Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cell Reports ; 18(3): 672-687, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36764296

RESUMO

The adult spinal cord contains a population of ependymal-derived neural stem/progenitor cells (epNSPCs) that are normally quiescent, but are activated to proliferate, differentiate, and migrate after spinal cord injury. The mechanisms that regulate their response to injury cues, however, remain unknown. Here, we demonstrate that excitotoxic levels of glutamate promote the proliferation and astrocytic fate specification of adult spinal cord epNSPCs. We show that glutamate-mediated calcium influx through calcium-permeable alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors (CP-AMPARs) in concert with Notch signaling increases the proliferation of epNSPCs via pCREB, and induces astrocytic differentiation through Hes1 upregulation. Furthermore, the in vivo targeting of this pathway via positive modulation of AMPARs after spinal cord injury enhances epNSPC proliferation, astrogliogenesis, neurotrophic factor production and increases neuronal survival. Our study uncovers an important mechanism by which CP-AMPARs regulate the growth and phenotype of epNSPCs, which can be targeted therapeutically to harness the regenerative potential of these cells after injury.


Assuntos
Ácido Glutâmico , Traumatismos da Medula Espinal , Humanos , Ácido Glutâmico/metabolismo , Cálcio/metabolismo , Medula Espinal , Receptores de AMPA/metabolismo , Traumatismos da Medula Espinal/metabolismo , Proliferação de Células
2.
Neurobiol Dis ; 172: 105812, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-35810963

RESUMO

Spinal cord injury (SCI) elicits a cascade of degenerative events including cell death, axonal degeneration, and the upregulation of inhibitory molecules which limit repair. Repulsive guidance molecule A (RGMa) is an axon growth inhibitor which is also involved in neuronal cell death and differentiation. SCI causes upregulation of RGMa in the injured rodent, non-human primate, and human spinal cord. Recently, we showed that delayed administration of elezanumab, a high affinity human RGMa-specific monoclonal antibody, promoted neuroprotective and regenerative effects following thoracic SCI. Since most human traumatic SCI is at the cervical level, and level-dependent anatomical and molecular differences may influence pathophysiological responses to injury and treatment, we examined the efficacy of elezanumab and its therapeutic time window of administration in a clinically relevant rat model of cervical impact-compression SCI. Pharmacokinetic analysis of plasma and spinal cord tissue lysate showed comparable levels of RGMa antibodies with delayed administration following cervical SCI. At 12w after SCI, elezanumab promoted long term benefits including perilesional sparing of motoneurons and increased neuroplasticity of key descending pathways involved in locomotion and fine motor function. Elezanumab also promoted growth of corticospinal axons into spinal cord gray matter and enhanced serotonergic innervation of the ventral horn to form synaptic connections caudal to the cervical lesion. Significant recovery in grip and trunk/core strength, locomotion and gait, and spontaneous voiding ability was found in rats treated with elezanumab either immediately post-injury or at 3 h post-SCI, and improvements in specific gait parameters were found when elezanumab was delayed to 24 h post-injury. We also developed a new locomotor score, the Cervical Locomotor Score, a simple and sensitive measure of trunk/core and limb strength and stability during dynamic locomotion.


Assuntos
Medula Cervical , Traumatismos da Medula Espinal , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Medula Cervical/metabolismo , Proteínas Ligadas por GPI , Humanos , Proteínas de Membrana , Proteínas do Tecido Nervoso/metabolismo , Ratos , Recuperação de Função Fisiológica/fisiologia , Medula Espinal/patologia , Traumatismos da Medula Espinal/patologia
3.
Neurobiol Dis ; 150: 105259, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33434618

RESUMO

Neuronal regeneration in the injured central nervous system is hampered by multiple extracellular proteins. These proteins exert their inhibitory action through interactions with receptors that are located in cholesterol rich compartments of the membrane termed lipid rafts. Here we show that cholesterol-synthesis inhibition prevents the association of the Neogenin receptor with lipid rafts. Furthermore, we show that cholesterol-synthesis inhibition enhances axonal growth both on inhibitory -myelin and -RGMa substrates. Following optic nerve injury, lowering cholesterol synthesis with both drugs and siRNA-strategies allows for robust axonal regeneration and promotes neuronal survival. Cholesterol inhibition also enhanced photoreceptor survival in a model of Retinitis Pigmentosa. Our data reveal that Lovastatin leads to several opposing effects on regenerating axons: cholesterol synthesis inhibition promotes regeneration whereas altered prenylation impairs regeneration. We also show that the lactone prodrug form of lovastatin has differing effects on regeneration when compared to the ring-open hydroxy-acid form. Thus the association of cell surface receptors with lipid rafts contributes to axonal regeneration inhibition, and blocking cholesterol synthesis provides a potential therapeutic approach to promote neuronal regeneration and survival in the diseased Central Nervous System. SIGNIFICANCE STATEMENT: Statins have been intensively used to treat high levels of cholesterol in humans. However, the effect of cholesterol inhibition in both the healthy and the diseased brain remains controversial. In particular, it is unclear whether cholesterol inhibition with statins can promote regeneration and survival following injuries. Here we show that late stage cholesterol inhibition promotes robust axonal regeneration following optic nerve injury. We identified distinct mechanisms of action for activated vs non-activated Lovastatin that may account for discrepancies found in the literature. We show that late stage cholesterol synthesis inhibition alters Neogenin association with lipid rafts, thereby i) neutralizing the inhibitory function of its ligand and ii) offering a novel opportunity to promote CNS regeneration and survival following injuries.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Lovastatina/farmacologia , Regeneração Nervosa/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Nervo Óptico/efeitos dos fármacos , Animais , Anticolesterolemiantes/farmacologia , Axônios/efeitos dos fármacos , Axônios/patologia , Sobrevivência Celular , Embrião de Galinha , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Bainha de Mielina , Neurônios/metabolismo , Nervo Óptico/metabolismo , Nervo Óptico/patologia , Traumatismos do Nervo Óptico/metabolismo , Traumatismos do Nervo Óptico/patologia , Células Fotorreceptoras , Prenilação , Pró-Fármacos , Ratos , Retina , Retinose Pigmentar , Dicloridrato de trans-1,4-Bis(2-clorobenzaminometil)ciclo-hexano/farmacologia
4.
Neurobiol Dis ; 143: 104995, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32590037

RESUMO

Spinal cord injury (SCI) often results in permanent functional loss due to a series of degenerative events including cell death, axonal damage, and the upregulation of inhibitory proteins that impede regeneration. Repulsive Guidance Molecule A (RGMa) is a potent inhibitor of axonal growth that is rapidly upregulated following injury in both the rodent and human central nervous system (CNS). Previously, we showed that monoclonal antibodies that specifically block inhibitory RGMa signaling promote neuroprotective and regenerative effects when administered acutely in a clinically relevant rat model of thoracic SCI. However, it is unknown whether systemic administration of RGMa blocking antibodies are effective for SCI after delayed administration. Here, we administered elezanumab, a human monoclonal antibody targeting RGMa, intravenously either acutely or at 3 h or 24 h following thoracic clip impact-compression SCI. Rats treated with elezanumab acutely and at 3 h post-injury showed improvements in overground locomotion and fine motor function and gait. Rats treated 24 h post-SCI trended towards better recovery demonstrating significantly greater stride length and swing speed. Treated rats also showed greater tissue preservation with reduced lesion areas. As seen with acute treatment, delayed administration of elezanumab at 3 h post-SCI also increased perilesional neuronal sparing and serotonergic and corticospinal axonal plasticity. In addition, all elezanumab treated rats showed earlier spontaneous voiding ability and less post-trauma bladder wall hypertrophy. Together, our data demonstrate the therapeutic efficacy of delayed systemic administration of elezanumab in a rat model of SCI, and uncovers a new role for RGMa inhibition in bladder recovery following SCI.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Proteínas Ligadas por GPI/antagonistas & inibidores , Proteínas de Membrana/antagonistas & inibidores , Proteínas do Tecido Nervoso/antagonistas & inibidores , Recuperação de Função Fisiológica/efeitos dos fármacos , Traumatismos da Medula Espinal/fisiopatologia , Animais , Feminino , Humanos , Ratos , Ratos Wistar , Micção/efeitos dos fármacos
5.
Stem Cells ; 38(2): 187-194, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31648407

RESUMO

Nearly a century ago, the concept of the secondary injury in spinal cord trauma was first proposed to explain the complex cascade of molecular and cellular events leading to widespread neuronal and glial cell death after trauma. In recent years, it has been established that the ependymal region of the adult mammalian spinal cord contains a population of multipotent neural stem/progenitor cells (NSPCs) that are activated after spinal cord injury (SCI) and likely play a key role in endogenous repair and regeneration. How these cells respond to the various components of the secondary injury remains poorly understood. Emerging evidence suggests that many of the biochemical components of the secondary injury cascade which have classically been viewed as deleterious to host neuronal and glial cells may paradoxically trigger NSPC activation, proliferation, and differentiation thus challenging our current understanding of secondary injury mechanisms in SCI. Herein, we highlight new findings describing the response of endogenous NSPCs to spinal cord trauma, redefining the secondary mechanisms of SCI through the lens of the endogenous population of stem/progenitor cells. Moreover, we outline how these insights can fuel novel stem cell-based therapeutic strategies to repair the injured spinal cord.


Assuntos
Células-Tronco Neurais/metabolismo , Traumatismos da Medula Espinal/fisiopatologia , Humanos
6.
Neuroscientist ; 25(1): 8-21, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-29283022

RESUMO

The ideal biomarker for central nervous system (CNS) trauma in patients would be a molecular marker specific for injured nervous tissue that would provide a consistent and reliable assessment of the presence and severity of injury and the prognosis for recovery. One candidate biomarker is the protein tau, a microtubule-associated protein abundant in the axonal compartment of CNS neurons. Following axonal injury, tau becomes modified primarily by hyperphosphorylation of its various amino acid residues and cleavage into smaller fragments. These posttrauma products can leak into the cerebrospinal fluid or bloodstream and become candidate biomarkers of CNS injury. This review examines the primary molecular changes that tau undergoes following traumatic brain injury and spinal cord injury, and reviews the current literature in traumatic CNS biomarker research with a focus on the potential for hyperphosphorylated and cleaved tau as sensitive biomarkers of injury.


Assuntos
Lesões Encefálicas Traumáticas/diagnóstico , Lesões Encefálicas Traumáticas/metabolismo , Processamento de Proteína Pós-Traducional , Traumatismos da Medula Espinal/diagnóstico , Traumatismos da Medula Espinal/metabolismo , Proteínas tau/metabolismo , Animais , Biomarcadores/sangue , Biomarcadores/líquido cefalorraquidiano , Humanos , Fosforilação
7.
J Neurotrauma ; 35(16): 1929-1941, 2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-29644915

RESUMO

Current biomarker research in spinal cord injury (SCI) and traumatic brain injury has focused on a number of structural protein candidates, including the microtubule-associated protein tau. Evidence from models of traumatic brain injury has demonstrated that hyperphosphorylation of tau (p-tau) occurs in injured axons and demonstrates its utility as a biomarker for brain injury; however, the potential of p-tau as a biomarker for SCI is not yet known. Therefore, the present study determined whether tau is hyperphosphorylated in injured spinal cord axons, and then examined cerebrospinal fluid (CSF) and serum concentrations of p-tau and total-tau protein after a clinically relevant severe impact-compression SCI in rats. We found that severe SCI at T8 showed the presence of p-tau in damaged axons with a similar time course and distribution pattern to ß-APP, a biomarker of axonal injury. The presence of p-tau and ß-APP positive axons extended no farther than 5000 µm rostral and caudal to the injury epicenter, and was at its maximum at one day post-SCI. CSF levels of p-tau and total-tau significantly increased at one day post-SCI; however, only serum p-tau levels were significantly elevated in rats with SCI compared with naïve rats. These results suggest that CSF and serum p-tau may be a useful biomarker for severe traumatic SCI.


Assuntos
Biomarcadores/análise , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Proteínas tau/análise , Animais , Axônios/metabolismo , Axônios/patologia , Feminino , Fosforilação , Ratos , Ratos Wistar , Proteínas tau/metabolismo
8.
Stem Cells Dev ; 26(23): 1675-1681, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28950762

RESUMO

Transplantation of neural stem/progenitor cells (NSPCs) following spinal cord injury (SCI) is a promising strategy to enhance regeneration but is limited by poor survival of grafted cells. Determining methods to enhance survival of NSPCs is therefore essential. Positive modulation of AMPA receptors has been shown to enhance neurogenesis in various models of brain injury. Here we examined the effect of selective AMPA receptor modulation in adult rat spinal cord-derived NSPCs using a class of allosteric AMPA receptor modulators known as ampakines. NSPCs from the periventricular region of the adult rat spinal cord were treated with ampakines CX614 and CX546 for 72 h either alone or in the presence of low-dose glutamate (50 µM). Treatment with either agent in the presence of glutamate significantly increased cell survival and proliferation and reduced cell death. Moreover, ampakine/glutamate treatment reduced cell death in the setting of oxidative stress. Treatment with ampakines did not significantly alter cell phenotype. These findings offer important insight into a potential therapeutic strategy to positively regulate transplanted and endogenous adult spinal cord-derived NSPCs after SCI.


Assuntos
Morte Celular , Proliferação de Células , Dioxóis/farmacologia , Células-Tronco Neurais/efeitos dos fármacos , Oxazinas/farmacologia , Piperidinas/farmacologia , Receptores de AMPA/metabolismo , Medula Espinal/citologia , Animais , Células Cultivadas , Feminino , Ácido Glutâmico/farmacologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/fisiologia , Estresse Oxidativo , Ratos , Ratos Wistar
9.
Sci Rep ; 7(1): 10529, 2017 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-28874746

RESUMO

Traumatic spinal cord injury (SCI) causes a cascade of degenerative events including cell death, axonal damage, and the upregulation of inhibitory molecules which prevent regeneration and limit recovery. Repulsive guidance molecule A (RGMa) is a potent neurite growth inhibitor in the central nervous system, exerting its repulsive activity by binding the Neogenin receptor. Here, we show for the first time that inhibitory RGMa is markedly upregulated in multiple cell types after clinically relevant impact-compression SCI in rats, and importantly, also in the injured human spinal cord. To neutralize inhibitory RGMa, clinically relevant human monoclonal antibodies were systemically administered after acute SCI, and were detected in serum, cerebrospinal fluid, and in the injured tissue. Rats treated with RGMa blocking antibodies showed significantly improved recovery of motor function and gait. Furthermore, RGMa blocking antibodies promoted neuronal survival, and enhanced the plasticity of descending serotonergic pathways and corticospinal tract axonal regeneration. RGMa antibody also attenuated neuropathic pain responses, which was associated with fewer activated microglia and reduced CGRP expression in the dorsal horn caudal to the lesion. These results show the therapeutic potential of the first human RGMa antibody for SCI and uncovers a new role for the RGMa/Neogenin pathway on neuropathic pain.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/uso terapêutico , Proteínas de Membrana/imunologia , Proteínas do Tecido Nervoso/imunologia , Neuralgia/terapia , Traumatismos da Medula Espinal/terapia , Regeneração da Medula Espinal , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Neutralizantes/administração & dosagem , Células Cultivadas , Feminino , Proteínas Ligadas por GPI , Humanos , Camundongos , Plasticidade Neuronal , Ratos , Ratos Wistar
10.
J Neurotrauma ; 34(6): 1209-1226, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-27775474

RESUMO

The immune system plays a critical and complex role in the pathobiology of spinal cord injury (SCI), exerting both beneficial and detrimental effects. Increasing evidence suggests that there are injury level-dependent differences in the immune response to SCI. Patients with traumatic SCI have elevated levels of circulating autoantibodies against components of the central nervous system, but the role of these antibodies in SCI outcomes remains unknown. In rodent models of mid-thoracic SCI, antibody-mediated autoimmunity appears to be detrimental to recovery. However, whether autoantibodies against the spinal cord are generated following cervical SCI (cSCI), the most common level of injury in humans, remains undetermined. To address this knowledge gap, we investigated the antibody responses following cSCI in a rat model of injury. We found increased immunoglobulin G (IgG) and IgM antibodies in the spinal cord in the subacute phase of injury (2 weeks), but not in more chronic phases (10 and 20 weeks). At 2 weeks post-cSCI, antibodies were detected at the injury epicenter and co-localized with the astroglial scar and neurons of the ventral horn. These increased levels of antibodies corresponded with enhanced activation of immune responses in the spleen. Higher counts of antibody-secreting cells were observed in the spleen of injured rats. Further, increased levels of secreted IgG antibodies and enhanced proliferation of T-cells in splenocyte cultures from injured rats were found. These findings suggest the potential development of autoantibody responses following cSCI in the rat. The impact of the post-traumatic antibody responses on functional outcomes of cSCI is a critical topic that requires further investigation.


Assuntos
Autoanticorpos/imunologia , Medula Cervical/lesões , Traumatismos da Medula Espinal/imunologia , Animais , Células Produtoras de Anticorpos/imunologia , Astrócitos/imunologia , Modelos Animais de Doenças , Feminino , Ratos , Ratos Wistar , Baço/imunologia
11.
Stem Cells Dev ; 25(16): 1223-33, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27316370

RESUMO

Traumatic spinal cord injury (SCI) leads to a cascade of secondary chemical insults, including oxidative stress and glutamate excitotoxicity, which damage host neurons and glia. Transplantation of exogenous neural stem/progenitor cells (NSPCs) has shown promise in enhancing regeneration after SCI, although survival of transplanted cells remains poor. Understanding the response of NSPCs to the chemical mediators of secondary injury is essential in finding therapies to enhance survival. We examined the in vitro effects of glutamate and glutamate receptor agonists on adult rat spinal cord-derived NSPCs. NSPCs isolated from the periventricular region of the adult rat spinal cord were exposed to various concentrations of glutamate for 96 h. We found that glutamate treatment (500 µM) for 96 h significantly increased live cell numbers, reduced cell death, and increased proliferation, but did not significantly alter cell phenotype. Concurrent glutamate treatment (500 µM) in the setting of H2O2 exposure (500 µM) for 10 h increased NSPC survival compared to H2O2 exposure alone. The effects of glutamate on NSPCs were blocked by the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)/kainate receptor antagonist GYKI-52466, but not by the N-methyl-D-aspartic acid receptor antagonist MK-801 or DL-AP5, or the mGluR3 antagonist LY-341495. Furthermore, treatment of NSPCs with AMPA, kainic acid, or the kainate receptor-specific agonist (RS)-2-amino-3-(3-hydroxy-5-tert-butylisoxazol-4-yl)propanoic acid mimicked the responses seen with glutamate both alone and in the setting of oxidative stress. These findings offer important insights into potential mechanisms to enhance NSPC survival and implicate a potential role for glutamate in promoting NSPC survival and proliferation after traumatic SCI.


Assuntos
Células-Tronco Adultas/citologia , Ácido Glutâmico/farmacologia , Células-Tronco Neurais/citologia , Estresse Oxidativo/efeitos dos fármacos , Receptores Ionotrópicos de Glutamato/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Medula Espinal/citologia , Células-Tronco Adultas/efeitos dos fármacos , Células-Tronco Adultas/metabolismo , Envelhecimento , Animais , Encéfalo/citologia , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Fenótipo , Ratos Transgênicos , Ratos Wistar , Receptores de AMPA/metabolismo , Receptores Ionotrópicos de Glutamato/antagonistas & inibidores , Receptores de Ácido Caínico/metabolismo
12.
Int J Dev Neurosci ; 47(Pt B): 140-6, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26390954

RESUMO

Riluzole, a sodium/glutamate antagonist, has shown significant neuroprotective effects in experimental models of spinal cord injury (SCI) and is currently under clinical trial for patients with SCI. However, the effect of riluzole on adult spinal cord-derived NSPCs remains unknown. In this study, we examined the effects of riluzole on NSPC survival both in vitro and in vivo. NSPCs harvested from the adult rat spinal cord were exposed to riluzole (1-30 µM) either alone or in combination with hydrogen peroxide or glutamate in vitro. Measures of intracellular reactive oxygen species (ROS), cell viability and proliferation were assessed. To examine the effects of riluzole on transplanted NSPCs in vivo, a rodent clip compression model of SCI was used. One week after injury, NSPCs were transplanted into the spinal cord and rats received either riluzole or vehicle treatment for two weeks (similar to the clinically accepted dosing regimen) at which time cords were processed for analysis. Exposure to riluzole (≥ 10 µM) for more than 48 h in vitro reduced NSPC viability. Riluzole treatment (1-10 µM) did not significantly affect intracellular ROS levels or cell viability in the setting of in vitro oxidative stress. While glutamate (500 µM) exposure for 96 h significantly increased adult NSPC proliferation and survival, this response was not blocked by concurrent treatment with riluzole (1-10 µM) thus supporting the notion that the known anti-glutamatergic properties of riluzole are not mediated through direct inhibition of glutamate receptors. Furthermore, riluzole treatment did not impair the survival of transplanted NSPCs in a rodent model of SCI. These results suggest that although NSPCs may have a narrow tolerance to riluzole treatment in vitro, riluzole does not impair NSPC survival at doses that would be used clinically.


Assuntos
Antagonistas de Aminoácidos Excitatórios/farmacologia , Células-Tronco Neurais/efeitos dos fármacos , Riluzol/farmacologia , Medula Espinal/citologia , Animais , Contagem de Células , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Ácido Glutâmico/farmacologia , Hidroliases/metabolismo , Peróxido de Hidrogênio/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Oxidantes/farmacologia , Ratos , Ratos Transgênicos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo
13.
Biores Open Access ; 4(1): 146-59, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26309791

RESUMO

Transplantation of neural stem/progenitor cells (NSPCs) is a promising strategy in spinal cord injury (SCI). However, poor survival of transplanted stem cells remains a major limitation of this therapy due to the hostile environment of the injured cord. Oxidative stress is a hallmark in the pathogenesis of SCI; however, its effects on NSPCs from the adult spinal cord have yet to be examined. We therefore developed in vitro models of mild and severe oxidative stress of adult spinal cord-derived NSPCs and used these models to examine potential cell survival factors. NSPCs harvested from the adult rat spinal cord were treated with hydrogen peroxide (H2O2) in vitro to induce oxidative stress. A mild 4 h exposure to H2O2 (500 µM) significantly increased the level of intracellular reactive oxygen species with minimal effect on viability. In contrast, 24 h of oxidative stress led to a marked reduction in cell survival. Pretreatment with brain-derived neurotrophic factor (BDNF) for 48 h attenuated the increase in intracellular reactive oxygen species and enhanced survival. This survival effect was associated with a significant reduction in the number of apoptotic cells and a significant increase in the activity of the antioxidant enzymes glutathione reductase and superoxide dismutase. BDNF treatment had no effect on NSPC differentiation or proliferation. In contrast, cyclosporin A and thyrotropin-releasing hormone had minimal or no effect on NSPC survival. Thus, these models of in vitro oxidative stress may be useful for screening neuroprotective factors administered prior to transplantation to enhance survival of stem cell transplants.

14.
Exp Neurol ; 269: 154-68, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25902036

RESUMO

Large animal and primate models of spinal cord injury (SCI) are being increasingly utilized for the testing of novel therapies. While these represent intermediary animal species between rodents and humans and offer the opportunity to pose unique research questions prior to clinical trials, the role that such large animal and primate models should play in the translational pipeline is unclear. In this initiative we engaged members of the SCI research community in a questionnaire and round-table focus group discussion around the use of such models. Forty-one SCI researchers from academia, industry, and granting agencies were asked to complete a questionnaire about their opinion regarding the use of large animal and primate models in the context of testing novel therapeutics. The questions centered around how large animal and primate models of SCI would be best utilized in the spectrum of preclinical testing, and how much testing in rodent models was warranted before employing these models. Further questions were posed at a focus group meeting attended by the respondents. The group generally felt that large animal and primate models of SCI serve a potentially useful role in the translational pipeline for novel therapies, and that the rational use of these models would depend on the type of therapy and specific research question being addressed. While testing within these models should not be mandatory, the detection of beneficial effects using these models lends additional support for translating a therapy to humans. These models provides an opportunity to evaluate and refine surgical procedures prior to use in humans, and safety and bio-distribution in a spinal cord more similar in size and anatomy to that of humans. Our results reveal that while many feel that these models are valuable in the testing of novel therapies, important questions remain unanswered about how they should be used and how data derived from them should be interpreted.


Assuntos
Traumatismos da Medula Espinal , Pesquisa Translacional Biomédica , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos , Modelos Animais de Doenças , Grupos Focais , Humanos , Primatas , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/terapia , Inquéritos e Questionários , Pesquisa Translacional Biomédica/métodos
15.
Cell Rep ; 8(4): 1146-59, 2014 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-25127134

RESUMO

Ideal strategies to ameliorate CNS damage should promote both neuronal survival and axon regeneration. The receptor Neogenin promotes neuronal apoptosis. Its ligand prevents death, but the resulting repulsive guidance molecule a (RGMa)-Neogenin interaction also inhibits axonal growth, countering any prosurvival benefits. Here, we explore strategies to inhibit Neogenin, thus simultaneously enhancing survival and regeneration. We show that bone morphogenetic protein (BMP) and RGMa-dependent recruitment of Neogenin into lipid rafts requires an interaction between RGMa and Neogenin subdomains. RGMa or Neogenin peptides that prevent this interaction, BMP inhibition by Noggin, or reduction of membrane cholesterol all block Neogenin raft localization, promote axon outgrowth, and prevent neuronal apoptosis. Blocking Neogenin raft association influences axonal pathfinding, enhances survival in the developing CNS, and promotes survival and regeneration in the injured adult optic nerve and spinal cord. Moreover, lowering cholesterol disrupts rafts and restores locomotor function after spinal cord injury. These data reveal a unified strategy to promote both survival and regeneration in the CNS.


Assuntos
Microdomínios da Membrana/fisiologia , Regeneração Nervosa , Animais , Axônios/fisiologia , Galinhas , Feminino , Cones de Crescimento/fisiologia , Macrolídeos , Proteínas de Membrana/metabolismo , Neurônios Motores/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Nervo Óptico/patologia , Nervo Óptico/fisiopatologia , Ratos Wistar , Células Ganglionares da Retina/fisiologia , Medula Espinal/metabolismo , Medula Espinal/patologia , Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/fisiopatologia
16.
Int J Dev Neurosci ; 31(7): 701-13, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23928260

RESUMO

Spinal cord injury (SCI) is a debilitating condition often resulting in paralysis, yet currently there is no effective treatment. Stem cell transplantation is a promising therapeutic strategy for promoting tissue repair after SCI. Stem cells offer a renewable source of cells with inherent plasticity for tissue regeneration. Neural stem/progenitor cells (NSPCs) are multipotent cells that self-renew and are committed to the neural lineage, and thus, they are especially suited to SCI repair. NSPCs may differentiate into neural cells after transplantation into the injured spinal cord, replacing lost or damaged cells, providing trophic support, restoring connectivity, and facilitating regeneration. Here, we review experimental studies and considerations for clinical translation of NSPC transplantation for SCI.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Neurais/fisiologia , Células-Tronco Neurais/transplante , Traumatismos da Medula Espinal/cirurgia , Animais , Humanos
17.
Biomaterials ; 34(15): 3775-83, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23465486

RESUMO

Traumatic injury to the spinal cord causes cell death, demyelination, axonal degeneration, and cavitation resulting in functional motor and sensory loss. Stem cell therapy is a promising approach for spinal cord injury (SCI); however, this strategy is currently limited by the poor survival and uncontrolled differentiation of transplanted stem cells. In an attempt to achieve greater survival and integration with the host tissue, we examined the survival and efficacy of adult brain-derived neural stem/progenitor cells (NSPCs) injected within a hydrogel blend of hyaluronan and methyl cellulose (HAMC) into a subacute, clinically relevant model of rat SCI. Prior to use, HAMC was covalently modified with recombinant rat platelet-derived growth factor-A (rPDGF-A) to promote oligodendrocytic differentiation. SCI rats transplanted with NSPCs in HAMC-rPDGF-A showed improved behavioral recovery compared to rats transplanted with NSPCs in media. Rats with NSPC/HAMC-rPDGF-A transplants had a significant reduction in cavitation, improved graft survival, increased oligodendrocytic differentiation, and sparing of perilesional host oligodendrocytes and neurons. These data suggest that HAMC-rPDGF-A is a promising vehicle for cell delivery to the injured spinal cord.


Assuntos
Ácido Hialurônico/química , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Células-Tronco Neurais/citologia , Traumatismos da Medula Espinal/terapia , Transplante de Células-Tronco , Cicatrização , Animais , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Sobrevivência de Enxerto/efeitos dos fármacos , Inflamação/patologia , Metilcelulose/química , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Oligodendroglia/citologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Fator de Crescimento Derivado de Plaquetas , Ratos , Ratos Wistar , Recuperação de Função Fisiológica/efeitos dos fármacos , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Cicatrização/efeitos dos fármacos
18.
Stem Cells Dev ; 22(3): 359-73, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22900481

RESUMO

Although transplantation of neural stem/progenitor cells (NSPC) encourages regeneration and repair after spinal cord injury (SCI), the survival of transplanted NSPC is limited. Ephrin-B3 has been shown to reduce the death of endogenous NSPC in the subventricular zone of the mouse brain without inducing uncontrolled proliferation. Due to similarities in the environment of the brain and spinal cord, we hypothesized that ephrin-B3 might reduce the death of both transplanted and endogenous spinal cord-derived NSPC. Both normal and injured (26 g clip compression) spinal cords were examined. Ephrin-B3-Fc was tested, and Fc fragments and phosphate-buffered saline (PBS) were used as controls. We found that EphA4 receptors were expressed by spinal cord-derived NSPC and expressed in the normal and injured rat spinal cord (higher expression in the latter). In vitro, ephrin-B3-Fc did not significantly reduce the survival of NSPC except at 1 µg/mL (P<0.05), but Fc fragments alone reduced NSPC survival at all doses in a dose-dependent fashion. In vivo, intrathecal infusion of ephrin-B3-Fc increased the proliferation of endogenous ependymal cells and the proportion of proliferating cells that expressed the glial fibrillary acidic protein astrocytic marker in the injured spinal cord compared with the infusion of PBS (P<0.05). However, in the injured spinal cord, the infusion of either ephrin-B3-Fc or Fc fragments alone caused a 20-fold reduction in the survival of transplanted NSPC (P<0.001). Thus, after SCI, ephrin-B3-Fc and Fc fragments are toxic to transplanted NSPC.


Assuntos
Efrina-B3/administração & dosagem , Células-Tronco Neurais/fisiologia , Traumatismos da Medula Espinal/terapia , Medula Espinal/efeitos dos fármacos , Animais , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Epêndima/patologia , Efrina-B3/farmacologia , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Infusão Espinal , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/transplante , Ratos , Ratos Transgênicos , Ratos Wistar , Receptor EphA4/metabolismo , Medula Espinal/metabolismo , Medula Espinal/patologia
19.
J Biomed Mater Res A ; 101(5): 1472-7, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23129254

RESUMO

No effective clinical treatment currently exists for traumatic spinal cord injury. Cell replacement therapy holds promise for attaining functional repair. Cells may be delivered directly or near the injury site; however, this strategy requires a delivery vehicle to maintain cell viability. We have identified an injectable, biocompatible, and biodegradable hydrogel scaffold composed of hyaluronan (HA) and methylcellulose (MC) that may be an effective scaffold for therapeutic cell delivery. The purpose of the present study was to determine the effects of polymer concentration on HAMC mechanical strength, gelation time, and cell viability. The yield stress of HAMC, a measure of mechanical stiffness, was tunable via manipulation of MC and HA content. Measurement of the elastic and storage moduli as functions of time revealed that HAMC gels in less than 5 min at physiological temperatures. Human umbilical tissue-derived cells encapsulated in HAMC were homogenously and stably distributed over 3 days in culture and extended processes into the scaffold. Cell viability was stable over this period in all but the most concentrated HAMC formulation. Because of its strength-tunability, rapid gelation, and ability to maintain cell viability, HAMC is a promising vehicle for cell delivery and is being tested in ongoing in vivo studies.


Assuntos
Ácido Hialurônico/química , Hidrogéis/química , Metilcelulose/química , Alicerces Teciduais/química , Sobrevivência Celular , Terapia Baseada em Transplante de Células e Tecidos , Células Cultivadas , Módulo de Elasticidade , Humanos , Reologia , Traumatismos da Medula Espinal/terapia
20.
J Clin Invest ; 122(11): 3824-34, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23114605

RESUMO

Spinal cord injury (SCI) is a devastating condition producing great personal and societal costs and for which there is no effective treatment. Stem cell transplantation is a promising therapeutic strategy, though much preclinical and clinical research work remains. Here, we briefly describe SCI epidemiology, pathophysiology, and experimental and clinical stem cell strategies. Research in stem cell biology and cell reprogramming is rapidly advancing, with the hope of moving stem cell therapy closer to helping people with SCI. We examine issues important for clinical translation and provide a commentary on recent developments, including termination of the first human embryonic stem cell transplantation trial in human SCI.


Assuntos
Pesquisa Biomédica , Desdiferenciação Celular , Traumatismos da Medula Espinal , Transplante de Células-Tronco , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos , Humanos , Traumatismos da Medula Espinal/epidemiologia , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...