6
The inflammatory NADPH oxidase enzyme modulates motor neuron degeneration in amyotrophic lateral sclerosis mice Du-Chu Wu* , Diane Be ´ range ` re Re ´* , Makiko Nagai* , Harry Ischiropoulos ‡§ , and Serge Przedborski* †¶ Departments of *Neurology and Pathology and Cell Biology and Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032; Stokes Research Institute, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104; and § Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104 Edited by L. L. Iversen, University of Oxford, Oxford, United Kingdom, and approved June 19, 2006 (received for review May 5, 2006) ALS is a fatal paralytic disorder characterized by a progressive loss of spinal cord motor neurons. Herein, we show that NADPH oxidase, the main reactive oxygen species-producing enzyme dur- ing inflammation, is activated in spinal cords of ALS patients and in spinal cords in a genetic animal model of this disease. We demonstrate that inactivation of NADPH oxidase in ALS mice delays neurodegeneration and extends survival. We also show that NADPH oxidase-derived oxidant products damage proteins such as insulin-like growth factor 1 (IGF1) receptors, which are located on motor neurons. Our in vivo and in vitro data indicate that such an oxidative modification hinders the IGF1Akt survival pathway in motor neurons. These findings suggest a non-cell-autonomous mechanism through which inflammation could hasten motor neu- ron death and contribute to the selective motor neuronal degen- eration in ALS. Akt ALS microglia oxidation non-cell autonomous A LS is the most common adult-onset paralytic disease and is characterized by a loss of motor neurons in the cerebral cortex, brainstem, and spinal cord (1). It is invariably fatal, usually within 3–5 years after onset (1). Insights into its neurodegenerative mechanisms followed the discovery that dominant mutations in the gene for superoxide dismutase 1 (SOD1) cause familial ALS (2, 3). Overexpression of SOD1 mutants in rodents emulate clinical and pathological hallmarks of ALS through a toxic gain of function (4). Development of chimeric mice provided animals with a mixture of neuronal and nonneuronal cells expressing wild-type or mutant SOD1 (5); investigation of these animals suggested that nonneu- ronal cells influence the fate of spinal cord motor neurons (5). Corroborating this hypothesis is the demonstration that reduction of mutant SOD1, selectively in microglia, extended survival in transgenic SOD1 G37R mice (6). In light of the latter results, micro- glia are now thought to contribute to the non-cell-autonomous killing of motor neurons. Among the microglia-derived mediators that could promote neurodegeneration are reactive oxygen species (ROS) produced by the enzyme NADPH oxidase complex (7). Although this ROS- generating multimeric oxidase is indispensable for protecting the host against invading microorganisms in infectious disorders (8), its inappropriate activation may be harmful in noninfectious neuro- degenerative disorders. Such bystander cytotoxicity is thought to lead to the death of developing oligodendrocytes in periventricular leukomalacia (9), one of the most important causes of cerebral palsy. In light of these facts, we undertook the study of NADPH oxidase in both human ALS and one of its genetic models. Our results for both human and mouse postmortem tissues indicate that spinal cord microgliosis in ALS is accompanied with an up- regulation of NADPH oxidase. Furthermore, by using mutant deficient mice in functional NADPH oxidase as well as in neuron- like cell culture systems, we provide compelling evidence that supports the concept that this microglial ROS-generating enzy- matic complex promotes spinal cord motor neuron degeneration by a mechanism involving oxidative damage to critical survival signal- ing pathways. Results NADPH Oxidase Is Up-Regulated in Inflamed Spinal Cords of ALS Mice. To determine the role of NADPH oxidase in motor neuron degeneration, we first evaluated its expression at different stages of the disease in transgenic mice expressing mutant human SOD1 with a substitution of glycine to alanine in position 93 (SOD1 G93A ), the most widely studied model of ALS. Expression of NADPH oxidase in the spinal cord, which carries the brunt of the pathology in this ALS model, was determined by analyzing its catalytic subunit, gp91 phox . Both gp91 phox message and protein contents in whole- tissue extracts of spinal cord rose over time in transgenic SOD1 G93A mice (Fig. 1 A, B, D, and E) in concert with the development of a glial response (Fig. 6, which is published as supporting information on the PNAS web site) and the progression of the paralytic phenotype. The levels of the p67 phox subunit that contains the NADPH-binding site of the NADPH oxidase complex (10) were increased in membrane fractions of spinal cord extracts from symptomatic transgenic SOD1 G93A mice (Fig. 1C), indicating that this cytosolic subunit did translocate to the plasma membrane in these animals. Conversely, none of these NADPH oxidase alter- ations were seen in age-matched nontransgenic littermates (Fig. 1 AE). Histological evaluation of the spinal cord of symptomatic transgenic SOD1 G93A mice showed numerous gp91 phox -positive cells, primarily in the gray matter of the anterior horn (Fig. 1G), whereas sparse staining was observed in the spinal cord of age- matched nontransgenic controls (Fig. 1 F). Consistent with NADPH oxidase expression by professional phagocytes, confocal microscopy demonstrated the colocalization of the gp91 phox subunit with a microglial marker, the ricinus communis agglutinin lectin (Fig. 1 HJ); no gp91 phox colocalization was detected with the motor neuron marker, nonphosphorylated neurofilament heavy chain, or with the astrocyte marker, glial fibrillary acid protein (data not shown). NADPH Oxidase Causes Protein Oxidation in Transgenic SOD1 G93A Mice. We further characterized the status of spinal cord NADPH oxidase in transgenic SOD1 G93A mice by probing for formation of ROS and evidence of protein oxidation. In nontransgenic mice, spinal cord production of ROS, evidenced by the fluorescence emitted by ethidium, the oxidation product of hydroethidine (11), was minimal (Fig. 1K). In contrast, in symptomatic transgenic Conflict of interest statement: No conflicts declared. This paper was submitted directly (Track II) to the PNAS office. Abbreviations: ROS, reactive oxygen species; SOD1, superoxide dismutase 1; IGF1, insulin- like growth factor 1. To whom correspondence should be addressed at: Departments of Neurology and Pathol- ogy, Columbia University, BB-302, 650 West 168th Street, New York, NY 10032. E-mail: [email protected]. © 2006 by The National Academy of Sciences of the USA 12132–12137 PNAS August 8, 2006 vol. 103 no. 32 www.pnas.orgcgidoi10.1073pnas.0603670103 Downloaded by guest on January 20, 2021

The inflammatory NADPH oxidase enzyme modulates motor ...The inflammatory NADPH oxidase enzyme modulates motor neuron degeneration in amyotrophic lateral sclerosis mice Du-Chu Wu*†,

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: The inflammatory NADPH oxidase enzyme modulates motor ...The inflammatory NADPH oxidase enzyme modulates motor neuron degeneration in amyotrophic lateral sclerosis mice Du-Chu Wu*†,

The inflammatory NADPH oxidase enzyme modulatesmotor neuron degeneration in amyotrophic lateralsclerosis miceDu-Chu Wu*†, Diane Berangere Re*†, Makiko Nagai*†, Harry Ischiropoulos‡§, and Serge Przedborski*†¶�

Departments of *Neurology and ¶Pathology and Cell Biology and †Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032;‡Stokes Research Institute, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104; and §Department of Pharmacology,University of Pennsylvania School of Medicine, Philadelphia, PA 19104

Edited by L. L. Iversen, University of Oxford, Oxford, United Kingdom, and approved June 19, 2006 (received for review May 5, 2006)

ALS is a fatal paralytic disorder characterized by a progressive lossof spinal cord motor neurons. Herein, we show that NADPHoxidase, the main reactive oxygen species-producing enzyme dur-ing inflammation, is activated in spinal cords of ALS patients andin spinal cords in a genetic animal model of this disease. Wedemonstrate that inactivation of NADPH oxidase in ALS micedelays neurodegeneration and extends survival. We also show thatNADPH oxidase-derived oxidant products damage proteins such asinsulin-like growth factor 1 (IGF1) receptors, which are located onmotor neurons. Our in vivo and in vitro data indicate that such anoxidative modification hinders the IGF1�Akt survival pathway inmotor neurons. These findings suggest a non-cell-autonomousmechanism through which inflammation could hasten motor neu-ron death and contribute to the selective motor neuronal degen-eration in ALS.

Akt � ALS � microglia � oxidation � non-cell autonomous

ALS is the most common adult-onset paralytic disease and ischaracterized by a loss of motor neurons in the cerebral cortex,

brainstem, and spinal cord (1). It is invariably fatal, usually within3–5 years after onset (1). Insights into its neurodegenerativemechanisms followed the discovery that dominant mutations in thegene for superoxide dismutase 1 (SOD1) cause familial ALS (2, 3).Overexpression of SOD1 mutants in rodents emulate clinical andpathological hallmarks of ALS through a toxic gain of function (4).Development of chimeric mice provided animals with a mixture ofneuronal and nonneuronal cells expressing wild-type or mutantSOD1 (5); investigation of these animals suggested that nonneu-ronal cells influence the fate of spinal cord motor neurons (5).Corroborating this hypothesis is the demonstration that reductionof mutant SOD1, selectively in microglia, extended survival intransgenic SOD1G37R mice (6). In light of the latter results, micro-glia are now thought to contribute to the non-cell-autonomouskilling of motor neurons.

Among the microglia-derived mediators that could promoteneurodegeneration are reactive oxygen species (ROS) produced bythe enzyme NADPH oxidase complex (7). Although this ROS-generating multimeric oxidase is indispensable for protecting thehost against invading microorganisms in infectious disorders (8), itsinappropriate activation may be harmful in noninfectious neuro-degenerative disorders. Such bystander cytotoxicity is thought tolead to the death of developing oligodendrocytes in periventricularleukomalacia (9), one of the most important causes of cerebralpalsy. In light of these facts, we undertook the study of NADPHoxidase in both human ALS and one of its genetic models. Ourresults for both human and mouse postmortem tissues indicate thatspinal cord microgliosis in ALS is accompanied with an up-regulation of NADPH oxidase. Furthermore, by using mutantdeficient mice in functional NADPH oxidase as well as in neuron-like cell culture systems, we provide compelling evidence thatsupports the concept that this microglial ROS-generating enzy-matic complex promotes spinal cord motor neuron degeneration by

a mechanism involving oxidative damage to critical survival signal-ing pathways.

ResultsNADPH Oxidase Is Up-Regulated in Inflamed Spinal Cords of ALS Mice.To determine the role of NADPH oxidase in motor neurondegeneration, we first evaluated its expression at different stages ofthe disease in transgenic mice expressing mutant human SOD1 witha substitution of glycine to alanine in position 93 (SOD1G93A), themost widely studied model of ALS. Expression of NADPH oxidasein the spinal cord, which carries the brunt of the pathology in thisALS model, was determined by analyzing its catalytic subunit,gp91phox. Both gp91phox message and protein contents in whole-tissue extracts of spinal cord rose over time in transgenic SOD1G93A

mice (Fig. 1 A, B, D, and E) in concert with the development of aglial response (Fig. 6, which is published as supporting informationon the PNAS web site) and the progression of the paralyticphenotype. The levels of the p67phox subunit that contains theNADPH-binding site of the NADPH oxidase complex (10) wereincreased in membrane fractions of spinal cord extracts fromsymptomatic transgenic SOD1G93A mice (Fig. 1C), indicating thatthis cytosolic subunit did translocate to the plasma membrane inthese animals. Conversely, none of these NADPH oxidase alter-ations were seen in age-matched nontransgenic littermates (Fig. 1A–E). Histological evaluation of the spinal cord of symptomatictransgenic SOD1G93A mice showed numerous gp91phox-positivecells, primarily in the gray matter of the anterior horn (Fig. 1G),whereas sparse staining was observed in the spinal cord of age-matched nontransgenic controls (Fig. 1F). Consistent withNADPH oxidase expression by professional phagocytes, confocalmicroscopy demonstrated the colocalization of the gp91phox subunitwith a microglial marker, the ricinus communis agglutinin lectin(Fig. 1 H–J); no gp91phox colocalization was detected with the motorneuron marker, nonphosphorylated neurofilament heavy chain, orwith the astrocyte marker, glial fibrillary acid protein (data notshown).

NADPH Oxidase Causes Protein Oxidation in Transgenic SOD1G93A

Mice. We further characterized the status of spinal cord NADPHoxidase in transgenic SOD1G93A mice by probing for formation ofROS and evidence of protein oxidation. In nontransgenic mice,spinal cord production of ROS, evidenced by the fluorescenceemitted by ethidium, the oxidation product of hydroethidine (11),was minimal (Fig. 1K). In contrast, in symptomatic transgenic

Conflict of interest statement: No conflicts declared.

This paper was submitted directly (Track II) to the PNAS office.

Abbreviations: ROS, reactive oxygen species; SOD1, superoxide dismutase 1; IGF1, insulin-like growth factor 1.

�To whom correspondence should be addressed at: Departments of Neurology and Pathol-ogy, Columbia University, BB-302, 650 West 168th Street, New York, NY 10032. E-mail:[email protected].

© 2006 by The National Academy of Sciences of the USA

12132–12137 � PNAS � August 8, 2006 � vol. 103 � no. 32 www.pnas.org�cgi�doi�10.1073�pnas.0603670103

Dow

nloa

ded

by g

uest

on

Janu

ary

20, 2

021

Page 2: The inflammatory NADPH oxidase enzyme modulates motor ...The inflammatory NADPH oxidase enzyme modulates motor neuron degeneration in amyotrophic lateral sclerosis mice Du-Chu Wu*†,

SOD1G93A mice carrying the wild-type gp91phox allele (SODG93A�gp91phox�), spinal cord ethidium fluorescence was intense (Fig. 1L)and coincided anatomically with the areas of gp91phox expression(Fig. 1G) and microglial activation (Fig. 6). In symptomatic trans-genic SOD1G93A mice carrying the nonfunctional mutant allele(SODG93A�gp91phox�) (12), spinal cord ethidium fluorescence wasnegligible (Fig. 1M), as it was in the nontransgenic controls (Fig.1K). A similar genotypic difference was found for the analysis ofprotein carbonylation (13), which is used as a general marker ofprotein oxidation (14). Symptomatic transgenic SOD1G93A�gp91phox� mice, but not age-matched SOD1G93A�gp91phox� mice,had increased levels of spinal cord protein carbonyl adducts com-pared with nontransgenic controls expressing either wild-type ornull mutant gp91phox (Fig. 1N). Immunohistochemically, the mostrobust labeling for protein carbonyl adducts occurred in spinal cordsections from SOD1G93A�gp91phox� mice at the level of cells withmixed morphology, including large motor neurons (Fig. 1 O–Q).

NADPH Oxidase Induction and Neuronal Protein Carbonylation inSporadic ALS. We then sought to determine whether the NADPHoxidase alterations identified in transgenic SOD1G93A mice werealso present in human sporadic ALS, the most common form ofthe disease (1). Consistent with the mouse data, gp91phox contentwas low (Fig. 2 A and B) and its immunoreactivity was faint incontrol postmortem spinal cords (Fig. 2D), whereas gp91phox

content was �3-fold higher and its immunoreactivity robust insporadic ALS spinal cords (Fig. 2E). In the latter, gp91phox-positive cells colocalized with the microglial-associated antigenCD68 (Fig. 2F) and were identified in all of the typical ALS lociof neurodegeneration, including the anterior horn and the lateralcorticospinal track (Fig. 2C). Also comparable with the mousedata, there was a robust labeling for protein carbonyl adducts inpostmortem spinal cord sections from sporadic ALS cases, whichseemed to be mainly associated with large motor neurons (Fig.2H). On average, 6–12 of 30 counted motor neurons were

positive for protein carbonyl adducts per lumbar spinal cordsection in ALS patients, whereas no such immunoreactive motorneurons were seen in controls (Fig. 2G).

Fig. 2. NADPH oxidase is up-regulated and associated with motor neuroncarbonylation in the spinal cord of sporadic ALS patients. (A and B) Immuno-blots and bar graph for gp91phox using spinal cord extracts from six normalcontrols and six age-matched ALS patients. Values in B are means � SEM andwere compared by Student’s t test. *, P � 0.05, higher than normal controls.(C) Drawing of a spinal cord transversal plan showing the gray matter (areadelineated by the green dashed line) and the loci of degeneration in ALS(areas delineated by the red dashed line). (D and E) Spinal cord gp91phox

immunohistochemistry in tissue sections from controls (D) and ALS patients(E). (F) In ALS patient samples, gp91phox-positive cells (arrow) exhibit a brownmembrane labeling, which colocalizes with the blue-gray cytosol labeling forthe microglial marker CD68 (arrowhead). (G and H) Immunohistochemicaldetection of carbonyl adducts in spinal cord sections of normal controls andage-matched ALS patients obtained by using the same technique as in Fig. 1O–Q. (Scale bar in H: D, E, G, and H, 0.2 mm; F, 0.5 mm.)

Fig. 1. Microglial NADPH oxidasestimulates carbonylation of spinalcord motor neurons in transgenicSOD1G93A mice. (A–E ) Spinal cordgp91phox mRNA (A and D) and pro-tein (B and E) in 1-month-old(asymptomatic) to 4-month-old(end-stage) transgenic SOD1G93A

(TG) and nontransgenic (NTG) mice.(C) Spinal cord p67phox protein in4-month-old TG SOD1G93A andNTG littermates. �, positive control(mouse macrophage lysate). (F andG) Spinal cord gp91phox immunohis-tochemistry in NTG (F ) and TG (G)mice. (H–J) Confocal analysis of aTG spinal cord showing gp91phox ingreen (H) and the microglial markerricinus communis agglutinin lectinin red (I); merged image is shownin J. (K–M) Ethidium fluorescence (inred) in nontransgenic�gp91phox�

(NTG�gp91�) (K), transgenicSOD1G93A�gp91phox� (TG�gp91�)(L), and transgenic SOD1G93A�gp91phox� (TG�gp91�) (M) spinalcords. (N)HPLCquantificationofpro-tein carbonyls in spinal cords from allfour genotypes. (O–Q) Immunohistochemistry for carbonyl adducts in NTG�gp91� (O), TG�gp91� (P), and TG�gp91� (Q) spinal cords by using an anti-dinitrophenylhydrazine antibody. (P Inset) Carbonylated motor neuron. Studies in F–Q are in 4-month-old mice. Values (means � SEM; n � 4–8 mice per group) werecompared by ANOVA followed by Newman–Keuls post hoc testing. *, P � 0.05, more than NTG mice; #, P � 0.05, lower than TG�gp91� mice. (Scale bar in Q: F andG, 0.4 mm; H–J, 40 �m; K–M, 0.5 mm; O–Q, 0.15 mm.)

Wu et al. PNAS � August 8, 2006 � vol. 103 � no. 32 � 12133

NEU

ROSC

IEN

CE

Dow

nloa

ded

by g

uest

on

Janu

ary

20, 2

021

Page 3: The inflammatory NADPH oxidase enzyme modulates motor ...The inflammatory NADPH oxidase enzyme modulates motor neuron degeneration in amyotrophic lateral sclerosis mice Du-Chu Wu*†,

Deletion of gp91phox Mitigates the Disease Phenotype in TransgenicSOD1G93A Mice. Next, we explored the contribution of NADPHoxidase activation on the disease phenotype in the SOD1G93A

mouse model of ALS. Transgenic SOD1G93A�gp91phox� micereached end-stage paralysis (defined as a loss of the righting reflex)later than their transgenic SOD1G93A�gp91phox� counterparts (Fig.3A), which resulted in a longer lifespan of transgenic SOD1G93A�gp91phox� mice (log-rank test � 15.3; P � 0.001). In addition to theprolonged survival, inactivation of NADPH oxidase did mitigateneurodegeneration. Compared with end-stage transgenicSOD1G93A�gp91phox� mice, age-matched transgenic SOD1G93A�gp91phox� mice had �50% more anterior horn motor neurons in thespinal cord (Fig. 3 B–E) and myelinated axons in the fifth lumbaranterior roots (Fig. 3 F–L) as well as significantly more innervatedendplates (Fig. 3 M–P) and larger muscle fibers in the fibularis andperoneus longus muscles (Fig. 3 Q–T). Spinal cord microgliosis,evidenced by macrophage antigen complex 1 immunostaining, andlevels of the glial cytokine IL-1� did not differ between age-matched transgenic SOD1G93A�gp91phox� mice and SOD1G93A�gp91phox� mice (Fig. 7, which is published as supporting informationon the PNAS web site). Also not affected by the deficit of gp91phox

were the levels of human SOD1 in transgenic SOD1G93A mice (Fig.7) or the size of muscle fibers in the fibularis and peroneus longusmuscles in nontransgenic mice (Fig. 3T). The selected muscles aremainly composed of fast-twitch fibers, and by immunostaining formyosin heavy chain, we did not observe any obvious alteration inthe makeup of muscle fiber types among the different mouse groups(Fig. 7).

NADPH Oxidase Impairs the Insulin-Like Growth Factor 1 (IGF1)�AktPathway in Transgenic SOD1G93A Mice. We then explored whetherNADPH oxidase-mediated protein modifications might promoteneurodegeneration in ALS by damaging essential surviving path-

ways for motor neurons such as IGF1. After IGF1 was immuno-precipitated from spinal cord extracts, it was probed for evidenceof carbonylation. This approach failed to reveal evidence of IGF1oxidation in any of the studied mouse genotypes (data not shown).However, protein carbonyl adducts were evident in the �-chain ofthe IGF1 tyrosine kinase cognate receptor in the spinal cord ofsymptomatic transgenic SOD1G93A�gp91phox� mice (Fig. 4 A andB); similar results were obtained for the �-chain of IGF1 receptor(data not shown). This finding might be quite significant becauseIGF1 receptors in mouse spinal cords were detected almost exclu-sively on motor neurons (Fig. 8, which is published as supportinginformation on the PNAS web site). Contrasting with the IGF1receptor findings, oxidation indices in the intracellular serine�threonine kinase Akt, which transduces IGF1 receptor signaling(15), did not differ between symptomatic transgenic SOD1G93A

mice and their nontransgenic littermates. These results suggest thatthe entire IGF1 molecular pathway is not oxidatively modified byinflammation in this ALS model.

Next, we compared selected IGF1 transduction events among thedifferent mouse groups. Although mutant SOD1 is expressed in allcells, markers of IGF1 transduction such as phospho-IGF1 recep-tor, phospho-Akt (data not shown), and phospho-BAD were iden-tified by immunocytochemistry mainly in large cells with a motorneuron-like appearance and to a much lesser extent in smallerglia-like cells in spinal cord sections of symptomatic transgenicSOD1G93A mice. However, there were fewer phospho-IGF1 recep-tor-immunoreactive cells in spinal cord sections from symptomatictransgenic SOD1G93A�gp91phox� mice than from age-matchedSOD1G93A�gp91phox� mice (Fig. 4 C–E). There were also smallerphospho-Akt:total Akt ratios (Fig. 4 F and G) as well as fewer cellsthat were immunoreactive for a downstream target of Akt, phos-pho-BAD (Fig. 4 H–J), and smaller phospho-BAD:total BAD ratios(Fig. 4 K and L) in symptomatic transgenic SOD1G93A�gp91phox�

Fig. 3. Deletion of gp91phox in-creases lifespan and lessens neuro-degeneration in transgenicSOD1G93A mice. (A) Survival com-parison of transgenic SOD1G93A�gp91phox� mice (red) (122.0 � 1.7days; n � 19) and transgenicSOD1G93A�gp91phox� littermates(green) (135.2 � 1.9 days; n � 17)(log-rank test � 15.3; P � 0.001).(B–D) Immunohistochemistry fornonphosphorylated heavy-chainneurofilament in the spinal cord.(E) Quantification of large (�25�m) motor neurons per 14-�m-thick section from the fourth tofifth lumbar spinal cord segments.(F–K) Toluidine blue-stained L5 an-terior root sections. (L) Quantifica-tion of myelinated axons in fifth-lumbar (L5) anterior roots. (M–P)Immunolabeling of the muscularacetylcholine receptor by an anti-�-bungarotoxin antibody (red) (M)and of the nerve terminals by ananti-heavy-chain neurofilament(green) (N) in the fibularis and per-oneus longus muscles; the mergedimage (O) shows a normally inner-vated end plate. (P) Quantificationof innervated end plates. (Q–S)H&E-stained fibularis and peroneuslongus muscle sections. (T) Quantification of muscle fiber size. See the legend of Fig. 1 for abbreviations. Except for the survival analysis, mice were all 115 daysold. In quantifications, values (means � SEM; n � 4–6 mice per group) were compared by using ANOVA followed by Newman–Keuls post hoc testing. *, P � 0.05,lower than nontransgenic controls; #, P � 0.05, higher than transgenic SOD1G93A�gp91phox� mice. (Scale bar in S: B–D and F–H, 80 �m; I–K, 8 �m; M–O, 15 �m;Q–S, 30 �m.)

12134 � www.pnas.org�cgi�doi�10.1073�pnas.0603670103 Wu et al.

Dow

nloa

ded

by g

uest

on

Janu

ary

20, 2

021

Page 4: The inflammatory NADPH oxidase enzyme modulates motor ...The inflammatory NADPH oxidase enzyme modulates motor neuron degeneration in amyotrophic lateral sclerosis mice Du-Chu Wu*†,

mice compared with their age-matched SOD1G93A�gp91phox�

counterparts. These data further support the idea that oxidativemodification of IGF1 receptor in symptomatic transgenicSOD1G93A�gp91phox� mice is associated with a range of molecularperturbations.

Microglial-Derived ROS Recapitulate the IGF1�Akt Pathway Defect inVitro. To test the idea that NADPH oxidase-derived ROS couldimpair IGF1 pathway function, an in vitro cell system using theneuron-like cell lines SH-SY5Y and IMR32 was used. First, cellswere briefly incubated with 0.1–100 �M human recombinant IGF1in the presence of overnight-preconditioned serum-free mediumsupplemented with or without 0.1 milliunits�ml glucose oxidase toprovide a constant flux of H2O2; IGF1 pathway responsiveness wasmonitored by Akt phosphorylation. Exposure to IGF1 caused adose-dependent phosphorylation of Akt in SH-SY5Y cells (Fig. 5A and B) and IMR32 cells (data not shown) in the presence ofconditioned medium lacking glucose oxidase. Conversely, IGF1barely increased Akt phosphorylation in the neuroblastoma cell

lines that were exposed to conditioned medium containing 0.1milliunits�ml glucose oxidase, generating an average stable con-centration of 75 �M H2O2 (Fig. 5E); this effect was abolished byadding 1,000 units�ml catalase to the medium (data not shown).Next, SH-SY5Y cells were incubated with or without conditionedmedium from LPS-activated BV2 microglial cells. Akin to theglucose oxidase experiments, brief exposure to LPS-activated mi-croglial-conditioned medium, which contained increased levels ofH2O2 (Fig. 5E), attenuated IGF1-mediated Akt phosphorylation inthe neuroblastoma cell line (Fig. 5 C and D). Upon longer exposureto LPS-activated microglial-conditioned medium, the Akt phos-phorylation response to the IGF1 recombinant remained de-pressed, and, at 72 h, a reduction of cell viability, indistinguishablefrom the condition without IGF1, was observed (Fig. 5F). However,both the alteration of IGF1-mediated Akt phosphorylation and theloss of cell viability mediated by LPS-activated microglia werecounteracted by the specific NADPH oxidase inhibitor apocynin(Fig. 5 C, D, and F).

DiscussionExperimental evidence supports a model for ALS neurodegenera-tion in which nonneuronal cells such as microglia contribute to thedemise of motor neurons (6, 16, 17). Germane to the molecularbasis of this deleterious effect on motor neurons is our finding thatvirtually all spinal cord microglial cells express the gp91phox subunitof the oxidant-producing enzyme NADPH oxidase (Fig. 1). Agree-ing with the fact that in nonactivated phagocytes NADPH oxidaseis quiescent (7), gp91phox-positive cells in spinal cords from 1- to4-month-old nontransgenic mice had a morphology of restingmicroglia and did not seem to produce ROS (Figs. 1 and 6).Conversely, in transgenic SOD1G93A mice, paralleling the worsen-ing of the ALS phenotype there was an intensification of spinal cordmicrogliosis accompanied by an up-regulation and activation ofNADPH oxidase. These results suggest that over the course of thedisease, spinal cord microglial cells become activated and acquirethe capacity of oxidatively damaging nearby macromolecules andcells homed within inflamed ALS tissues. Corroborating this vieware the levels of protein carbonyls, which were markedly elevated inspinal cord extracts of symptomatic transgenic SOD1G93A mice, forthe most part in a NADPH oxidase-dependent manner (Fig. 1).Evidence of microgliosis, NADPH oxidase up-regulation, andprotein carbonylation was also found in postmortem spinal cordsfrom human sporadic ALS cases (Fig. 2), supporting the conclusionthat the occurrence of inflammation-mediated oxidative damage isnot restricted to familial ALS caused by SOD1 mutations but is alsoa pathological hallmark of the prevalent nonfamilial, sporadic formof ALS.

Our results also show that abrogation of the gp91phox subunitof NADPH oxidase in transgenic SOD1G93A mice eliminates theproduction of microglial-derived ROS (Fig. 1M) and, impor-tantly, prolongs survival and retards neurodegeneration in thisALS model (Fig. 3). Deletion of gp91phox in transgenic SOD1G93A

mice did not alter the spinal cord microglial response or theexpression of human SOD1 in transgenic SOD1G93A mice (Fig. 7),which is a known determinant of disease severity in this ALS model(18). Consequently, the attenuated phenotype seen in transgenicSOD1G93A�gp91phox� mice is attributable to the lack of NADPHoxidase activity and not to either an impaired microglial responseor expression of the human SOD1G93A transgene. These dataprovide compelling evidence that NADPH oxidase contributes tothe degeneration of motor neurons in ALS. They also underscorethe significance of inflammatory-mediated oxidative stress in thepathogenesis of chronic, noninfectious pathological conditions suchas ALS. However, the magnitude of benefit afforded by gp91phox

deletion in transgenic SOD1G93A mice argues that targeting neu-roinflammation by inhibiting just one of its mediators, such asNADPH oxidase, may not be sufficient to produce robust andlasting neuroprotection in ALS patients.

Fig. 4. Modulation of the IGF1�Akt pathway by NADPH oxidase-derivedROS. (A) Immunoprecipitation of IGF1 receptor �-chain followed by OxyBlot(upper blot) and immunoblot for spinal cord IGF1 receptor �-chain (lowerblot). (B) Bar graph showing carbonylated�total �-chain ratios in the fourgenotypes shown in A. (C–E) Immunostaining for spinal cord phospho-IGF1receptor in the different mouse groups. (F) Spinal cord phospho-Akt (P-Akt)(upper blot) and total Akt (lower blot) immunoblots. (G) Bar graph showingP-Akt�total Akt ratios in the four genotypes in F. (H–J) Immunostaining forspinal cord phospho-BAD in the different mouse groups. (K) Immunoprecipi-tation of spinal cord BAD followed by immunoblot for phospho-BAD (P-BAD)(upper blot) and total BAD (lower blot). (L) Bar graph showing P-BAD�totalBAD ratios in the four genotypes in K. The mice were all 115 days old. In thequantifications, values (means � SEM; n � 4–8 mice per group) were com-pared by ANOVA followed by Newman–Keuls post hoc testing. *, P � 0.05,different from nontransgenic controls; #, P � 0.05, different from transgenicSOD1G93A�gp91phox� mice. (Scale bar in J: C–E and H–J, 0.2 mm.)

Wu et al. PNAS � August 8, 2006 � vol. 103 � no. 32 � 12135

NEU

ROSC

IEN

CE

Dow

nloa

ded

by g

uest

on

Janu

ary

20, 2

021

Page 5: The inflammatory NADPH oxidase enzyme modulates motor ...The inflammatory NADPH oxidase enzyme modulates motor neuron degeneration in amyotrophic lateral sclerosis mice Du-Chu Wu*†,

All cells, not motor neurons only, which are located in the vicinityof activated microglia, may indiscriminately have their plasmamembrane proteins and lipids damaged by NADPH oxidase-derived ROS. However, the chronic nature of ALS suggests thatneuroinflammation is likely protracted and not as strong as in acuteencephalitis. Accordingly, two hypotheses that are not mutuallyexclusive can be formulated to reconcile the expected nonselectivenature of the oxidative stress with the selective demise of motorneurons in ALS. First, at that lower level of ROS production,oxidative stress may not kill cells but instead may accelerate thedemise of those already compromised, as motor neurons probablyare in ALS. Second, in this paralytic disease, microglial-derivedoxidants may preferentially worsen motor neuron degeneration bydamaging proteins that are specifically important for their survival.Relevant to the latter scenario are our results for IGF1, a trophicfactor that is known to promote motor neuron survival (19–21).

In this study, we indeed found that receptors for IGF1 wereprimarily expressed on motor neurons in mouse spinal cords (Fig.8) and that the IGF1 signaling pathway was impaired by a NADPHoxidase-dependent mechanism in symptomatic transgenicSOD1G93A mice (Fig. 4). Although IGF1 per se did not seem to bedamaged by inflammation, NADPH oxidase did stimulate theoxidative modification of IGF1 receptors (Fig. 4). The ligand-dependent kinase activation of IGF1 receptor relies on its arrange-ment into a heterotetrameric 2��2�-chain complex (22). It maythus be predicted that oxidation of the IGF1 receptor mainextracellular domains (i.e., the �-chains) could distort its properassembly and transduction activity. Our in vivo and in vitro results(Figs. 4 and 5), in aggregate, show that several molecular events thatare normally elicited by ligation of the IGF1 receptor, includingautophosphorylation and Akt phosphorylation, were indeed abatedby ROS in a microglial NADPH oxidase-dependent manner.

Our data also show that microglial NADPH oxidase, by impair-ing the IGF1 signaling pathway, renders SH-SY5Y cells in our invitro system more prone to die upon exposure to a hostile environ-ment such as that emulated by LPS-activated microglial-conditioned medium (Fig. 5). These results thus suggest that

reduction in vital surviving pathways may be one mechanism bywhich inflammation attenuates the capability of motor neurons towithstand the toxicity of etiologic agents such as mutant SOD1.Muscle-specific expression of IGF1 stabilizes neuromuscular junc-tions, reduces inflammation in the spinal cord, and enhances motorneuronal survival in transgenic SOD1G93A mice (23). In our study,however, we did not find any evidence that the rescue of the IGF1pathway by abrogating NADPH oxidase was associated with musclehypertrophy (Fig. 3). Nevertheless, whether transgenic SOD1G93A

mice carrying the gp91phox null mutation reach end-stage paralysislater and exhibit an attenuated neurodegenerative process becauseof some effects at the skeletal muscle level is an interestingpossibility that cannot be excluded.

Injection of transgenic SOD1G93A mice with an adeno-associatedvirus carrying an IGF1 gene prolongs survival in these animals (20,24). Together with our data, these previous studies suggest thatinflammation may not abrogate, but instead may blunt, the motorneuron survival response to IGF1 in ALS. Perhaps the modestchange in ALS progression that is seen in patients treated withhuman recombinant IGF1 (25) may be related to the issue raisedabove. It may thus be argued that optimal therapeutic response toIGF1 in diseases such as ALS may rely on a concomitant admin-istration of this trophic factor and an antiinflammatory agent.

Materials and MethodsAnimals. Transgenic SOD1G93A mice [C57BL�6J-TgN(SOD1-G93A)1Gurdl] were crossed with gp91phox-deficient mice(B6.129S6-Cybbtm1din). Both lines were obtained from The JacksonLaboratory (Bar Harbor, ME) and maintained in the C57BL�6Jstrain. Progeny were genotyped by PCR as described in refs. 26 and27. Refer to Supporting Text, which is published as supportinginformation on the PNAS web site, for details about the timeline ofbehavioral abnormalities in transgenic SOD1G93A mice.

RNA Extraction and RT-PCR. Total RNA was extracted as describedin ref. 28. Primer sequences for gp91phox, glial fibrillary acidicprotein, macrophage antigen complex 1, and GAPDH and PCR

Fig. 5. Glucose oxidase- and microglial-derived ROS impair the IGF1 Akt pathway in vitro. (A) Phospho-Akt (upper blot) and total Akt (lower blot) immunoblotsof IGF1-treated SH-SY5Y cells exposed or not exposed to 75 �M glucose oxidase-generated H2O2. (B) Bar graph showing phospho-Akt�total Akt ratios for the differentIGF1 doses. #, P � 0.05, higher than 0.1 �M IGF1; $, P � 0.05, lower than the same IGF1 concentration without glucose oxidase. (C) Phospho-Akt (upper blot) and totalAkt (lower blot) immunoblots of SH-SY5Y cells exposed to various BV2 serum-free conditioned media supplemented or not supplemented with 10 �M IGF1. (D) Bargraph showing phospho-Akt�total Akt ratios. *, P � 0.05, lower than control and �LPS �APO in the IGF1-treated group. (E) Bar graph showing the H2O2 mediumconcentrations for the different conditions. *, P � 0.05, higher H2O2 concentration than controls and �LPS �APO. (F) SH-SY5Y survival kinetic over 72-h exposure tovarious BV2 serum-free conditioned media supplemented or not supplemented with 10 �M IGF1. APO, apocynin. Cell survival was assessed by counting DAPI-labelednormalnucleiandconfirmedby3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazoliumbromideassay (datanotshown).Dataareaveragesof threeormore independentexperimentsandwerecomparedbyANOVAorrepeated-measuresANOVAfor thesurvivalkinetic, followedbyNewman–Keulsposthoc testing.For thesurvivalkinetic,only LPS and IGF1 � LPS within the 72-h time point are not significantly different from each other (P � 0.386).

12136 � www.pnas.org�cgi�doi�10.1073�pnas.0603670103 Wu et al.

Dow

nloa

ded

by g

uest

on

Janu

ary

20, 2

021

Page 6: The inflammatory NADPH oxidase enzyme modulates motor ...The inflammatory NADPH oxidase enzyme modulates motor neuron degeneration in amyotrophic lateral sclerosis mice Du-Chu Wu*†,

conditions are presented in Supporting Text. Products were quan-tified with a phosphorimager (Bio-Rad, Hercules, CA).

Immunohistochemistry and Histological Methods. Mouse spinal cordswere fixed and processed for immunostaining as described in ref. 29.All antibodies used in this study are listed in Supporting Text. Forquantification of motor neurons, fixed spinal cord slices wereimmunostained with SMI-32 monoclonal antibody directed againstthe nonphosphorylated neurofilament heavy chain (SternbergerMonoclonals, Lutherville, MD). SMI-32-positive large (�25 �m indiameter) motor neurons were counted as described in ref. 29 byusing 14-�m-thick sections. Preparation and processing of L5anterior roots and fibularis and peroneus longus muscles were alsoperformed as described in ref. 29 with only minor modifications.Paraffin-embedded muscle sections were stained with H&E, andfiber cross-sectional areas were determined by averaging the areaof 100 fibers; it was assumed that fibularis and peroneus longusmuscle fibers are cylindrical.

Immunoprecipitation and Western Blots. Total and plasma mem-brane proteins were prepared as described in ref. 27. All antibodiesused are listed in Supporting Text. Bound primary antibody wasdetected by using HRP-conjugated secondary antibodies againstIgG and a chemiluminescent substrate (SuperSignal Ultra; Pierce,Rockford, IL). Films were quantified by using the NIH Imageanalysis system.

In Situ Visualization of ROS. In situ visualization of superoxideradicals and derived oxidants was assessed on 14-�m-thick spinalcord sections by hydroethidine histochemistry, following a methoddescribed in ref. 11.

Measurement of Protein Oxidation. Protein carbonyls were detectedafter derivatization of spinal cord homogenates with 2,4-dinitrophenylhydrazine by using a modification of a method de-scribed in ref. 13.

OxyBlot on Immunoprecipitated Proteins. Supernatants of mousespinal cord tissue homogenized in Nonidet P-40 buffer contain-ing 50 mM DTT were collected and incubated (at 4°C for 1 h)with rabbit anti-IGF1 receptor � (Santa Cruz Biotechnology,Santa Cruz, CA) or rabbit anti-Akt (Cell Signaling Technology,Danvers, MA). Immunoprecipitation was performed as de-scribed above, followed by an immunoblot detection of carbonylgroups introduced into proteins by oxidative reaction using theOxyBlot protein oxidation kit (Chemicon, Temecula, CA) as

specified by the manufacturer. Refer to Supporting Text for moretechnical details. For immunohistochemistry, tissue sectionswere incubated with 3 �M 2,4-dinitrophenylhydrazine (DNPH)in 3% trif luoroacetic acid for 15 min at room temperature toconvert carbonyl to hydrazone derivatives. Sections were thenincubated with 1:500 rabbit anti-DNPH antibody and finallyvisualized with VECTOR SG (blue-gray).

IL-1� Measurement. Spinal cord content of mouse IL-1� was de-termined as specified by the manufacturer by using an ELISA kitthat is specific for this cytokine (R & D Systems, Minneapolis, MN).

Human Samples. Age at death and interval from death to tissueprocessing were as follows: control group (n � 6), 60.5 � 10.2 yearsand 8.0 � 2.6 h, respectively; ALS group, (n � 6), 60.5 � 4.2 yearsand 7.7 � 1.4 h, respectively. For the ALS patients, the meanduration of disease was 19.3 � 2.6 months. All human samples(anonymized) were kindly provided by A. Hays (Columbia Uni-versity Medical Center).

In Vitro Experiments. SH-SY5Y and IMR32 neuroblastoma cell lineswere purchased from the American Type Culture Collection (Ma-nassas, VA). The BV2 microglial cell line was kindly provided byTong H. Joh (Burke Medical Research Institute, New York, NY).All cell culture products were obtained from Gibco�Invitrogen(Carlsbad, CA). Phosphorylation of Akt and cell viability in re-sponse to IGF1 recombinant and to H2O2 or activated BV2 cellswere assessed at selected time points as described in SupportingText.

Statistical Analysis. Values are means � SEM. Differences be-tween means were tested by Student’s t test, and differencesamong means were tested by ANOVA followed by Newman–Keuls post hoc testing. Survival statistics were obtained byKaplan–Meyer analysis. The null hypothesis was rejected at the0.05 level.

We thank Ms. Julia Jeon for assistance in preparing the manuscript andDrs. Lewis P. Rowland and Irwin Fridovich for editorial comments. Thiswork was supported by the Muscular Dystrophy Association�Wings overWall Street; National Institutes of Health (NIH)�National Institute ofNeurological Disorders and Stroke Grants NS42269, NS38370, andNS11766; NIH Grant AG 021617; National Institute of EnvironmentalHealth Sciences (NIEHS) Grant ES013177; U.S. Department of DefenseGrant DAMD 17-03-1; the Parkinson’s Disease Foundation GrantCU51523606; and a Gardner’s Fellowship from the Muscular DystrophyAssociation (to M.N.).

1. Rowland, L. P. & Shneider, N. A. (2001) N. Engl. J. Med. 344, 1688–1700.2. Rosen, D. R., Siddique, T., Patterson, D., Figlewicz, D. A., Sapp, P., Hentati, A.,

Donaldson, D., Goto, J., O’Regan, J. P., Deng, H.-X., et al. (1993) Nature 362, 59–62.3. Deng, H.-X., Hentati, A., Tainer, J. A., Iqbal, Z., Cayabyab, A., Hung, W.-Y., Getzoff,

E. D., Hu, P., Herzfeldt, B., Roos, R. P., et al. (1993) Science 261, 1047–1051.4. Julien, J. P. (2001) Cell 104, 581–591.5. Clement, A. M., Nguyen, M. D., Roberts, E. A., Garcia, M. L., Boillee, S., Rule, M.,

McMahon, A. P., Doucette, W., Siwek, D., Ferrante, R. J., et al. (2003) Science 302,113–117.

6. Boillee, S., Yamanaka, K., Lobsiger, C. S., Copeland, N. G., Jenkins, N. A., Kassiotis,G., Kollias, G. & Cleveland, D. W. (2006) Science 312, 1389–1392.

7. Babior, B. M. (1999) Blood 93, 1464–1476.8. Heyworth, P. G., Cross, A. R. & Curnutte, J. T. (2003) Curr. Opin. Immunol. 15,

578–584.9. Li, J., Baud, O., Vartanian, T., Volpe, J. J. & Rosenberg, P. A. (2005) Proc. Natl.

Acad. Sci. USA 102, 9936–9941.10. Smith, R. M., Connor, J. A., Chen, L. M. & Babior, B. M. (1996) J. Clin. Invest. 98,

977–983.11. Bindokas, V. P., Jordan, J., Lee, C. C. & Miller, R. J. (1996) J. Neurosci. 16,

1324–1336.12. Pollock, J. D., Williams, D. A., Gifford, M. A., Li, L. L., Du, X., Fisherman, J., Orkin,

S. H., Doerschuk, C. M. & Dinauer, M. C. (1995) Nat. Genet. 9, 202–209.13. Levine, R. L., Garland, D., Oliver, C. N., Amici, A., Climent, I., Lenz, A.-G., Ahn,

B.-W., Shaltiel, S. & Stadtman, E. R. (1990) Methods Enzymol. 186, 464–478.14. Dalle-Donne, I., Giustarini, D., Colombo, R., Rossi, R. & Milzani, A. (2003) Trends

Mol. Med. 9, 169–176.15. Brazil, D. P. & Hemmings, B. A. (2001) Trends Biochem. Sci. 26, 657–664.

16. Nguyen, M. D., D’Aigle, T., Gowing, G., Julien, J. P. & Rivest, S. (2004) J. Neurosci.24, 1340–1349.

17. Urushitani, M., Sik, A., Sakurai, T., Nukina, N., Takahashi, R. & Julien, J. P. (2006)Nat. Neurosci. 9, 108–118.

18. Raoul, C., Abbas-Terki, T., Bensadoun, J. C., Guillot, S., Haase, G., Szulc, J.,Henderson, C. E. & Aebischer, P. (2005) Nat. Med. 11, 423–428.

19. Rind, H. B. & von Bartheld, C. S. (2002) Mol. Cell. Neurosci. 19, 58–71.20. Kaspar, B. K., Llado, J., Sherkat, N., Rothstein, J. D. & Gage, F. H. (2003) Science

301, 839–842.21. Neff, N. T., Prevette, D., Houenou, L. J., Lewis, M. E., Glicksman, M. A., Yin, Q. W.

& Oppenheim, R. W. (1993) J. Neurobiol. 24, 1578–1588.22. Boni-Schnetzler, M., Rubin, J. B. & Pilch, P. F. (1986) J. Biol. Chem. 261,

15281–15287.23. Dobrowolny, G., Giacinti, C., Pelosi, L., Nicoletti, C., Winn, N., Barberi, L., Molinaro,

M., Rosenthal, N. & Musaro, A. (2005) J. Cell Biol. 168, 193–199.24. Kaspar, B. K., Frost, L. M., Christian, L., Umapathi, P. & Gage, F. H. (2005) Ann.

Neurol. 57, 649–655.25. Lai, E. C., Felice, K. J., Festoff, B. W., Gawel, M. J., Gelinas, D. F., Kratz, R., Murphy,

M. F., Natter, H. M., Norris, F. H. & Rudnicki, S. A. (1997) Neurology 49, 1621–1630.26. Kikuchi, H., Almer, G., Yamashita, S., Guegan, C., Nagai, M., Xu, Z., Sosunov, A. A.,

McKhann, G. M. & Przedborski, S. (2006) Proc. Natl. Acad. Sci. USA 103, 6025–6030.27. Wu, D. C., Teismann, P., Tieu, K., Vila, M., Jackson-Lewis, V., Ischiropoulos, H. &

Przedborski, S. (2003) Proc. Natl. Acad. Sci. USA 100, 6145–6150.28. Almer, G., Guegan, C., Teismann, P., Naini, A., Rosoklija, G., Hays, A. P., Chen, C.

& Przedborski, S. (2001) Ann. Neurol. 49, 176–185.29. Kostic, V., Jackson-Lewis, V., De Bilbao, F., Dubois-Dauphin, M. & Przedborski, S.

(1997) Science 277, 559–562.

Wu et al. PNAS � August 8, 2006 � vol. 103 � no. 32 � 12137

NEU

ROSC

IEN

CE

Dow

nloa

ded

by g

uest

on

Janu

ary

20, 2

021