Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Nat Med ; 28(9): 1813-1822, 2022 09.
Article in English | MEDLINE | ID: mdl-36064599

ABSTRACT

Amyotrophic lateral sclerosis (ALS) involves progressive motor neuron loss, leading to paralysis and death typically within 3-5 years of diagnosis. Dysfunctional astrocytes may contribute to disease and glial cell line-derived neurotrophic factor (GDNF) can be protective. Here we show that human neural progenitor cells transduced with GDNF (CNS10-NPC-GDNF) differentiated to astrocytes protected spinal motor neurons and were safe in animal models. CNS10-NPC-GDNF were transplanted unilaterally into the lumbar spinal cord of 18 ALS participants in a phase 1/2a study (NCT02943850). The primary endpoint of safety at 1 year was met, with no negative effect of the transplant on motor function in the treated leg compared with the untreated leg. Tissue analysis of 13 participants who died of disease progression showed graft survival and GDNF production. Benign neuromas near delivery sites were common incidental findings at post-mortem. This study shows that one administration of engineered neural progenitors can provide new support cells and GDNF delivery to the ALS patient spinal cord for up to 42 months post-transplantation.


Subject(s)
Amyotrophic Lateral Sclerosis , Neural Stem Cells , Amyotrophic Lateral Sclerosis/therapy , Animals , Disease Models, Animal , Glial Cell Line-Derived Neurotrophic Factor/genetics , Humans , Spinal Cord , Superoxide Dismutase
2.
Stem Cell Reports ; 10(6): 1696-1704, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29706501

ABSTRACT

Trophic factor delivery to the brain using stem cell-derived neural progenitors is a powerful way to bypass the blood-brain barrier. Protection of diseased neurons using this technology is a promising therapy for neurodegenerative diseases. Glial cell line-derived neurotrophic factor (GDNF) has provided benefits to Parkinsonian patients and is being used in a clinical trial for amyotrophic lateral sclerosis. However, chronic trophic factor delivery prohibits dose adjustment or cessation if side effects develop. To address this, we engineered a doxycycline-regulated vector, allowing inducible and reversible expression of a therapeutic molecule. Human induced pluripotent stem cell (iPSC)-derived neural progenitors were stably transfected with the vector and transplanted into the adult mouse brain. Doxycycline can penetrate the graft, with addition and withdrawal providing inducible and reversible GDNF expression in vivo, over multiple cycles. Our findings provide proof of concept for combining gene and stem cell therapy for effective modulation of ectopic protein expression in transplanted cells.


Subject(s)
Gene Expression Regulation, Developmental , Glial Cell Line-Derived Neurotrophic Factor/genetics , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Stem Cell Transplantation , Cell- and Tissue-Based Therapy , Gene Expression , Genes, Reporter , Genetic Therapy , Genetic Vectors/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Plants, Genetically Modified , Stem Cell Transplantation/methods , Transduction, Genetic , Transgenes
3.
Prog Brain Res ; 230: 99-132, 2017.
Article in English | MEDLINE | ID: mdl-28552237

ABSTRACT

Ex vivo gene therapy involves the genetic modification of cells outside of the body to produce therapeutic factors and their subsequent transplantation back into patients. Various cell types can be genetically engineered. However, with the explosion in stem cell technologies, neural stem/progenitor cells and mesenchymal stem cells are most often used. The synergy between the effect of the new cell and the additional engineered properties can often provide significant benefits to neurodegenerative changes in the brain. In this review, we cover both preclinical animal studies and clinical human trials that have used ex vivo gene therapy to treat neurological disorders with a focus on Parkinson's disease, Huntington's disease, Alzheimer's disease, ALS, and stroke. We highlight some of the major advances in this field including new autologous sources of pluripotent stem cells, safer ways to introduce therapeutic transgenes, and various methods of gene regulation. We also address some of the remaining hurdles including tunable gene regulation, in vivo cell tracking, and rigorous experimental design. Overall, given the current outcomes from researchers and clinical trials, along with exciting new developments in ex vivo gene and cell therapy, we anticipate that successful treatments for neurological diseases will arise in the near future.


Subject(s)
Genetic Therapy , Nervous System Diseases/therapy , Animals , Cell- and Tissue-Based Therapy , Humans , Mesenchymal Stem Cell Transplantation , Nervous System Diseases/genetics , Neural Stem Cells , Stem Cell Transplantation
4.
Nat Neurosci ; 19(9): 1256-67, 2016 09.
Article in English | MEDLINE | ID: mdl-27428653

ABSTRACT

Modeling amyotrophic lateral sclerosis (ALS) with human induced pluripotent stem cells (iPSCs) aims to reenact embryogenesis, maturation and aging of spinal motor neurons (spMNs) in vitro. As the maturity of spMNs grown in vitro compared to spMNs in vivo remains largely unaddressed, it is unclear to what extent this in vitro system captures critical aspects of spMN development and molecular signatures associated with ALS. Here, we compared transcriptomes among iPSC-derived spMNs, fetal spinal tissues and adult spinal tissues. This approach produced a maturation scale revealing that iPSC-derived spMNs were more similar to fetal spinal tissue than to adult spMNs. Additionally, we resolved gene networks and pathways associated with spMN maturation and aging. These networks enriched for pathogenic familial ALS genetic variants and were disrupted in sporadic ALS spMNs. Altogether, our findings suggest that developing strategies to further mature and age iPSC-derived spMNs will provide more effective iPSC models of ALS pathology.


Subject(s)
Aging , Amyotrophic Lateral Sclerosis/metabolism , Motor Neurons/cytology , Neurogenesis/physiology , Amyotrophic Lateral Sclerosis/physiopathology , Gene Expression , Gene Regulatory Networks , Humans , Induced Pluripotent Stem Cells/physiology , Neurogenesis/genetics
5.
Exp Neurol ; 280: 41-9, 2016 06.
Article in English | MEDLINE | ID: mdl-27032721

ABSTRACT

Age-associated health decline presents a significant challenge to healthcare, although there are few animal models that can be used to test potential treatments. Here, we show that there is a significant reduction in both spinal cord motor neurons and motor function over time in the aging rat. One explanation for this motor neuron loss could be reduced support from surrounding aging astrocytes. Indeed, we have previously shown using in vitro models that aging rat astrocytes are less supportive to rat motor neuron function and survival over time. Here, we test whether rejuvenating the astrocyte niche can improve the survival of motor neurons in an aging spinal cord. We transplanted fetal-derived human neural progenitor cells (hNPCs) into the aging rat spinal cord and found that the cells survive and differentiate into astrocytes with a much higher efficiency than when transplanted into younger animals, suggesting that the aging environment stimulates astrocyte maturation. Importantly, the engrafted astrocytes were able to protect against motor neuron loss associated with aging, although this did not result in an increase in motor function based on behavioral assays. We also transplanted hNPCs genetically modified to secrete glial cell line-derived neurotrophic factor (GDNF) into the aging rat spinal cord, as this combination of cell and protein delivery can protect motor neurons in animal models of ALS. During aging, GDNF-expressing hNPCs protected motor neurons, though to the same extent as hNPCs alone, and again had no effect on motor function. We conclude that hNPCs can survive well in the aging spinal cord, protect motor neurons and mature faster into astrocytes when compared to transplantation into the young spinal cord. While there was no functional improvement, there were no functional deficits either, further supporting a good safety profile of hNPC transplantation even into the older patient population.


Subject(s)
Aging/physiology , Astrocytes/physiology , Cell Differentiation/physiology , Motor Neurons/physiology , Movement Disorders/surgery , Neural Stem Cells/physiology , Age Factors , Animals , Body Weight/physiology , Cerebral Cortex/cytology , Disease Models, Animal , Exploratory Behavior/physiology , Fetus/cytology , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Male , Movement Disorders/pathology , Movement Disorders/physiopathology , Muscle Strength/physiology , Nerve Tissue Proteins/metabolism , Neural Stem Cells/transplantation , Neuromuscular Junction/physiology , Rats , Rats, Sprague-Dawley , Spinal Cord/cytology , Spinal Cord/transplantation
6.
eNeuro ; 2(3)2015.
Article in English | MEDLINE | ID: mdl-26464984

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disease in which upper and lower motor neurons degenerate, leading to muscle atrophy, paralysis, and death within 3 to 5 years of onset. While a small percentage of ALS cases are genetically linked, the majority are sporadic with unknown origin. Currently, etiological links are associated with disease onset without mechanistic understanding. Of all the putative risk factors, however, head trauma has emerged as a consistent candidate for initiating the molecular cascades of ALS. Here, we test the hypothesis that traumatic brain injury (TBI) in the SOD1 (G93A) transgenic rat model of ALS leads to early disease onset and shortened lifespan. We demonstrate, however, that a one-time acute focal injury caused by controlled cortical impact does not affect disease onset or survival. Establishing the negligible involvement of a single acute focal brain injury in an ALS rat model increases the current understanding of the disease. Critically, untangling a single focal TBI from multiple mild injuries provides a rationale for scientists and physicians to increase focus on repeat injuries to hopefully pinpoint a contributing cause of ALS.

7.
J Neurosci ; 34(47): 15587-600, 2014 Nov 19.
Article in English | MEDLINE | ID: mdl-25411487

ABSTRACT

Sporadic amyotrophic lateral sclerosis (ALS) is a fatal disease with unknown etiology, characterized by a progressive loss of motor neurons leading to paralysis and death typically within 3-5 years of onset. Recently, there has been remarkable progress in understanding inherited forms of ALS in which well defined mutations are known to cause the disease. Rodent models in which the superoxide dismutase-1 (SOD1) mutation is overexpressed recapitulate hallmark signs of ALS in patients. Early anatomical changes in mouse models of fALS are seen in the neuromuscular junctions (NMJs) and lower motor neurons, and selective reduction of toxic mutant SOD1 in the spinal cord and muscle of these models has beneficial effects. Therefore, much of ALS research has focused on spinal motor neuron and NMJ aspects of the disease. Here we show that, in the SOD1(G93A) rat model of ALS, spinal motor neuron loss occurs presymptomatically and before degeneration of ventral root axons and denervation of NMJs. Although overt cell death of corticospinal motor neurons does not occur until disease endpoint, we wanted to establish whether the upper motor neuron might still play a critical role in disease progression. Surprisingly, the knockdown of mutant SOD1 in only the motor cortex of presymptomatic SOD1(G93A) rats through targeted delivery of AAV9-SOD1-shRNA resulted in a significant delay of disease onset, expansion of lifespan, enhanced survival of spinal motor neurons, and maintenance of NMJs. This datum suggests an early dysfunction and thus an important role of the upper motor neuron in this animal model of ALS and perhaps patients with the disease.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Motor Cortex/enzymology , Motor Cortex/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase/physiology , Amyotrophic Lateral Sclerosis/mortality , Animals , Cell Death/drug effects , Female , Gene Knockdown Techniques , Herpesvirus 1, Suid/genetics , Humans , Male , Mice , Neuromuscular Junction/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Superoxide Dismutase-1 , Transfection
8.
J Vis Exp ; (88)2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24962813

ABSTRACT

A cell expansion technique to amass large numbers of cells from a single specimen for research experiments and clinical trials would greatly benefit the stem cell community. Many current expansion methods are laborious and costly, and those involving complete dissociation may cause several stem and progenitor cell types to undergo differentiation or early senescence. To overcome these problems, we have developed an automated mechanical passaging method referred to as "chopping" that is simple and inexpensive. This technique avoids chemical or enzymatic dissociation into single cells and instead allows for the large-scale expansion of suspended, spheroid cultures that maintain constant cell/cell contact. The chopping method has primarily been used for fetal brain-derived neural progenitor cells or neurospheres, and has recently been published for use with neural stem cells derived from embryonic and induced pluripotent stem cells. The procedure involves seeding neurospheres onto a tissue culture Petri dish and subsequently passing a sharp, sterile blade through the cells effectively automating the tedious process of manually mechanically dissociating each sphere. Suspending cells in culture provides a favorable surface area-to-volume ratio; as over 500,000 cells can be grown within a single neurosphere of less than 0.5 mm in diameter. In one T175 flask, over 50 million cells can grow in suspension cultures compared to only 15 million in adherent cultures. Importantly, the chopping procedure has been used under current good manufacturing practice (cGMP), permitting mass quantity production of clinical-grade cell products.


Subject(s)
Brain/cytology , Neural Stem Cells/cytology , Pluripotent Stem Cells/cytology , Brain/embryology , Cell Aggregation/physiology , Cell Communication/physiology , Cell Growth Processes/physiology , Cytological Techniques/methods , Humans
9.
Exp Neurol ; 262 Pt B: 127-37, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24613827

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disorder that is characterized by progressive degeneration of motor neurons in the cortex, brainstem and spinal cord. This leads to paralysis, respiratory insufficiency and death within an average of 3 to 5 years from disease onset. While the genetics of ALS are becoming more understood in familial cases, the mechanisms underlying disease pathology remain unclear and there are no effective treatment options. Without understanding what causes ALS it is difficult to design treatments. However, in recent years stem cell transplantation has emerged as a potential new therapy for ALS patients. While motor neuron replacement remains a focus of some studies trying to treat ALS with stem cells, there is more rationale for using stem cells as support cells for dying motor neurons as they are already connected to the muscle. This could be through reducing inflammation, releasing growth factors, and other potential less understood mechanisms. Prior to moving into patients, stringent pre-clinical studies are required that have at least some rationale and efficacy in animal models and good safety profiles. However, given our poor understanding of what causes ALS and whether stem cells may ameliorate symptoms, there should be a push to determine cell safety in pre-clinical models and then a quick translation to the clinic where patient trials will show if there is any efficacy. Here, we provide a critical review of current clinical trials using either mesenchymal or neural stem cells to treat ALS patients. Pre-clinical data leading to these trials, as well as those in development are also evaluated in terms of mechanisms of action, validity of conclusions and rationale for advancing stem cell treatment strategies for this devastating disorder.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , Clinical Trials as Topic , Stem Cell Transplantation/methods , Stem Cell Transplantation/trends , Animals , Clinical Trials as Topic/history , Clinical Trials as Topic/methods , Clinical Trials as Topic/trends , History, 20th Century , History, 21st Century , Humans
10.
J Comp Neurol ; 522(12): 2707-28, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24610630

ABSTRACT

Transplantation of human neural progenitor cells (NPCs) into the brain or spinal cord to replace lost cells, modulate the injury environment, or create a permissive milieu to protect and regenerate host neurons is a promising therapeutic strategy for neurological diseases. Deriving NPCs from human fetal tissue is feasible, although problematic issues include limited sources and ethical concerns. Here we describe a new and abundant source of NPCs derived from human induced pluripotent stem cells (iPSCs). A novel chopping technique was used to transform adherent iPSCs into free-floating spheres that were easy to maintain and were expandable (EZ spheres) (Ebert et al. [2013] Stem Cell Res 10:417-427). These EZ spheres could be differentiated towards NPC spheres with a spinal cord phenotype using a combination of all-trans retinoic acid (RA) and epidermal growth factor (EGF) and fibroblast growth factor-2 (FGF-2) mitogens. Suspension cultures of NPCs derived from human iPSCs or fetal tissue have similar characteristics, although they were not similar when grown as adherent cells. In addition, iPSC-derived NPCs (iNPCs) survived grafting into the spinal cord of athymic nude rats with no signs of overgrowth and with a very similar profile to human fetal-derived NPCs (fNPCs). These results suggest that human iNPCs behave like fNPCs and could thus be a valuable alternative for cellular regenerative therapies of neurological diseases.


Subject(s)
Cell Differentiation/physiology , Cell Movement/physiology , Induced Pluripotent Stem Cells/physiology , Neural Stem Cells/physiology , Spinal Cord/cytology , Analysis of Variance , Animals , Aquaporin 4/metabolism , Astrocytes/physiology , Humans , Ki-67 Antigen/metabolism , Nerve Tissue Proteins/metabolism , Neural Stem Cells/transplantation , Neurons/metabolism , Rats , Rats, Nude , Spinal Cord/surgery , Transcriptome/physiology
11.
Neuroreport ; 25(6): 367-72, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24284956

ABSTRACT

Human neural progenitor cells (hNPCs) derived from the fetal cortex can be expanded in vitro and genetically modified through lentiviral transduction to secrete growth factors shown to have a neurotrophic effect in animal models of neurological disease. hNPCs survive and mature following transplantation into the central nervous system of large and small animals including the rat model of amyotrophic lateral sclerosis. Here we report that hNPCs engineered to express glial cell line-derived neurotrophic factor (GDNF) survive long-term (7.5 months) following transplantation into the spinal cord of athymic nude rats and continue to secrete GDNF. Cell proliferation declined while the number of astrocytes increased, suggesting final maturation of the cells over time in vivo. Together these data show that GDNF-producing hNPCs may be useful as a source of cells for long-term delivery of both astrocytes and GDNF to the damaged central nervous system.


Subject(s)
Fetal Stem Cells/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Graft Survival/physiology , Neural Stem Cells/metabolism , Spinal Cord/transplantation , Animals , Cell Differentiation/physiology , Cell Line , Cell Survival/physiology , Fetal Stem Cells/transplantation , Glial Fibrillary Acidic Protein/metabolism , Humans , Male , Neural Stem Cells/transplantation , Rats , Rats, Nude , Spinal Cord/pathology
12.
Mol Ther ; 21(8): 1602-10, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23712039

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive loss of motor neurons in the brain and spinal cord. We have recently shown that human mesenchymal stem cells (hMSCs) modified to release glial cell line-derived neurotrophic factor (GDNF) decrease disease progression in a rat model of ALS when delivered to skeletal muscle. In the current study, we determined whether or not this effect could be enhanced by delivering GDNF in concert with other trophic factors. hMSC engineered to secrete GDNF (hMSC-GDNF), vascular endothelial growth factor (hMSC-VEGF), insulin-like growth factor-I (hMSC-IGF-I), or brain-derived neurotrophic factor (hMSC-BDNF), were prepared and transplanted bilaterally into three muscle groups. hMSC-GDNF and hMSC-VEGF prolonged survival and slowed the loss of motor function, but hMSC-IGF-I and hMSC-BDNF did not have any effect. We then tested the efficacy of a combined ex vivo delivery of GDNF and VEGF in extending survival and protecting neuromuscular junctions (NMJs) and motor neurons. Interestingly, the combined delivery of these neurotrophic factors showed a strong synergistic effect. These studies further support ex vivo gene therapy approaches for ALS that target skeletal muscle.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Glial Cell Line-Derived Neurotrophic Factor/genetics , Longevity/genetics , Vascular Endothelial Growth Factor A/genetics , Amyotrophic Lateral Sclerosis/mortality , Animals , Cell Survival/genetics , Disease Models, Animal , Disease Progression , Female , Gene Expression , Gene Transfer Techniques , Genetic Therapy , Humans , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Motor Neurons/metabolism , Muscle, Skeletal/metabolism , Neuromuscular Junction/metabolism , Rats
13.
Am J Respir Crit Care Med ; 187(5): 535-42, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23220913

ABSTRACT

RATIONALE: Amyotrophic lateral sclerosis (ALS) is a devastating motor neuron disease causing paralysis and death from respiratory failure. Strategies to preserve and/or restore respiratory function are critical for successful treatment. Although breathing capacity is maintained until late in disease progression in rodent models of familial ALS (SOD1(G93A) rats and mice), reduced numbers of phrenic motor neurons and decreased phrenic nerve activity are observed. Decreased phrenic motor output suggests imminent respiratory failure. OBJECTIVES: To preserve or restore phrenic nerve activity in SOD1(G93A) rats at disease end stage. METHODS: SOD1(G93A) rats were injected with human neural progenitor cells (hNPCs) bracketing the phrenic motor nucleus before disease onset, or exposed to acute intermittent hypoxia (AIH) at disease end stage. MEASUREMENTS AND MAIN RESULTS: The capacity to generate phrenic motor output in anesthetized rats at disease end stage was: (1) transiently restored by a single presentation of AIH; and (2) preserved ipsilateral to hNPC transplants made before disease onset. hNPC transplants improved ipsilateral phrenic motor neuron survival. CONCLUSIONS: AIH-induced respiratory plasticity and stem cell therapy have complementary translational potential to treat breathing deficits in patients with ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , Respiratory Insufficiency/prevention & control , Respiratory Therapy/methods , Stem Cell Transplantation , Animals , Brain-Derived Neurotrophic Factor/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Hypoxia , Inspiratory Capacity , Male , Motor Neurons/metabolism , Phrenic Nerve/metabolism , Phrenic Nerve/physiopathology , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Superoxide Dismutase
14.
PLoS One ; 7(4): e34932, 2012.
Article in English | MEDLINE | ID: mdl-22523565

ABSTRACT

Proliferation of glia and immune cells is a common pathological feature of many neurodegenerative diseases including amyotrophic lateral sclerosis (ALS). Here, to investigate the role of proliferating cells in motor neuron disease, SOD1(G93A) transgenic mice were treated intracerebroventicularly (i.c.v.) with the anti-mitotic drug cytosine arabinoside (Ara-C). I.c.v. delivery of Ara-C accelerated disease progression in SOD1(G93A) mouse model of ALS. Ara-C treatment caused substantial decreases in the number of microglia, NG2+ progenitors, Olig2+ cells and CD3+ T cells in the lumbar spinal cord of symptomatic SOD1(G93A) transgenic mice. Exacerbation of disease was also associated with significant alterations in the expression inflammatory molecules IL-1ß, IL-6, TGF-ß and the growth factor IGF-1.


Subject(s)
Motor Neuron Disease/complications , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Proliferation/drug effects , Cytarabine/pharmacology , Disease Models, Animal , Disease Progression , Humans , Insulin-Like Growth Factor I/metabolism , Interleukin-6/metabolism , Mice , Mice, Transgenic , Microglia/drug effects , Transforming Growth Factor beta/metabolism
15.
Neurotherapeutics ; 8(4): 591-606, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21904789

ABSTRACT

Motor neuron degeneration leading to muscle atrophy and death is a pathological hallmark of disorders, such as amyotrophic lateral sclerosis or spinal muscular atrophy. No effective treatment is available for these devastating diseases. At present, cell-based therapies targeting motor neuron replacement, support, or as a vehicle for the delivery of neuroprotective molecules are being investigated. Although many challenges and questions remain, the beneficial effects observed following transplantation therapy in animal models of motor neuron disease has sparked hope and a number of clinical trials. Here, we provide a comprehensive review of cell-based therapeutics for motor neuron disorders, with a particular emphasis on amyotrophic lateral sclerosis.


Subject(s)
Motor Neuron Disease/surgery , Neurons/physiology , Stem Cell Transplantation/methods , Animals , Humans , Mice , Stem Cell Transplantation/trends
16.
Neurobiol Dis ; 40(1): 245-50, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20573565

ABSTRACT

Approximately 10% of the cases of amyotrophic lateral sclerosis (ALS) are inherited, with the majority of identified linkages in the gene encoding Cu/Zn superoxide dismutase (SOD1). Recent studies showed that human wild-type SOD1 (SOD1(WT)) overexpression accelerated disease in mice expressing human SOD1 mutants linked to ALS. However, there is a controversy whether the exacerbation mechanism occurs through coaggregation of human SOD1(WT) with SOD1 mutants, stabilization by SOD1(WT) of toxic soluble SOD1 species, or conversion of SOD1(WT) into toxic species through oxidative damage. To further address whether the exacerbation of disease requires misfolding, modifications, and/or interaction of SOD1(WT) with pathogenic forms of SOD1 species, we have studied the effect of human SOD1(WT) overexpression in mice expressing the murine mutant Sod1(G86R). Surprisingly, unlike a previous report with SOD1(G85R) mice, SOD1(WT) overexpression did not affect the life span of Sod1(G86R) mice. Our analysis of spinal cord extracts revealed a lack of heterodimerization or aggregation between human SOD1(WT) and mouse Sod1(G86R) proteins. Moreover, there was no evidence of conversion of SOD1(WT) into misfolded or abnormal SOD1 isoforms based on immunoreactivity with monoclonal antibodies specific to misfolded forms of SOD1 mutants and on analysis of SOD1 isoforms after two-dimensional gel electrophoresis. We conclude that a direct interaction between wild type and mutant forms of SOD1 is required for exacerbation of ALS disease by SOD1(WT) protein.


Subject(s)
Disease Models, Animal , Motor Neuron Disease/enzymology , Motor Neuron Disease/genetics , Spinal Cord/enzymology , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics , Amino Acid Sequence , Animals , Base Sequence , Disease Progression , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Motor Neuron Disease/pathology , Mutation , Spinal Cord/pathology , Superoxide Dismutase/physiology , Superoxide Dismutase-1
17.
Exp Neurol ; 220(2): 267-75, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19733170

ABSTRACT

Macrophage colony stimulating factor (M-CSF) is a cytokine that regulates the survival, proliferation and maturation of microglial cells. Administration of M-CSF can promote neuronal survival in various models of central nervous system (CNS) injury. Here, in an attempt to induce a neuroprotective microglial cell phenotype and enhance motor neuron survival, mutant SOD1(G37R) transgenic mice were treated, weekly, with M-CSF starting at onset of disease. Unexpectedly, M-CSF accelerated disease progression in SOD1(G37R) mouse model of ALS. The shortened survival of M-CSF-treated animals was associated with diminished muscle innervation and enhanced adoption of a macrophage-like phenotype by microglial cells characterised by the upregulation of pro-inflammatory cytokines TNF-alpha and IL-1 beta and of the phagocytic marker CD68.


Subject(s)
Amyotrophic Lateral Sclerosis/chemically induced , Macrophage Colony-Stimulating Factor/pharmacology , Microglia/drug effects , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Immunohistochemistry , Interleukin-1beta/metabolism , Mice , Mice, Transgenic , Mutation/physiology , Peripheral Nerves/pathology , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/pathology , Tumor Necrosis Factor-alpha/metabolism
18.
J Neurosci ; 28(41): 10234-44, 2008 Oct 08.
Article in English | MEDLINE | ID: mdl-18842883

ABSTRACT

Microglial activation is a hallmark of all neurodegenerative diseases including amyotrophic lateral sclerosis (ALS). Here, a detailed characterization of the microglial cell population within the spinal cord of a mouse model of familial ALS was performed. Using flow cytometry, we detected three distinct microglial populations within the spinal cord of mice overexpressing mutant superoxide dismutase (SOD1): mature microglial cells (CD11b(+), CD45(low)), myeloid precursor cells (CD11b(+), CD45(int)), and macrophages (CD11b(+), CD45(high)). Characterization of cell proliferation within the CNS of SOD1(G93A) mice revealed that the expansion in microglial cell population is mainly attributable to the proliferation of myeloid precursor cells. To assess the contribution of proliferating microglia in motor neuron degeneration, we generated CD11b-TK(mut-30); SOD1(G93A) doubly transgenic mice that allow the elimination of proliferating microglia on administration of ganciclovir. Surprisingly, a 50% reduction in reactive microglia specifically in the lumbar spinal cord of CD11b-TK(mut-30); SOD1(G93A) doubly transgenic mice had no effect on motor neuron degeneration. This suggests that proliferating microglia-expressing mutant SOD1 are not central contributors of the neurodegenerative process in ALS caused by mutant SOD1.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Microglia/pathology , Motor Neurons/pathology , Mutation , Nerve Degeneration/pathology , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/complications , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Biomarkers/metabolism , CD11b Antigen/metabolism , Cell Count , Cell Proliferation , Cellular Senescence , Dendritic Cells/metabolism , Disease Progression , Mice , Mice, Transgenic , Microglia/immunology , Nerve Degeneration/etiology , Neuroglia/pathology , Phenotype , Spinal Cord/pathology , Stem Cells/pathology , Superoxide Dismutase-1 , T-Lymphocytes/pathology
19.
J Neurosci ; 28(38): 9363-76, 2008 Sep 17.
Article in English | MEDLINE | ID: mdl-18799670

ABSTRACT

The role of CD11b+ myeloid cells in axonal regeneration was assessed using axonal injury models and CD11b-TK(mt-30) mice expressing a mutated HSV-1 thymidine kinase (TK) gene regulated by the myeloid-specific CD11b promoter. Continuous delivery of ganciclovir at a sciatic nerve lesion site greatly decreased the number of granulocytes/inflammatory monocytes and macrophages in the distal stump of CD11b-TK(mt-30) mice. Axonal regeneration and locomotor function recovery were severely compromised in ganciclovir-treated CD11b-TK(mt-30) mice. This was caused by an unsuitable growth environment rather than an altered regeneration capacity of neurons. In absence of CD11b+ cells, the clearance of inhibitory myelin debris was prevented, neurotrophin synthesis was abolished, and blood vessel formation/maintenance was severely compromised in the sciatic nerve distal stump. Spinal cord-injured axons also failed to regenerate through peripheral nerve grafts in the absence of CD11b+ cells. Therefore, myeloid cells support axonal regeneration and functional recovery by creating a growth-permissive milieu for injured axons.


Subject(s)
CD11 Antigens/immunology , Growth Cones/immunology , Myeloid Cells/immunology , Nerve Regeneration/immunology , Animals , Antiviral Agents/pharmacology , Cells, Cultured , Disease Models, Animal , Female , Ganciclovir/pharmacology , Graft Survival/physiology , Granulocytes/drug effects , Granulocytes/immunology , Growth Cones/metabolism , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Knockout , Mice, Transgenic , Monocytes/drug effects , Monocytes/immunology , Motor Activity/drug effects , Motor Activity/physiology , Recovery of Function/drug effects , Recovery of Function/physiology , Sciatic Neuropathy/immunology , Sciatic Neuropathy/physiopathology , Thymidine Kinase/genetics , Thymidine Kinase/immunology , Tissue Transplantation
20.
J Neurochem ; 107(1): 253-64, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18680552

ABSTRACT

Mutations in the gigaxonin gene are responsible for giant axonal neuropathy (GAN), a progressive neurodegenerative disorder associated with abnormal accumulations of Intermediate Filaments (IFs). Gigaxonin is the substrate-specific adaptor for a new Cul3-E3-ubiquitin ligase family that promotes the proteasome dependent degradation of its partners MAP1B, MAP8 and tubulin cofactor B. Here, we report the generation of a mouse model with targeted deletion of Gan exon 1 (Gan(Deltaexon1;Deltaexon1)). Analyses of the Gan(Deltaexon1;Deltaexon1) mice revealed increased levels of various IFs proteins in the nervous system and the presence of IFs inclusion bodies in the brain. Despite deficiency of full length gigaxonin, the Gan(Deltaexon1;Deltaexon1) mice do not develop overt neurological phenotypes and giant axons reminiscent of the human GAN disease. Nonetheless, at 6 months of age the Gan(Deltaexon1;Deltaexon1) mice exhibit a modest hind limb muscle atrophy, a 10% decrease of muscle innervation and a 27% axonal loss in the L5 ventral roots. This new mouse model should provide a useful tool to test potential therapeutic approaches for GAN disease.


Subject(s)
Brain/metabolism , Cytoskeletal Proteins/deficiency , Muscular Atrophy/metabolism , Neurodegenerative Diseases/metabolism , Peripheral Nervous System Diseases/metabolism , Wallerian Degeneration/metabolism , Animals , Axons/metabolism , Axons/pathology , Brain/physiopathology , Cells, Cultured , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Embryonic Stem Cells , Exons/genetics , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Ganglia, Spinal/physiopathology , Gene Targeting/methods , Inclusion Bodies/genetics , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Intermediate Filament Proteins/metabolism , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Motor Neurons/metabolism , Motor Neurons/pathology , Muscle, Skeletal/innervation , Muscle, Skeletal/physiopathology , Muscular Atrophy/genetics , Muscular Atrophy/physiopathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/physiopathology , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/physiopathology , Phenotype , Spinal Nerve Roots/metabolism , Spinal Nerve Roots/pathology , Spinal Nerve Roots/physiopathology , Wallerian Degeneration/genetics , Wallerian Degeneration/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...