Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
J Neurosci ; 41(41): 8508-8531, 2021 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-34417332

RESUMO

Axon regenerative failure in the mature CNS contributes to functional deficits following many traumatic injuries, ischemic injuries, and neurodegenerative diseases. The complement cascade of the innate immune system responds to pathogen threat through inflammatory cell activation, pathogen opsonization, and pathogen lysis, and complement is also involved in CNS development, neuroplasticity, injury, and disease. Here, we investigated the involvement of the classical complement cascade and microglia/monocytes in CNS repair using the mouse optic nerve injury (ONI) model, in which axons arising from retinal ganglion cells (RGCs) are disrupted. We report that central complement C3 protein and mRNA, classical complement C1q protein and mRNA, and microglia/monocyte phagocytic complement receptor CR3 all increase in response to ONI, especially within the optic nerve itself. Importantly, genetic deletion of C1q, C3, or CR3 attenuates RGC axon regeneration induced by several distinct methods, with minimal effects on RGC survival. Local injections of C1q function-blocking antibody revealed that complement acts primarily within the optic nerve, not retina, to support regeneration. Moreover, C1q opsonizes and CR3+ microglia/monocytes phagocytose growth-inhibitory myelin debris after ONI, a likely mechanism through which complement and myeloid cells support axon regeneration. Collectively, these results indicate that local optic nerve complement-myeloid phagocytic signaling is required for CNS axon regrowth, emphasizing the axonal compartment and highlighting a beneficial neuroimmune role for complement and microglia/monocytes in CNS repair.SIGNIFICANCE STATEMENT Despite the importance of achieving axon regeneration after CNS injury and the inevitability of inflammation after such injury, the contributions of complement and microglia to CNS axon regeneration are largely unknown. Whereas inflammation is commonly thought to exacerbate the effects of CNS injury, we find that complement proteins C1q and C3 and microglia/monocyte phagocytic complement receptor CR3 are each required for retinal ganglion cell axon regeneration through the injured mouse optic nerve. Also, whereas studies of optic nerve regeneration generally focus on the retina, we show that the regeneration-relevant role of complement and microglia/monocytes likely involves myelin phagocytosis within the optic nerve. Thus, our results point to the importance of the innate immune response for CNS repair.


Assuntos
Axônios/metabolismo , Complemento C1q/metabolismo , Complemento C3/metabolismo , Células Mieloides/metabolismo , Traumatismos do Nervo Óptico/metabolismo , Células Ganglionares da Retina/metabolismo , Animais , Axônios/imunologia , Complemento C1q/imunologia , Complemento C3/imunologia , Feminino , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/imunologia , Regeneração Nervosa/fisiologia , Traumatismos do Nervo Óptico/imunologia , Traumatismos do Nervo Óptico/patologia , Células Ganglionares da Retina/imunologia
3.
Restor Neurol Neurosci ; 37(6): 525-544, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31609715

RESUMO

The optic nerve conveys information about the outside world from the retina to multiple subcortical relay centers. Until recently, the optic nerve was widely believed to be incapable of re-growing if injured, with dire consequences for victims of traumatic, ischemic, or neurodegenerative diseases of this pathway. Over the past 10-20 years, research from our lab and others has made considerable progress in defining factors that normally suppress axon regeneration and the ability of retinal ganglion cells, the projection neurons of the retina, to survive after nerve injury. Here we describe research from our lab on the role of inflammation-derived growth factors, suppression of inter-cellular signals among diverse retinal cell types, and combinatorial therapies, along with related studies from other labs, that enable animals with optic nerve injury to regenerate damaged retinal axons back to the brain. These studies raise the possibility that vision might one day be restored to people with optic nerve damage.


Assuntos
Regeneração Nervosa/fisiologia , Traumatismos do Nervo Óptico/metabolismo , Nervo Óptico/fisiologia , Células Ganglionares da Retina/metabolismo , Animais , Axônios/metabolismo , Axônios/ultraestrutura , Humanos , Mediadores da Inflamação/metabolismo , Nervo Óptico/ultraestrutura , Traumatismos do Nervo Óptico/patologia , Células Ganglionares da Retina/ultraestrutura
4.
Neural Regen Res ; 13(6): 1046-1053, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29926832

RESUMO

In spite of advances in surgical care and rehabilitation, the consequences of spinal cord injury (SCI) are still challenging. Several experimental therapeutic strategies have been studied in the SCI field, and recent advances have led to the development of therapies that may act on the inhibitory microenvironment. Assorted lineages of stem cells are considered a good treatment for SCI. This study investigated the effect of systemic transplantation of mesenchymal stem cells (MSCs) in a compressive SCI model. Here we present results of the intraperitoneal route, which has not been used previously for MSC administration after compressive SCI. We used adult female C57BL/6 mice that underwent laminectomy at the T9 level, followed by spinal cord compression for 1 minute with a 30-g vascular clip. The animals were divided into five groups: sham (anesthesia and laminectomy but without compression injury induction), MSC i.p. (intraperitoneal injection of 8 × 105 MSCs in 500 µL of DMEM at 7 days after SCI), MSC i.v. (intravenous injection of 8 × 105 MSCs in 500 µL of DMEM at 7 days after SCI), DMEM i.p. (intraperitoneal injection of 500 µL of DMEM at 7 days after SCI), DMEM i.v. (intravenous injection of 500 µL of DMEM at 7 days after SCI). The effects of MSCs transplantation in white matter sparing were analyzed by luxol fast blue staining. The number of preserved fibers was counted in semithin sections stained with toluidine blue and the presence of trophic factors was analyzed by immunohistochemistry. In addition, we analyzed the locomotor performance with Basso Mouse Scale and Global Mobility Test. Our results showed white matter preservation and a larger number of preserved fibers in the MSC groups than in the DMEM groups. Furthermore, the MSC groups had higher levels of trophic factors (brain-derived neurotrophic factor, nerve growth factor, neurotrophin-3 and neurotrophin-4) in the spinal cord and improved locomotor performance. Our results indicate that injection of MSCs by either intraperitoneal or intravenous routes results in beneficial outcomes and can be elected as a choice for SCI treatment.

5.
Dev Biol ; 429(1): 105-117, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28716713

RESUMO

Myc proto-oncogenes regulate diverse cellular processes during development, but their roles during morphogenesis of specific tissues are not fully understood. We found that c-myc regulates cell proliferation in mouse lens development and previous genome-wide studies suggested functional roles for N-myc in developing lens. Here, we examined the role of N-myc in mouse lens development. Genetic inactivation of N-myc in the surface ectoderm or lens vesicle impaired eye and lens growth, while "late" inactivation in lens fibers had no effect. Unexpectedly, defective growth of N-myc-deficient lenses was not associated with alterations in lens progenitor cell proliferation or survival. Notably, N-myc-deficient lens exhibited a delay in degradation of DNA in terminally differentiating lens fiber cells. RNA-sequencing analysis of N-myc-deficient lenses identified a cohort of down-regulated genes associated with fiber cell differentiation that included DNaseIIß. Further, an integrated analysis of differentially expressed genes in N-myc-deficient lens using normal lens expression patterns of iSyTE, N-myc-binding motif analysis and molecular interaction data from the String database led to the derivation of an N-myc-based gene regulatory network in the lens. Finally, analysis of N-myc and c-myc double-deficient lens demonstrated that these Myc genes cooperate to drive lens growth prior to lens vesicle stage. Together, these findings provide evidence for exclusive and cooperative functions of Myc transcription factors in mouse lens development and identify novel mechanisms by which N-myc regulates cell differentiation during eye morphogenesis.


Assuntos
Diferenciação Celular , Cristalino/citologia , Cristalino/crescimento & desenvolvimento , Proteína Proto-Oncogênica N-Myc/metabolismo , Animais , Diferenciação Celular/genética , Núcleo Celular/metabolismo , Proliferação de Células/genética , Sobrevivência Celular/genética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Cristalino/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transcrição Gênica , Transcriptoma/genética
7.
Proc Natl Acad Sci U S A ; 114(2): E209-E218, 2017 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-28049831

RESUMO

Retinal ganglion cells (RGCs), the projection neurons of the eye, cannot regenerate their axons once the optic nerve has been injured and soon begin to die. Whereas RGC death and regenerative failure are widely viewed as being cell-autonomous or influenced by various types of glia, we report here that the dysregulation of mobile zinc (Zn2+) in retinal interneurons is a primary factor. Within an hour after the optic nerve is injured, Zn2+ increases several-fold in retinal amacrine cell processes and continues to rise over the first day, then transfers slowly to RGCs via vesicular release. Zn2+ accumulation in amacrine cell processes involves the Zn2+ transporter protein ZnT-3, and deletion of slc30a3, the gene encoding ZnT-3, promotes RGC survival and axon regeneration. Intravitreal injection of Zn2+ chelators enables many RGCs to survive for months after nerve injury and regenerate axons, and enhances the prosurvival and regenerative effects of deleting the gene for phosphatase and tensin homolog (pten). Importantly, the therapeutic window for Zn2+ chelation extends for several days after nerve injury. These results show that retinal Zn2+ dysregulation is a major factor limiting the survival and regenerative capacity of injured RGCs, and point to Zn2+ chelation as a strategy to promote long-term RGC protection and enhance axon regeneration.


Assuntos
Regeneração Nervosa , Traumatismos do Nervo Óptico/metabolismo , Nervo Óptico/fisiologia , Retina/fisiologia , Zinco/metabolismo , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Proteínas de Transporte de Cátions , Quelantes/farmacologia , Etilaminas/farmacologia , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Proteínas de Membrana Transportadoras , Camundongos Endogâmicos C57BL , Camundongos Knockout , Piridinas/farmacologia , Ácidos Sulfanílicos/farmacologia
8.
Histol Histopathol ; 32(3): 253-262, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27255346

RESUMO

A trauma to the mature central nervous system (CNS) often leads to persistent deficits, due to the inability of axons to regenerate after being injured. Increasing evidence suggests that pro-inflammatory and pro-apoptotic genes can present a major obstacle to promoting neuroprotection of retinal ganglion cells and consequently succeed in axonal regeneration. This study evaluated the effect of the absence of galectin-3 (Gal-3) on retinal ganglion cells (RGC) survival and axonal regeneration/degeneration after optic nerve crush injury. Two weeks after crush there was a 2.6 fold increase in the rate of cell survival in Gal-3-/- mice (1283±79.15) compared to WT animals (495.4±53.96). However, no regeneration was observed in the Gal-3-/- mice two weeks after lesion. Furthermore, axonal degeneration presented a particular pattern on those mice; Electron Microscopy (EM) analysis showed incomplete axon degeneration while the WT mice presented an advanced stage of degeneration. This suggests that the removal of the nerve fibers in the Gal 3-/- mice could be deficient and this would cause a delay in the process of Wallerian degeneration once there is a decrease in the number of macrophages/microglia in the nerve. This study demonstrates how the absence of Gal-3 can affect RGC survival and optic nerve regeneration/degeneration after lesion. Our results suggest that the absence of Gal-3 plays an important role in the survival of RGC and thus can be a potential target for therapeutic intervention in RGC neuroprotection.


Assuntos
Galectina 3/metabolismo , Regeneração Nervosa/fisiologia , Neuroproteção/fisiologia , Traumatismos do Nervo Óptico/patologia , Células Ganglionares da Retina/patologia , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Compressão Nervosa , Degeneração Neural/metabolismo , Traumatismos do Nervo Óptico/metabolismo , Células Ganglionares da Retina/metabolismo
9.
J Neurosci ; 36(35): 9148-60, 2016 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-27581456

RESUMO

UNLABELLED: Action potential initiation and propagation in myelinated axons require ion channel clustering at axon initial segments (AIS) and nodes of Ranvier. Disruption of these domains after injury impairs nervous system function. Traditionally, injured CNS axons are considered refractory to regeneration, but some recent approaches challenge this view by showing robust long-distance regeneration. However, whether these approaches allow remyelination and promote the reestablishment of AIS and nodes of Ranvier is unknown. Using mouse optic nerve crush as a model for CNS traumatic injury, we performed a detailed analysis of AIS and node disruption after nerve crush. We found significant disruption of AIS and loss of nodes within days of the crush, and complete loss of nodes 1 week after injury. Genetic deletion of the tumor suppressor phosphatase and tensin homolog (Pten) in retinal ganglion cells (RGCs), coupled with stimulation of RGCs by inflammation and cAMP, dramatically enhanced regeneration. With this treatment, we found significant reestablishment of RGC AIS, remyelination, and even reassembly of nodes in regions proximal, within, and distal to the crush site. Remyelination began near the retina, progressed distally, and was confirmed by electron microscopy. Although axons grew rapidly, remyelination and nodal ion channel clustering was much slower. Finally, genetic deletion of ankyrinG from RGCs to block AIS reassembly did not affect axon regeneration, indicating that preservation of neuronal polarity is not required for axon regeneration. Together, our results demonstrate, for the first time, that regenerating CNS axons can be remyelinated and reassemble new AIS and nodes of Ranvier. SIGNIFICANCE STATEMENT: We show, for the first time, that regenerated CNS axons have the capacity to both remyelinate and reassemble the axon initial segments and nodes of Ranvier necessary for rapid and efficient action potential propagation.


Assuntos
Axônios/fisiologia , Regeneração Nervosa/fisiologia , Doenças do Nervo Óptico/patologia , Doenças do Nervo Óptico/fisiopatologia , Animais , Anquirinas/genética , Anquirinas/metabolismo , Axônios/ultraestrutura , Moléculas de Adesão Celular Neuronais , Toxina da Cólera/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Regeneração Nervosa/genética , Proteínas do Tecido Nervoso/metabolismo , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Nós Neurofibrosos/metabolismo , Nós Neurofibrosos/patologia , Nós Neurofibrosos/ultraestrutura , Espectrina/metabolismo , Estatísticas não Paramétricas , Fatores de Tempo
10.
PLoS One ; 9(10): e108919, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25330147

RESUMO

The ganglioside 9-O-acetyl GD3 is overexpressed in peripheral nerves after lesioning, and its expression is correlated with axonal degeneration and regeneration in adult rodents. However, the biological roles of this ganglioside during the regenerative process are unclear. We used mice lacking GD3 synthase (Siat3a KO), an enzyme that converts GM3 to GD3, which can be further converted to 9-O-acetyl GD3. Morphological analyses of longitudinal and transverse sections of the sciatic nerve revealed significant differences in the transverse area and nerve thickness. The number of axons and the levels of myelin basic protein were significantly reduced in adult KO mice compared to wild-type (WT) mice. The G-ratio was increased in KO mice compared to WT mice based on quantification of thin transverse sections stained with toluidine blue. We found that neurite outgrowth was significantly reduced in the absence of GD3. However, addition of exogenous GD3 led to neurite growth after 3 days, similar to that in WT mice. To evaluate fiber regeneration after nerve lesioning, we compared the regenerated distance from the lesion site and found that this distance was one-fourth the length in KO mice compared to WT mice. KO mice in which GD3 was administered showed markedly improved regeneration compared to the control KO mice. In summary, we suggest that 9-O-acetyl GD3 plays biological roles in neuron-glia interactions, facilitating axonal growth and myelination induced by Schwann cells. Moreover, exogenous GD3 can be converted to 9-O-acetyl GD3 in mice lacking GD3 synthase, improving regeneration.


Assuntos
Técnicas de Inativação de Genes , Regeneração Nervosa , Neurônios/citologia , Nervo Isquiático/citologia , Nervo Isquiático/fisiologia , Sialiltransferases/deficiência , Sialiltransferases/genética , Animais , Axônios/efeitos dos fármacos , Contagem de Células , Feminino , Gangliosídeos/farmacologia , Integrina beta1/metabolismo , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Bainha de Mielina/metabolismo , Regeneração Nervosa/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Nervo Isquiático/efeitos dos fármacos
11.
PLoS One ; 9(10): e110090, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25333892

RESUMO

BACKGROUND: Despite the regenerative potential of the peripheral nervous system, severe nerve lesions lead to loss of target-organ innervation, making complete functional recovery a challenge. Few studies have given attention to combining different approaches in order to accelerate the regenerative process. OBJECTIVE: Test the effectiveness of combining Schwann-cells transplantation into a biodegradable conduit, with treadmill training as a therapeutic strategy to improve the outcome of repair after mouse nerve injury. METHODS: Sciatic nerve transection was performed in adult C57BL/6 mice; the proximal and distal stumps of the nerve were sutured into the conduit. Four groups were analyzed: acellular grafts (DMEM group), Schwann cell grafts (3×105/2 µL; SC group), treadmill training (TMT group), and treadmill training and Schwann cell grafts (TMT + SC group). Locomotor function was assessed weekly by Sciatic Function Index and Global Mobility Test. Animals were anesthetized after eight weeks and dissected for morphological analysis. RESULTS: Combined therapies improved nerve regeneration, and increased the number of myelinated fibers and myelin area compared to the DMEM group. Motor recovery was accelerated in the TMT + SC group, which showed significantly better values in sciatic function index and in global mobility test than in the other groups. The TMT + SC group showed increased levels of trophic-factor expression compared to DMEM, contributing to the better functional outcome observed in the former group. The number of neurons in L4 segments was significantly higher in the SC and TMT + SC groups when compared to DMEM group. Counts of dorsal root ganglion sensory neurons revealed that TMT group had a significant increased number of neurons compared to DMEM group, while the SC and TMT + SC groups had a slight but not significant increase in the total number of motor neurons. CONCLUSION: These data provide evidence that this combination of therapeutic strategies can significantly improve functional and morphological recovery after sciatic injury.


Assuntos
Transplante de Células , Regeneração Nervosa , Condicionamento Físico Animal , Células de Schwann/citologia , Nervo Isquiático/citologia , Nervo Isquiático/fisiologia , Animais , Axônios/fisiologia , Sobrevivência Celular , Modelos Animais de Doenças , Masculino , Camundongos , Neurônios Motores/fisiologia , Fatores de Crescimento Neural/metabolismo , Junção Neuromuscular , Traumatismos dos Nervos Periféricos/patologia , Traumatismos dos Nervos Periféricos/fisiopatologia , Traumatismos dos Nervos Periféricos/terapia , Poliésteres/metabolismo , Recuperação de Função Fisiológica , Nervo Isquiático/ultraestrutura
12.
Brain Nerve ; 66(3): 265-72, 2014 Mar.
Artigo em Japonês | MEDLINE | ID: mdl-24607951

RESUMO

The optic nerve has been widely studied in search for insights into mechanisms that suppress or promote axon regeneration after injury. Like other CNS neurons, adult retinal ganglion cells (RGCs) normally fail to regenerate their axons after optic nerve injury. Recent studies have identified molecular pathways able to allow partial regeneration of damaged RGCs axons in mature rodents; however, it is still unknown, whether regrowing optic axons can re-enter the brain in large numbers, innervate the correct target areas, and thus restore vision. We investigated these questions by using three manipulations that synergistically increase regeneration far above the level induced by any of the three used alone. Oncomodulin is a calcium-binding protein secreted by activated macrophages and neutrophils and stimulates RGCs to regenerate axons. Its ability to bind to RGCs and activate a downstream response is enhanced by elevating intracellular cAMP. Studies were carried out in mice with a conditional deletion of the gene encoding PTEN, a phosphatase and tensin homolog that suppresses signaling through the Akt/mTOR/S6K pathway. Our results showed that intraocular inflammation, deletion of the PTEN gene and elevation of intracellular cAMP exert synergistic effects that enable RGCs to regenerate the full length of axons, form synapses, and restore simple visual functions. These results demonstrate the feasibility of reconstructing central circuitry for vision after optic nerve damage in mature mammals.


Assuntos
Proteínas de Ligação ao Cálcio/farmacologia , Regeneração Nervosa/efeitos dos fármacos , Traumatismos do Nervo Óptico/tratamento farmacológico , Nervo Óptico/efeitos dos fármacos , Visão Ocular/efeitos dos fármacos , Envelhecimento , Animais , Regeneração Nervosa/genética , Traumatismos do Nervo Óptico/genética , Traumatismos do Nervo Óptico/metabolismo , Visão Ocular/fisiologia
13.
Int Rev Neurobiol ; 108: 59-77, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24083431

RESUMO

Although the peripheral nervous system has an inherent capacity for regeneration, injuries to nerves still result in considerable disabilities. The persistence of these disabilities along with the underlying problem of nerve reconstruction has motivated neuroscientists worldwide to seek additional therapeutic strategies. In recent years, cell-based therapy has emerged as a promising therapeutic tool. Schwann cells (SCs) are the main supportive cells for peripheral nerve regeneration; however, there are several technical limitations regarding its application for cell-based therapy. In this context, bone marrow mesenchymal stem cells (BM-MSCs) have been used as alternatives to SCs for treating peripheral neuropathies, showing great promise. Several studies have been trying to shed light on the mechanisms behind the nerve regeneration-promotion potential of BM-MSCs. Although not completely clarified, understanding how BM-MSCs exert tissue repair effects will facilitate their development as therapeutic agents before they become a clinically viable tool for encouraging peripheral nerve regeneration.


Assuntos
Transplante de Medula Óssea/tendências , Transplante de Células-Tronco Mesenquimais/tendências , Doenças do Sistema Nervoso Periférico/cirurgia , Animais , Transplante de Medula Óssea/métodos , Humanos , Transplante de Células-Tronco Mesenquimais/métodos , Regeneração Nervosa/fisiologia , Doenças do Sistema Nervoso Periférico/fisiopatologia
14.
Int Rev Neurobiol ; 106: 153-72, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23211463

RESUMO

The optic nerve has been widely studied for insights into mechanisms that suppress or promote axon regeneration after central nervous system injury. Following optic nerve damage in adult mammals, retinal ganglion cells (RGCs) normally fail to regenerate their axons, resulting in blindness in patients who suffer from neurodegenerative diseases such as glaucoma or who have sustained traumatic injury to the optic nerve. Over the past several decades, many groups have investigated the basis of regenerative failure in the hope of developing strategies to stimulate the regrowth of axons and restore visual function. New findings show that a combination of therapies that act synergistically to activate RGCs' intrinsic growth state enables these cells to regenerate their axons the full length of the optic nerve, across the optic chiasm, and into the brain, where they establish synapses in appropriate target zones and restore limited visual responses. These treatments involve the induction of a limited inflammatory response in the eye to increase levels of oncomodulin and other growth factors; elevation of intracellular cAMP; and deletion of the pten gene in RGCs. Although these methods cannot be applied in the clinic, they point to strategies that might be.


Assuntos
Regeneração Nervosa/fisiologia , Traumatismos do Nervo Óptico/terapia , Recuperação de Função Fisiológica/fisiologia , Visão Ocular/fisiologia , Animais , Terapia Genética/métodos , Humanos , Camundongos , Fatores de Crescimento Neural/farmacologia , Fatores de Crescimento Neural/uso terapêutico , Regeneração Nervosa/efeitos dos fármacos , Traumatismos do Nervo Óptico/genética , Traumatismos do Nervo Óptico/fisiopatologia , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/fisiologia , Visão Ocular/efeitos dos fármacos
15.
Proc Natl Acad Sci U S A ; 109(23): 9149-54, 2012 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-22615390

RESUMO

The mature optic nerve cannot regenerate when injured, leaving victims of traumatic nerve damage or diseases such as glaucoma with irreversible visual losses. Recent studies have identified ways to stimulate retinal ganglion cells to regenerate axons part-way through the optic nerve, but it remains unknown whether mature axons can reenter the brain, navigate to appropriate target areas, or restore vision. We show here that with adequate stimulation, retinal ganglion cells are able to regenerate axons the full length of the visual pathway and on into the lateral geniculate nucleus, superior colliculus, and other visual centers. Regeneration partially restores the optomotor response, depth perception, and circadian photoentrainment, demonstrating the feasibility of reconstructing central circuitry for vision after optic nerve damage in mature mammals.


Assuntos
Axônios/fisiologia , Traumatismos do Nervo Óptico/tratamento farmacológico , Nervo Óptico/fisiologia , Regeneração/fisiologia , Células Ganglionares da Retina/fisiologia , Zimosan/farmacologia , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Ritmo Circadiano/fisiologia , AMP Cíclico/metabolismo , Dependovirus , Deleção de Genes , Vetores Genéticos/genética , Corpos Geniculados/fisiologia , Integrases/metabolismo , Camundongos , Nervo Óptico/citologia , PTEN Fosfo-Hidrolase/genética , Células Ganglionares da Retina/citologia , Colículos Superiores/fisiologia , Zimosan/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...