Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Eur Neuropsychopharmacol ; 26(6): 1048-61, 2016 06.
Article in English | MEDLINE | ID: mdl-26996375

ABSTRACT

Prolyl oligopeptidase (PREP) has been implicated in neurodegeneration and neuroinflammation and has been considered a drug target to enhance memory in dementia. However, the true physiological role of PREP is not yet understood. In this paper, we report the phenotyping of a mouse line where the PREP gene has been knocked out. This work indicates that the lack of PREP in mice causes reduced anxiety but also hyperactivity. The cortical volumes of PREP knockout mice were smaller than those of wild type littermates. Additionally, we found increased expression of diazepam binding inhibitor protein in the cortex and of the somatostatin receptor-2 in the hippocampus of PREP knockout mice. Furthermore, immunohistochemistry and tail suspension test revealed lack of response of PREP knockout mice to lipopolysaccharide insult. Further analysis revealed significantly increased levels of polysialylated-neural cell adhesion molecule in PREP deficient mice. These findings might be explained as possible alteration in brain plasticity caused by PREP deficiency, which in turn affect behaviour and brain development.


Subject(s)
Anxiety/genetics , Anxiety/psychology , Behavior, Animal , Neuronal Plasticity/genetics , Serine Endopeptidases/deficiency , Synapses/genetics , Animals , Anxiety/pathology , Body Weight/genetics , Brain/pathology , Cytokines/blood , Hindlimb Suspension , Hyperkinesis/genetics , Hyperkinesis/psychology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Phenotype , Prolyl Oligopeptidases , Receptors, Somatostatin/biosynthesis , Receptors, Somatostatin/genetics
2.
J Neuropathol Exp Neurol ; 73(8): 752-69, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25003233

ABSTRACT

Understanding the pathophysiologic mechanisms underlying Alzheimer disease relies on knowledge of disease onset and the sequence of development of brain pathologies. We present a comprehensive analysis of early and progressive changes in a mouse model that demonstrates a full spectrum of characteristic Alzheimer disease-like pathologies. This model demonstrates an altered immune redox state reminiscent of the human disease and capitalizes on data indicating critical differences between human and mouse immune responses, particularly in nitric oxide levels produced by immune activation of the NOS2 gene. Using the APPSwDI(+)/(+)mNos2(-/-) (CVN-AD) mouse strain, we show a sequence of pathologic events leading to neurodegeneration,which include pathologically hyperphosphorylated tau in the perforant pathway at 6 weeks of age progressing to insoluble tau, early appearance of ß-amyloid peptides in perivascular deposits around blood vessels in brain regions known to be vulnerable to Alzheimer disease, and progression to damage and overt loss in select vulnerable neuronal populations in these regions. The role of species differences between hNOS2 and mNos2 was supported by generating mice in which the human NOS2 gene replaced mNos2. When crossed with CVN-AD mice, pathologic characteristics of this new strain (APPSwDI(+)/(-)/HuNOS2(tg+)/(+)/mNos2(-/-)) mimicked the pathologic phenotypes found in the CVN-AD strain.


Subject(s)
Alzheimer Disease/genetics , Disease Models, Animal , Mutation/genetics , Nitric Oxide Synthase Type II/deficiency , Nitric Oxide Synthase Type II/genetics , Age Factors , Alzheimer Disease/complications , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Brain/pathology , Conditioning, Classical/physiology , Gene Expression Regulation/genetics , Humans , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/genetics , Nitric Oxide Synthase Type II/metabolism , Phosphorylation/genetics
3.
Biochem Pharmacol ; 85(12): 1783-94, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23643808

ABSTRACT

Prolyl oligopeptidase (PREP) has been considered as a drug target for the treatment of neurodegenerative diseases. In plasma, PREP has been found altered in several disorders of the central nervous system including multiple sclerosis (MS). Oxidative stress and the levels of an endogenous plasma PREP inhibitor have been proposed to decrease PREP activity in MS. In this work, we measured the circulating levels of PREP in patients suffering of relapsing remitting (RR), secondary progressive (SP), primary progressive (PP) MS, and in subjects with clinically isolated syndrome (CIS). We found a significantly lower PREP activity in plasma of RRMS as well as in PPMS patients and a trend to reduced activity in subjects diagnosed with CIS, compared to controls. No signs of oxidative inactivation of PREP, and no correlation with the endogenous PREP inhibitor, identified as activated α-2-macroglobulin (α2M*), were observed in any of the patients studied. However, a significant decrease of α2M* was recorded in MS. In cell cultures, we found that PREP specifically stimulates immune active cells possibly by modifying the levels of fibrinogen ß, thymosin ß4, and collagen. Our results open new lines of research on the role of PREP and α2M* in MS, aiming to relate them to the diagnosis and prognosis of this devastating disease.


Subject(s)
Demyelinating Diseases/blood , Multiple Sclerosis, Chronic Progressive/blood , Multiple Sclerosis, Relapsing-Remitting/blood , Serine Endopeptidases/blood , alpha-Macroglobulins/metabolism , Adult , Aged , Animals , Biomarkers/blood , Cell Line, Tumor , Demyelinating Diseases/diagnosis , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Multiple Sclerosis/blood , Multiple Sclerosis/diagnosis , Multiple Sclerosis, Chronic Progressive/diagnosis , Multiple Sclerosis, Relapsing-Remitting/diagnosis , Prolyl Oligopeptidases , Young Adult
4.
PLoS One ; 7(12): e50717, 2012.
Article in English | MEDLINE | ID: mdl-23284644

ABSTRACT

Huntington's disease (HD) is an autosomal neurodegenerative disorder, characterized by severe behavioral, cognitive, and motor deficits. Since the discovery of the huntingtin gene (HTT) mutation that causes the disease, several mouse lines have been developed using different gene constructs of Htt. Recently, a new model, the zQ175 knock-in (KI) mouse, was developed (see description by Menalled et al, [1]) in an attempt to have the Htt gene in a context and causing a phenotype that more closely mimics HD in humans. Here we confirm the behavioral phenotypes reported by Menalled et al [1], and extend the characterization to include brain volumetry, striatal metabolite concentration, and early neurophysiological changes. The overall reproducibility of the behavioral phenotype across the two independent laboratories demonstrates the utility of this new model. Further, important features reminiscent of human HD pathology are observed in zQ175 mice: compared to wild-type neurons, electrophysiological recordings from acute brain slices reveal that medium spiny neurons from zQ175 mice display a progressive hyperexcitability; glutamatergic transmission in the striatum is severely attenuated; decreased striatal and cortical volumes from 3 and 4 months of age in homo- and heterozygous mice, respectively, with whole brain volumes only decreased in homozygotes. MR spectroscopy reveals decreased concentrations of N-acetylaspartate and increased concentrations of glutamine, taurine and creatine + phosphocreatine in the striatum of 12-month old homozygotes, the latter also measured in 12-month-old heterozygotes. Motor, behavioral, and cognitive deficits in homozygotes occur concurrently with the structural and metabolic changes observed. In sum, the zQ175 KI model has robust behavioral, electrophysiological, and histopathological features that may be valuable in both furthering our understanding of HD-like pathophyisology and the evaluation of potential therapeutic strategies to slow the progression of disease.


Subject(s)
Behavior, Animal , Brain/pathology , Disease Models, Animal , Gene Knock-In Techniques , Huntington Disease/pathology , Huntington Disease/physiopathology , Neurophysiology , Animals , Body Weight , Brain/metabolism , Brain/physiopathology , Cell Count , Disease Progression , Endpoint Determination , Female , Glutamic Acid/metabolism , Huntington Disease/genetics , Huntington Disease/metabolism , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Mice , Neostriatum/pathology , Nerve Tissue Proteins/genetics , Neurons/pathology , Organ Size , Repetitive Sequences, Nucleic Acid , Swimming , Synaptic Transmission
5.
J Alzheimers Dis ; 27(3): 499-510, 2011.
Article in English | MEDLINE | ID: mdl-21821875

ABSTRACT

The purpose of this study was to evaluate the efficacy of the radical scavenger IAC (bis(1-hydroxy-2,2,6,6-tetramethyl-4-piperidinyl) decantionate) in alleviating behavioral deficits and reducing amyloid-ß (Aß) accumulation in an Alzheimer's disease (AD) transgenic Tg2576 mouse model. Daily treatment with IAC (3-30 mg/kg, i.p.) was started at the age of 6 months and continued until the mice were 13 months old. At the age of 9 months and again at 12 months, the mice were tested in open field and water maze tests. At the age of 13 months, the mice were sacrificed and the brains processed for immunohistochemistry. Mortality was significantly reduced in all IAC-treated groups. In addition, IAC treatment improved the water maze hidden platform training performance but had no effect on motor activity in the open field or water maze swim speed in transgenic mice. Lastly, IAC treatment (10 mg/kg) significantly reduced the cortical Aß plaque burden. In vitro, IAC is able to increase the number of neurites and neurite branches in cultured cortical primary neurons. In conclusion, IAC slowed down the development of the AD-like phenotype in Tg2576 mice and accelerated neurite growth in cultured neurons.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Cognition/physiology , Maze Learning/physiology , Piperidines/therapeutic use , Plaque, Amyloid/drug therapy , Plaque, Amyloid/genetics , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cognition/drug effects , Cricetinae , Disease Models, Animal , Free Radical Scavengers/pharmacology , Free Radical Scavengers/therapeutic use , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Piperidines/pharmacology , Plaque, Amyloid/pathology , Rats , Rats, Wistar
6.
Phytother Res ; 20(10): 917-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16909443

ABSTRACT

Valeriana officinalis (Valerianaceae) has been of great interest for its therapeutic uses for treating mild nervous tension and temporary sleeping problems. In traditional European medicine it has been also reported as an antiinflammatory remedy. This study reports that the EtOAc extract of the underground parts of V. officinalis showed inhibitory activity against NF-kappaB at 100 microg/mL in the IL-6/Luc assay on HeLa cells and provided protection against excitotoxicity in primary brain cell cultures at micromolar concentrations. Bioassay-guided fractionation of the EtOAc extract led to the isolation of three known sesquiterpenes: acetylvalerenolic acid (1), valerenal (2) and valerenic acid (3), 1 and 3 were active as inhibitors of NF-kappaB at a concentration of 100 microg/mL. Acetylvalerenolic acid (1) reduced NF-kappaB activity to 4%, whereas valerenic acid (3) reduced NF-kappaB activity to 25%.


Subject(s)
NF-kappa B/antagonists & inhibitors , Plant Extracts/pharmacology , Valerian/chemistry , Astrocytes/cytology , Astrocytes/drug effects , Coculture Techniques , HeLa Cells , Humans , Kainic Acid/antagonists & inhibitors , N-Methylaspartate/antagonists & inhibitors , Neurons/cytology , Neurons/drug effects , Plant Extracts/chemistry , Plant Extracts/isolation & purification
7.
Proc Natl Acad Sci U S A ; 103(23): 8852-7, 2006 Jun 06.
Article in English | MEDLINE | ID: mdl-16723396

ABSTRACT

beta-Amyloid (Abeta) polypeptide plays a critical role in the pathogenesis of Alzheimer's disease (AD), which is characterized by progressive decline of cognitive functions, formation of Abeta deposits and neurofibrillary tangles, and loss of neurons. Increased genetic production or direct intracerebral administration of Abeta in animal models results in Abeta deposition, gliosis, and impaired cognitive functions. Whether aging renders the brain prone to Abeta and whether inflammation is required for Abeta-induced learning deficits is unclear. We show that intraventricular infusion of Abeta1-42 results in learning deficits in 9-month-old but not 2.5-month-old mice. Deficits that become detectable 12 weeks after the infusion are associated with a slight reduction in Cu,Zn superoxide dismutase activity but do not correlate with Abeta deposition and are not associated with gliosis. In rats, Abeta infusion induced learning deficits that were detectable 6 months after the infusion. Approximately 20% of the Abeta immunoreactivity in rats was associated with astrocytes. NMR spectrum analysis of the animals cerebrospinal fluid revealed a strong reduction trend in several metabolites in Abeta-infused rats, including lactate and myo-inositol, supporting the idea of dysfunctional astrocytes. Even a subtle increase in brain Abeta1-42 concentration may disrupt normal metabolism of astrocytes, resulting in altered neuronal functions and age-related development of learning deficits independent of Abeta deposition and inflammation.


Subject(s)
Aging/physiology , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/pharmacology , Learning Disabilities/chemically induced , Maze Learning/drug effects , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacology , Animals , Brain/cytology , Brain/enzymology , Brain/pathology , Inflammation/metabolism , Infusions, Intravenous , Learning Disabilities/metabolism , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Inbred SHR
8.
J Neurochem ; 91(3): 755-65, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15485504

ABSTRACT

Pyrrolidine dithiocarbamate (PDTC) is an antioxidant and inhibitor of transcription factor nuclear factor kappa-B (NF-kappa B). Because the role of NF-kappa B in brain injury is controversial and another NF-kappa B inhibiting thiocarbamate, DDTC, was recently shown to increase ischaemic brain damage, we investigated the effect of PDTC on transient brain ischaemia. Ischaemia was induced by occlusion of the middle cerebral artery (MCAO). In Wistar rats, the PDTC treatment started even 6 h after MCAO reduced the infarction volume by 48%. PDTC protected against MCAO also in spontaneously hypertensive rats and against forebrain ischaemia in Mongolian gerbils. PDTC prevented NF-kappa B activation in the ischaemic brain as determined by reduced DNA binding and nuclear translocation of NF-kappa B in neurons. PDTC had anti-inflammatory effect by preventing induction of NF-kappa B-regulated pro-inflammatory genes. In ischaemic rats, NF-kappa B was localized in cyclo-oxygenase-2-immunoreactive neurons. Blood cytokine levels were not altered by ischaemia or PDTC. When cultured neurons were exposed to an excitotoxin, no production of reactive oxygen species was detected, but PDTC provided protection and prevented nuclear translocation of NF-kappa B. The clinically approved PDTC and its analogues may act as anti-inflammatories and may be safe therapies in stroke with a wide time window.


Subject(s)
Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/prevention & control , NF-kappa B/metabolism , Neurons/drug effects , Neurons/metabolism , Pyrrolidines/therapeutic use , Thiocarbamates/therapeutic use , Animals , Cells, Cultured , Cyclooxygenase 2 , Cytokines/blood , Disease Models, Animal , Gerbillinae , Ischemic Attack, Transient/pathology , Male , Microglia/drug effects , Microglia/pathology , NF-kappa B/antagonists & inhibitors , Neurons/pathology , Neuroprotective Agents/therapeutic use , Neurotoxins/antagonists & inhibitors , Neurotoxins/toxicity , Prostaglandin-Endoperoxide Synthases/metabolism , Protein Transport/drug effects , Rats , Rats, Inbred SHR , Rats, Wistar , Time Factors , Treatment Outcome
9.
Stroke ; 34(3): 752-7, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12624303

ABSTRACT

BACKGROUND AND PURPOSE: Acetylsalicylic acid (ASA) is preventive against stroke and protects against focal brain ischemia in rats. We studied the mechanisms of the manner in which ASA provides neuroprotection against hypoxia/reoxygenation (H/R) injury. METHODS: Spinal cord cultures exposed to 20 hours of hypoxia followed by reoxygenation were treated with a vehicle, ASA or inhibitors of inducible nitric oxide synthase (iNOS), mitogen-activated protein kinases p38 MAPK and ERK1/2, or an N-methyl-d-aspartic acid (NMDA) receptor antagonist. Cell viability was assessed by LDH release measurement and cell counts. Prostaglandin production was measured by enzyme immunoassay, MAPK signaling by immunoblotting, and DNA binding of nuclear factor-kappaB (NF-kappaB) and activating protein-1 (AP-1) by electrophoretic mobility shift assay. RESULTS: One to 3 mmol/L ASA inhibited H/R-induced neuronal death when present during H/R but not when administered only for the reoxygenation period. Prostaglandin E2 production was very low and was not altered by ASA. The AP-1 and NF-kappaB DNA binding activities increased after H/R. ASA increased the H/R-induced AP-1 binding but had no effect on NF-kappaB binding. H/R induced a sustained ERK1/2 activation followed by neuronal death, whereas no changes in p38 or c-Jun N-terminal kinase were detected. ASA strongly inhibited this ERK1/2 activation. PD98059, an ERK1/2 inhibitor, was also neuroprotective, prevented H/R-induced ERK1/2 activation, and had no effect on NF-kappaB binding activity. Inhibition of NMDA receptors, iNOS, or p38 MAPK did not provide neuroprotection. CONCLUSIONS: Inhibition of the sustained activation of ERK1/2 may partially contribute to neuroprotection achieved by ASA against H/R injury.


Subject(s)
Aspirin/pharmacology , Enzyme Inhibitors/pharmacology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neurons/drug effects , Animals , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Survival/drug effects , Cells, Cultured , Dinoprostone/metabolism , Enzyme Activation/drug effects , Excitatory Amino Acid Antagonists/pharmacology , In Vitro Techniques , L-Lactate Dehydrogenase/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Neurons/cytology , Neurons/enzymology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type II , Oxygen/metabolism , Oxygen/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Spinal Cord/cytology , Spinal Cord/drug effects , Spinal Cord/enzymology , Transcription Factor AP-1/metabolism , p38 Mitogen-Activated Protein Kinases
10.
J Neurochem ; 82(2): 329-35, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12124433

ABSTRACT

Aspirin [acetylsalicylic acid (ASA)] is an anti-inflammatory drug that protects against cellular injury by inhibiting cyclooxygenases (COX), inducible nitric oxide synthase (iNOS) and p44/42 mitogen-activated protein kinase (p44/42 MAPK), or by preventing translocation of nuclear factor kappaB (NF-kappaB). We studied the effect of ASA pre-treatment on neuronal survival after hypoxia/reoxygenation damage in rat spinal cord (SC) cultures. In this injury model, COX, iNOS and NF-kappaB played no role in the early neuronal death. A 20-h treatment with 3 mm ASA prior to hypoxia/reoxygenation blocked the hypoxia/reoxygenation-induced lactate dehydrogenase (LDH) release from neurons. This neuroprotection was associated with increased phosphorylation of neurofilaments, which are substrates of p44/42 MAPK and cyclin-dependent kinase 5 (Cdk5). PD90859, a p44/42 MAPK inhibitor, had no effect on ASA-induced tolerance, but olomoucine and roscovitine, Cdk5 inhibitors, reduced ASA neuroprotection. Hypoxia/reoxygenation alone reduced both the protein amount and activity of Cdk5, and this reduction was inhibited by pre-treatment with ASA. Moreover, the protein amount of a neuronal Cdk5 activator, p35, recovered after reoxygenation only in ASA-treated samples. The prevention of the loss in Cdk5 activity during reoxygenation was crucial for ASA-induced protection, because co-administration of Cdk5 inhibitors at the onset ofreoxygenation abolished the protection. In conclusion, pre-treatment with ASA induces tolerance against hypoxia/reoxygenation damage in spinal cord cultures by restoring Cdk5 and p35 protein expression.


Subject(s)
Aspirin/pharmacology , Cell Hypoxia/drug effects , Cyclin-Dependent Kinases/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Survival/drug effects , Cells, Cultured , Cyclin-Dependent Kinase 5 , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Hypoxia, Brain/metabolism , Hypoxia, Brain/prevention & control , Immunohistochemistry , L-Lactate Dehydrogenase/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurofilament Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Phosphorylation/drug effects , Rats , Rats, Wistar , Spinal Cord/cytology , Spinal Cord/embryology
11.
Brain ; 125(Pt 4): 722-31, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11912107

ABSTRACT

CSF from patients with motor neurone disease (MND) has been reported to be toxic to cultured primary neurones. We found that CSF from MND patients homozygous for the D90A CuZn-superoxide dismutase (CuZn-SOD) mutation, patients with sporadic MND and patients with familial MND without CuZn-SOD mutations significantly increased apoptosis and reduced phosphorylation of neurofilaments in cultured spinal cord neurones when compared with the effects of CSF from patients with other neurological diseases. Exposure of spinal cord cultures to MND CSF also triggered microglial activation. The toxicity of MND CSF was independent of the presence of the CuZn-SOD mutation, and it did not correlate with gelatinase activity or the presence of immunoglobulin G autoantibodies in the CSF. The concentrations of glutamate, aspartate and glycine in MND CSF were not elevated. Antagonists of N-methyl-D-aspartate (NMDA) and alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid/kainate receptors prevented the toxic CSF-induced neuronal death but not microglial activation, whereas minocycline, a tetracycline derivative with anti-inflammatory potential independent of antimicrobial activity, reduced both the apoptotic neuronal death and microglial activation. We conclude that the cytotoxic action of CSF is prevalent in all MND cases and that microglia may mediate the toxicity of CSF by releasing excitotoxicity-enhancing factors.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cerebrospinal Fluid Proteins/toxicity , Minocycline/pharmacology , Motor Neuron Disease/cerebrospinal fluid , Motor Neuron Disease/drug therapy , Motor Neurons/drug effects , Neuroprotective Agents/pharmacology , Neurotoxins/toxicity , Spinal Cord/drug effects , Aged , Aged, 80 and over , Animals , Anti-Bacterial Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/physiology , Cells, Cultured , Cerebrospinal Fluid Proteins/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Female , Glutamic Acid/metabolism , Humans , Male , Microglia/drug effects , Microglia/metabolism , Middle Aged , Minocycline/therapeutic use , Motor Neuron Disease/genetics , Motor Neurons/metabolism , Motor Neurons/pathology , Mutation/genetics , Neuroprotective Agents/therapeutic use , Neurotoxins/metabolism , Rats , Spinal Cord/metabolism , Spinal Cord/physiopathology , Superoxide Dismutase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...