molecular Genetics, 2015, Vol. 24, No. 16

Downloaded from https://academic.oup.com/hmg/article-abstract/24/16/4504/744753by gueston 12 April 2018

Page 12: Striatal neuronal death mediated by astrocytes from the Gcdh ...

19. Maragakis, N.J. and Rothstein, J.D. (2006) Mechanisms of dis-ease: astrocytes in neurodegenerative disease.Nat. Clin. Pract.Neurol., 2, 679–689.

20. Verkhratsky, A., Sofroniew, M.V., Messing, A., de Lanerolle, N.C., Rempe, D., Rodriguez, J.J. andNedergaard,M. (2012) Neuro-logical diseases as primary gliopathies: a reassessment ofneurocentrism. ASN Neuro, 4:e00082.

21. Sofroniew, M.V. and Vinters, H.V. (2010) Astrocytes: biologyand pathology. Acta Neuropathol, 119, 7–35.

22. Wang, Z., Liu, D., Wang, J., Liu, S., Gao, M., Ling, E.A. and Hao,A. (2012) Cytoprotective effects of melatonin on astroglialcells subjected to palmitic acid treatment in vitro. J. PinealRes., 52, 253–264.

23. Kwon, K.J., Kim, J.N., Kim, M.K., Lee, J., Ignarro, L.J., Kim, H.J.,Shin, C.Y. and Han, S.H. (2011) Melatonin synergistically in-creases resveratrol-induced heme oxygenase-1 expressionthrough the inhibition of ubiquitin-dependent proteasomepathway: a possible role in neuroprotection. J. Pineal Res., 50,110–123.

24. Chow, H.S., Lynch, J.J. III, Rose, K. and Choi, D.W. (1994) Troloxattenuates cortical neuronal injury induced by iron, ultravioletlight, glucose deprivation, or AMPA. Brain Res., 639, 102–108.

25. Vargas, M.R., Pehar, M., Cassina, P., Martinez-Palma, L.,Thompson, J.A., Beckman, J.S. and Barbeito, L. (2005) Fibro-blast growth factor-1 induces heme oxygenase-1 via nuclearfactor erythroid 2-related factor 2 (Nrf2) in spinal cord astro-cytes: consequences for motor neuron survival. J. Biol. Chem.,280, 25571–25579.

26. Dowell, J.A. and Johnson, J.A. (2013) Mechanisms of Nrf2 pro-tection in astrocytes as identified by quantitative proteomicsand siRNA screening. PLoS ONE, 8, e70163.

27. Lee, J.M., Calkins, M.J., Chan, K., Kan, Y.W. and Johnson, J.A.(2003) Identification of the NF-E2-related factor-2-dependentgenes conferring protection against oxidative stress in pri-mary cortical astrocytes using oligonucleotide microarrayanalysis. J. Biol. Chem., 278, 12029–12038.

28. Seminotti, B., Fernandes, C.G., Leipnitz, G., Amaral, A.U., Za-natta, A. and Wajner, M. (2011) Neurochemical evidence thatlysine inhibits synaptic Na+,K+-ATPase activity and provokesoxidative damage in striatum of young rats in vivo. Neuro-chem. Res., 36, 205–214.

29. Seminotti, B., Leipnitz, G., Amaral, A.U., Fernandes, C.G., daSilva Lde, B., Tonin, A.M., Vargas, C.R. and Wajner, M. (2008)Lysine induces lipid and protein damage and decreases re-duced glutathione concentrations in brain of young rats.Int. J. Dev. Neurosci., 26, 693–698.

30. Lobsiger, C.S. and Cleveland, D.W. (2007) Glial cells as intrin-sic components of non-cell-autonomous neurodegenerativedisease. Nat. Neurosci., 10, 1355–1360.

31. Ballas, N., Lioy, D.T., Grunseich, C. and Mandel, G. (2009)Non-cell autonomous influence of MeCP2-deficient glia onneuronal dendritic morphology. Nat. Neurosci., 12, 311–317.

32. Messing,A., Brenner,M., Feany,M.B., Nedergaard,M. andGold-man, J.E. (2012) Alexander disease. J. Neurosci., 32, 5017–5023.

33. Diaz-Amarilla, P., Olivera-Bravo, S., Trias, E., Cragnolini, A.,Martinez-Palma, L., Cassina, P., Beckman, J. and Barbeito, L.(2011) Phenotypically aberrant astrocytes that promotemotoneuron damage in a model of inherited amyotrophiclateral sclerosis. Proc. Natl Acad. Sci. USA, 108, 18126–18131.

34. Soffer, D., Amir, N., Elpeleg, O.N., Gomori, J.M., Shalev, R.S.and Gottschalk-Sabag, S. (1992) Striatal degeneration andspongy myelinopathy in glutaric acidemia. J. Neurol. Sci.,107, 199–204.

35. Porciuncula, L.O., Emanuelli, T., Tavares, R.G., Schwarzbold,C., Frizzo, M.E., Souza, D.O. and Wajner, M. (2004) Glutaricacid stimulates glutamate binding and astrocytic uptakeand inhibits vesicular glutamate uptake in forebrain fromyoung rats. Neurochem. Int., 45, 1075–1086.

36. Rosa, R.B., Dalcin, K.B., Schmidt, A.L., Gerhardt, D., Ribeiro, C.A., Ferreira, G.C., Schuck, P.F., Wyse, A.T., Porciuncula, L.O.,Wofchuk, S. et al. (2007) Evidence that glutaric acid reducesglutamate uptake by cerebral cortex of infant rats. Life Sci.,81, 1668–1676.

37. Wajner, M., Kolker, S., Souza, D.O., Hoffmann, G.F. and deMello, C.F. (2004) Modulation of glutamatergic and GABAergicneurotransmission in glutaryl-CoA dehydrogenase defi-ciency. J. Inherit. Metab. Dis., 27, 825–828.

38. Bain, J.M., Ziegler, A., Yang, Z., Levison, S.W. and Sen, E. (2010)TGFbeta1 stimulates the over-production of white matter as-trocytes from precursors of the “brain marrow” in a rodentmodel of neonatal encephalopathy. PLoS ONE, 5, e9567.

39. Kulkens, S., Harting, I., Sauer, S., Zschocke, J., Hoffmann, G.F.,Gruber, S., Bodamer, O.A. and Kolker, S. (2005) Late-onsetneurologic disease in glutaryl-CoA dehydrogenase defi-ciency. Neurology, 64, 2142–2144.

40. Gerstner, B., Gratopp, A., Marcinkowski, M., Sifringer, M., Ob-laden, M. and Buhrer, C. (2005) Glutaric acid and its metabo-lites cause apoptosis in immature oligodendrocytes: a novelmechanism of white matter degeneration in glutaryl-CoAdehydrogenase deficiency. Pediatr. Res., 57, 771–776.

41. Harting, I., Neumaier-Probst, E., Seitz, A., Maier, E.M., Ass-mann, B., Baric, I., Troncoso, M., Muhlhausen, C., Zschocke,J., Boy, N.P. et al. (2009) Dynamic changes of striatal and extra-striatal abnormalities in glutaric aciduria type I. Brain, 132,1764–1782.

42. Ventimiglia, R. and Kindsay, R.M. (1998) Rat striatal neuronsin low-density, serum-free culture. In Goslin, G. and Ba, K.(eds), Culturing Nerve Cells. MIT Press, in press, pp. 371–393.

43. Sweetman, L. (1991) Organic acid analysis. In Hommes, F.A.(eds), Techniques in Diagnostic Human Biochemical Genetics: ALaboratory Manual. Wiley-Liss, New York, pp. 143.

44. Wajner, M., Coelho Dde, M., Ingrassia, R., de Oliveira, A.B., Bu-sanello, E.N., Raymond, K., Flores Pires, R., de Souza, C.F., Giu-gliani, R. and Vargas, C.R. (2009) Selective screening fororganic acidemias by urine organic acid GC-MS analysis inBrazil: fifteen-year experience. Clin. Chim. Acta, 400, 77–81.

45. Evelson, P., Travacio, M., Repetto, M., Escobar, J., Llesuy, S. andLissi, E.A. (2001) Evaluation of total reactive antioxidant po-tential (TRAP) of tissue homogenates and their cytosols.Arch. Biochem. Biophys., 388, 261–266.

46. Lowry, O.H., Rosebrough,N.J., Farr, A.L. andRandall, R.J. (1951)Protein measurement with the Folin phenol reagent. J. Biol.Chem., 193, 265–275.

47. Yagi, K. (1998) Simple procedure for specific assay of lipidhydroperoxides in serum or plasma. Methods Mol. Biol., 108,107–110.

48. Browne, R.W. and Armstrong, D. (1998) Reduced glutathioneand glutathione disulfide. Methods Mol. Biol., 108, 347–352.

Human Molecular Genetics, 2015, Vol. 24, No. 16 | 4515

Downloaded from https://academic.oup.com/hmg/article-abstract/24/16/4504/744753by gueston 12 April 2018