Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35101972

RESUMO

Neuroinflammation is well known to be associated with neurodegenerative diseases. Apoptosis signal-regulating kinase 1 (ASK1) is a mitogen-activated protein kinase kinase kinase that has been implicated in neuroinflammation, but its precise cellular and molecular mechanisms remain unknown. In this study, we generated conditional knockout (CKO) mice that lack ASK1 in T cells, dendritic cells, microglia/macrophages, microglia, or astrocytes, to assess the roles of ASK1 during experimental autoimmune encephalomyelitis (EAE). We found that neuroinflammation was reduced in both the early and later stages of EAE in microglia/macrophage-specific ASK1 knockout mice, whereas only the later-stage neuroinflammation was ameliorated in astrocyte-specific ASK1 knockout mice. ASK1 deficiency in T cells and dendritic cells had no significant effects on EAE severity. Further, we found that ASK1 in microglia/macrophages induces a proinflammatory environment, which subsequently activates astrocytes to exacerbate neuroinflammation. Microglia-specific ASK1 deletion was achieved using a CX3CR1CreER system, and we found that ASK1 signaling in microglia played a major role in generating and maintaining disease. Activated astrocytes produce key inflammatory mediators, including CCL2, that further activated and recruited microglia/macrophages, in an astrocytic ASK1-dependent manner. Astrocyte-specific analysis revealed CCL2 expression was higher in the later stage compared with the early stage, suggesting a greater proinflammatory role of astrocytes in the later stage. Our findings demonstrate cell-type-specific roles of ASK1 and suggest phase-specific ASK1-dependent glial cell interactions in EAE pathophysiology. We propose glial ASK1 as a promising therapeutic target for reducing neuroinflammation.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , MAP Quinase Quinase Quinase 5/imunologia , Microglia/imunologia , Esclerose Múltipla/imunologia , Transdução de Sinais/imunologia , Animais , Células Dendríticas/imunologia , Encefalomielite Autoimune Experimental/genética , Inflamação/genética , Inflamação/imunologia , MAP Quinase Quinase Quinase 5/genética , Macrófagos/imunologia , Camundongos , Camundongos Knockout , Esclerose Múltipla/genética , Transdução de Sinais/genética , Linfócitos T/imunologia
2.
J Pharmacol Sci ; 154(4): 326-333, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38485351

RESUMO

PURPOSE: To determine whether combination of topical ripasudil and brimonidine has more effective neuroprotection on retinal ganglion cells (RGCs) following injury to axons composing the optic nerve. METHODS: Topical ripasudil, brimonidine, or mixture of both drugs were administered to adult mice after optic nerve injury (ONI). The influence of drug conditions on RGC health were evaluated by the quantifications of surviving RGCs, phosphorylated p38 mitogen-activated protein kinase (phospho-p38), and expressions of trophic factors and proinflammatory mediators in the retina. RESULTS: Topical ripasudil and brimonidine suppressed ONI-induced RGC death respectively, and mixture of both drugs further stimulated RGC survival. Topical ripasudil and brimonidine suppressed ONI-induced phospho-p38 in the whole retina. In addition, topical ripasudil suppressed expression levels of TNFα, IL-1ß and monocyte chemotactic protein-1 (MCP-1), whereas topical brimonidine increased the expression level of basic fibroblast growth factor (bFGF). CONCLUSIONS: Combination of topical ripasudil and brimonidine may enhance RGC protection by modulating multiple signaling pathways in the retina.


Assuntos
Isoquinolinas , Traumatismos do Nervo Óptico , Sulfonamidas , Camundongos , Animais , Tartarato de Brimonidina , Traumatismos do Nervo Óptico/tratamento farmacológico , Traumatismos do Nervo Óptico/metabolismo , Neuroproteção , Combinação de Medicamentos
3.
Mol Ther ; 31(3): 810-824, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36463402

RESUMO

Activation of neurotrophic factor signaling is a promising therapy for neurodegeneration. However, the transient nature of ligand-dependent activation limits its effectiveness. In this study, we solved this problem by inventing a system that forces membrane localization of the intracellular domain of tropomyosin receptor kinase B (iTrkB), which results in constitutive activation without ligands. Our system overcomes the small size limitation of the genome packaging in adeno-associated virus (AAV) and allows high expression of the transgene. Using AAV-mediated gene therapy in the eyes, we demonstrate that iTrkB expression enhances neuroprotection in mouse models of glaucoma and stimulates robust axon regeneration after optic nerve injury. In addition, iTrkB expression in the retina was also effective in an optic tract transection model, in which the injury site is near the superior colliculus. Regenerating axons successfully formed pathways to their brain targets, resulting in partial recovery of visual behavior. Our system may also be applicable to other trophic factor signaling pathways and lead to a significant advance in the field of gene therapy for neurotrauma and neurodegenerative disorders, including glaucoma.


Assuntos
Glaucoma , Células Ganglionares da Retina , Camundongos , Animais , Células Ganglionares da Retina/metabolismo , Axônios/fisiologia , Regeneração Nervosa/genética , Retina , Glaucoma/genética , Glaucoma/terapia , Glaucoma/metabolismo , Modelos Animais de Doenças
4.
Neuropathology ; 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38775061

RESUMO

Multiple sclerosis (MS), the leading cause of disability in young adults, is an inflammatory disease of the central nervous system characterized by localized areas of demyelination. Apoptosis signal-regulating kinase 1 (ASK1) is a mitogen-activated protein kinase kinase kinase that has been shown to be implicated in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. Interestingly, ASK1 signaling regulates glial cell interactions and drives neuroinflammation in EAE mice. To further investigate its clinical significance, in the present study, we examined the activation of ASK1 in the post-mortem brain of MS patients. ASK1 activation was found in active lesions of the corpus callosum in both microglia/macrophages and astrocytes. Moreover, ASK1 activation in astrocytes was higher than that in microglia/macrophages, which was in line with our findings in EAE mice. Our results suggest an important role of ASK1 in glial cells, indicating that ASK1 might be a good therapeutic target for MS.

5.
Genes Cells ; 27(8): 526-536, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35703119

RESUMO

Glaucoma is a neurodegenerative disorder caused by the death of retinal ganglion cells (RGCs). Elevated intraocular pressure (IOP) is a cause of glaucoma. However, glaucoma often develops with normal IOP and is known as normal-tension glaucoma (NTG). Glutamate neurotoxicity is considered as one of the significant causes of NTG, resulting in excessive stimulation of retinal neurons via the N-methyl-D-aspartate (NMDA) receptors. The present study examined the phosphorylation of collapsin response mediator protein-2 (CRMP2), a protein that is abundantly expressed in neurons and involved in their development. In two mouse models, NMDA-injection and glutamate/aspartate transporter (GLAST) mutant, CRMP2 phosphorylation at the cyclin-dependent kinase-5 (Cdk5) site was elevated in RGCs. We confirmed that the decrease in the number of RGCs and thickness of the inner retinal layer (IRL) could be suppressed after NMDA administration in CRMP2KI/KI mice with genetically inhibited CRMP2 phosphorylation. Next, we investigated GLAST heterozygotes (GLAST+/-) with CRMP2KI/KI (GLAST+/-;CRMP2KI/KI) and GLAST knockout (GLAST-/-) mice with CRMP2KI/KI (GLAST-/-;CRMP2KI/KI) mice and compared them with GLAST+/- and GLAST-/- mice. pCRMP2 (S522) inhibition significantly reduced RGC loss and IRL thinning. These results suggest that the inhibition of CRMP2 phosphorylation could be a novel strategy for treating NTG.


Assuntos
Glaucoma , Células Ganglionares da Retina , Animais , Modelos Animais de Doenças , Glaucoma/genética , Glaucoma/metabolismo , Ácido Glutâmico/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , N-Metilaspartato , Proteínas do Tecido Nervoso , Fosforilação , Células Ganglionares da Retina/metabolismo
6.
Exp Eye Res ; 226: 109314, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36400285

RESUMO

Glaucoma is one of the most common causes of blindness worldwide. It is thought to be a multifactorial disease with underlying mechanisms that include mitochondrial dysfunction and oxidative stress. Here, we used NF-E2 related factor 2 (Nrf2) knockout (KO) mice, which are vulnerable to oxidative stress, to examine a neuroprotective effect against oxidative stress due to rotenone, a mitochondrial complex I inhibitor. Wild-type (WT) and Nrf2 KO mice received an oral solution of rotenone for 30 days. We then extracted the retinas and performed immunohistochemistry and quantitative RT-PCR. We also prepared a primary Müller cell culture of samples from each mouse, added 30 µM rotenone, and then measured cell viability, cytotoxicity and CellRox absorbance. We also examined gene expression. We found a significant increase in the number of 8-OHdG-positive retinal ganglion cells (RGCs) after rotenone administration in both the WT and Nrf2 KO mice. There was no difference in the number of RNA-binding protein with multiple splicing (RBPMS)-positive RGCs in the WT and Nrf2 KO mice, but Nrf2 KO mice that were given rotenone had significantly less retinal gene expression of RBPMS than Nrf2 KO mice given a control. Moreover, there was significantly higher mRNA gene expression of vimentin and glial fibrillary acidic protein (GFAP) in Nrf2 KO mice that received rotenone than WT mice that received rotenone. A statistical analysis of the in vitro experiment showed that cell viability was lower, cytotoxicity was higher, and oxidative stress was higher in the Müller cells of the Nrf2 KO mice than the WT mice. Finally, brain-derived neurotrophic factor (BDNF) and basic fibroblast growth factor (bFGF) were significantly higher in the Müller cells of the Nrf2 KO mice than the WT mice. These findings suggest that in Nrf2 KO mice under oxidative stress caused by rotenone, temporary neurotrophic factors are secreted from the Müller cells, conferring neuroprotection in these cells.


Assuntos
Fator 2 Relacionado a NF-E2 , Rotenona , Camundongos , Animais , Camundongos Knockout , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Rotenona/toxicidade , Rotenona/metabolismo , Fatores de Crescimento Neural/metabolismo , Estresse Oxidativo , Neuroglia/metabolismo , Camundongos Endogâmicos C57BL
7.
J Biol Chem ; 295(19): 6710-6720, 2020 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-32241915

RESUMO

The DOCK-D (dedicator of cytokinesis D) family proteins are atypical guanine nucleotide exchange factors that regulate Rho GTPase activity. The family consists of Zizimin1 (DOCK9), Zizimin2 (DOCK11), and Zizimin3 (DOCK10). Functions of the DOCK-D family proteins are presently not well-explored, and the role of the DOCK-D family in neuroinflammation is unknown. In this study, we generated three mouse lines in which DOCK9 (DOCK9-/-), DOCK10 (DOCK10-/-), or DOCK11 (DOCK11-/-) had been deleted and examined the phenotypic effects of these gene deletions in MOG35-55 peptide-induced experimental autoimmune encephalomyelitis, an animal model of the neuroinflammatory disorder multiple sclerosis. We found that all the gene knockout lines were healthy and viable. The only phenotype observed under normal conditions was a slightly smaller proportion of B cells in splenocytes in DOCK10-/- mice than in the other mouse lines. We also found that the migration ability of macrophages is impaired in DOCK10-/- and DOCK11-/- mice and that the severity of experimental autoimmune encephalomyelitis was ameliorated only in DOCK10-/- mice. No apparent phenotype was observed for DOCK9-/- mice. Further investigations indicated that lipopolysaccharide stimulation up-regulates DOCK10 expression in microglia and that microglial migration is decreased in DOCK10-/- mice. Up-regulation of C-C motif chemokine ligand 2 (CCL2) expression induced by activation of Toll-like receptor 4 or 9 signaling was reduced in DOCK10-/- astrocytes compared with WT astrocytes. Taken together, our findings suggest that DOCK10 plays a role in innate immunity and neuroinflammation and might represent a potential therapeutic target for managing multiple sclerosis.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Esclerose Múltipla/metabolismo , Animais , Astrócitos/patologia , Modelos Animais de Doenças , Camundongos , Microglia/patologia , Esclerose Múltipla/patologia
8.
J Biol Chem ; 294(36): 13421-13433, 2019 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-31337702

RESUMO

Dedicator of cytokinesis 8 (DOCK8) is a guanine nucleotide exchange factor whose loss of function results in immunodeficiency, but its role in the central nervous system (CNS) has been unclear. Microglia are the resident immune cells of the CNS and are implicated in the pathogenesis of various neurodegenerative diseases, including multiple sclerosis (MS) and glaucoma, which affects the visual system. However, the exact roles of microglia in these diseases remain unknown. Herein, we report that DOCK8 is expressed in microglia but not in neurons or astrocytes and that its expression is increased during neuroinflammation. To define the role of DOCK8 in microglial activity, we focused on the retina, a tissue devoid of infiltrating T cells. The retina is divided into distinct layers, and in a disease model of MS/optic neuritis, DOCK8-deficient mice exhibited a clear reduction in microglial migration through these layers. Moreover, neuroinflammation severity, indicated by clinical scores, visual function, and retinal ganglion cell (RGC) death, was reduced in the DOCK8-deficient mice. Furthermore, using a glaucoma disease model, we observed impaired microglial phagocytosis of RGCs in DOCK8-deficient mice. Our data demonstrate that DOCK8 is expressed in microglia and regulates microglial activity in disease states. These findings contribute to a better understanding of the molecular pathways involved in microglial activation and implicate a role of DOCK8 in several neurological diseases.


Assuntos
Modelos Animais de Doenças , Fatores de Troca do Nucleotídeo Guanina/biossíntese , Microglia/metabolismo , Doenças Neurodegenerativas/metabolismo , Animais , Células Cultivadas , Feminino , Fatores de Troca do Nucleotídeo Guanina/deficiência , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
9.
Biochem Biophys Res Commun ; 529(4): 943-949, 2020 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-32819603

RESUMO

Glaucoma is one of the leading causes of blindness characterized by progressive loss of retinal ganglion cells (RGCs) and their axons. We reported that glutamate/aspartate transporter (GLAST) knockout mice showed progressive RGC loss and optic nerve degeneration that are similar to glaucoma. To explore the possibility that rare variants in the EAAT1 gene (the human homolog of GLAST) cause susceptibility to glaucoma, we performed targeted sequencing of EAAT1 in 440 patients with glaucoma and 450 control subjects. We identified 8 rare variants in 20 out of 440 patients, including 4 synonymous and 4 missense variants located at protein coding regions. One of these rare variants (rs117295512) showed significant association with the risk of glaucoma (OR = 10.44, P = 0.005). Furthermore, the allele frequency for loss-of-function EAAT1 variants, pAla169Gly and pAla329Thr, was 5.5 folds higher in the glaucoma (1.1%) compared with the control cohort (0.2%). These findings suggest that these rare variants may contribute to the pathogenesis of glaucoma and that loss-of-function variants in EAAT1 are present in a small number of patients with glaucoma.


Assuntos
Transportador 1 de Aminoácido Excitatório/genética , Glaucoma de Ângulo Aberto/genética , Glaucoma de Baixa Tensão/genética , Mutação de Sentido Incorreto , Mutação Silenciosa , Alelos , Sequência de Aminoácidos , Animais , Estudos de Casos e Controles , Linhagem Celular , Transportador 1 de Aminoácido Excitatório/deficiência , Expressão Gênica , Frequência do Gene , Glaucoma de Ângulo Aberto/metabolismo , Glaucoma de Ângulo Aberto/patologia , Humanos , Pressão Intraocular , Glaucoma de Baixa Tensão/metabolismo , Glaucoma de Baixa Tensão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nervo Óptico/metabolismo , Nervo Óptico/patologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Fatores de Risco , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
10.
Am J Pathol ; 187(12): 2876-2885, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28919108

RESUMO

The major role of the renin-angiotensin system (RAS), including that of angiotensin II (Ang II), the principal effector molecule, in the cardiovascular system is well known. Increasing evidence suggests that the RAS also plays a role in the development of autoimmune diseases. Optic neuritis (ie, inflammation of the optic nerve, with retinal ganglion cell loss) is strongly associated with multiple sclerosis. We investigated the effects of candesartan, an Ang II receptor antagonist, on optic neuritis in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. The Ang II concentration was increased in the early phase of EAE. Oral administration of candesartan markedly attenuated demyelination of the optic nerve and spinal cord and reduced retinal ganglion cell loss and visual impairment in mice with EAE. In vitro analyses revealed that Ang II up-regulated the expression of Toll-like receptor (TLR)-4 in astrocytes via the NF-κB pathway. In addition, Ang II treatment enhanced lipopolysaccharide-induced production of monocyte chemoattractant protein 1 in astrocytes, and pretreatment with candesartan or SN50, an NF-κB inhibitor, suppressed the effects of Ang II. The novel pathway of RAS-NF-κB-TLR4 in glial cells identified in the present study may be a valid therapeutic target for neurodegeneration in neuroinflammatory diseases.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Degeneração Neural/patologia , Neurite Óptica/patologia , Sistema Renina-Angiotensina/efeitos dos fármacos , Animais , Benzimidazóis/farmacologia , Compostos de Bifenilo , Encefalomielite Autoimune Experimental/patologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Sistema Renina-Angiotensina/fisiologia , Tetrazóis/farmacologia
11.
Am J Pathol ; 185(12): 3238-47, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26476348

RESUMO

Brain-derived neurotrophic factor (BDNF) regulates neural cell survival mainly by activating TrkB receptors. Several lines of evidence support a key role for BDNF-TrkB signaling in survival of adult retinal ganglion cells in animal models of optic nerve injury (ONI), but the neuroprotective effect of exogenous BDNF is transient. Glial cells have recently attracted considerable attention as mediators of neural cell survival, and TrkB expression in retinal glia suggests its role in neuroprotection. To elucidate this point directly, we examined the effect of ONI on TrkB(flox/flox):glial fibrillary acidic protein (GFAP)-Cre+ (TrkB(GFAP)) knockout (KO) mice, in which TrkB is deleted in retinal glial cells. ONI markedly increased mRNA expression levels of basic fibroblast growth factor (bFGF) in wild-type (WT) mice but not in TrkB(GFAP) KO mice. Immunohistochemical analysis at 7 days after ONI (d7) revealed bFGF up-regulation mainly occurred in Müller glia. ONI-induced retinal ganglion cell loss in WT mice was consistently mild compared with TrkB(GFAP) KO mice at d7. On the other hand, ONI severely decreased TrkB expression in both WT and TrkB(GFAP) KO mice after d7, and the severity of retinal degeneration was comparable with TrkB(GFAP) KO mice at d14. Our data provide direct evidence that glial TrkB signaling plays an important role in the early stage of neural protection after traumatic injury.


Assuntos
Neuroglia/metabolismo , Neuroproteção/fisiologia , Traumatismos do Nervo Óptico/metabolismo , Receptor trkB/fisiologia , Animais , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fatores de Crescimento Neural/biossíntese , Traumatismos do Nervo Óptico/complicações , Traumatismos do Nervo Óptico/patologia , Degeneração Retiniana/etiologia , Degeneração Retiniana/metabolismo , Células Ganglionares da Retina/metabolismo , Transdução de Sinais/fisiologia
12.
Am J Pathol ; 185(3): 756-64, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25542970

RESUMO

Valproic acid (VPA) is widely prescribed for treatment of epilepsy, mood disorders, migraines, and neuropathic pain. It exerts its therapeutic benefits through multiple mechanisms, including enhancement of GABAergic activity, activation of prosurvival protein kinases, and inhibition of histone deacetylase. Increasing evidence suggests that VPA possesses neuroprotective properties. We examined neuroprotective effects of VPA in an N-methyl-d-aspartate (NMDA) excitotoxicity model, which mimics some of the pathological features of glaucoma. In vivo retinal imaging using optical coherence tomography revealed that NMDA-induced retinal degeneration was suppressed in the VPA-treated retina, and histological analyses confirmed that VPA reduced retinal ganglion cell death. In vivo electrophysiological analyses demonstrated that visual impairment was prevented in the VPA-treated retina, clearly establishing both histological and functional effects of VPA. Brain-derived neurotrophic factor (BDNF) expression was up-regulated in Müller glial cells, and neuroprotective effects of VPA on retinal ganglion cells were significantly reduced in a conditional knockout mouse strain with deletion of tropomyosin receptor kinase B (TrkB), a receptor for BDNF from retinal ganglion cells. The results show that VPA stimulates BDNF up-regulation in Müller glial cells and provides direct evidence that neuronal TrkB is important in VPA-mediated neuroprotection. Also, VPA suppresses oxidative stress induced by NMDA in the retina. Our findings raise intriguing possibilities that the widely prescribed drug VPA may be useful for treatment of glaucoma.


Assuntos
Morte Celular/efeitos dos fármacos , N-Metilaspartato/farmacologia , Fármacos Neuroprotetores/farmacologia , Receptor trkB/metabolismo , Células Ganglionares da Retina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Ácido Valproico/farmacologia , Animais , Camundongos , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Regulação para Cima/efeitos dos fármacos
13.
Int J Mol Sci ; 17(9)2016 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-27657046

RESUMO

Neurotrophic factors play key roles in the development and survival of neurons. The potent neuroprotective effects of neurotrophic factors, including brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF), glial cell-line derived neurotrophic factor (GDNF) and nerve growth factor (NGF), suggest that they are good therapeutic candidates for neurodegenerative diseases. Glaucoma is a neurodegenerative disease of the eye that causes irreversible blindness. It is characterized by damage to the optic nerve, usually due to high intraocular pressure (IOP), and progressive degeneration of retinal neurons called retinal ganglion cells (RGCs). Current therapy for glaucoma focuses on reduction of IOP, but neuroprotection may also be beneficial. BDNF is a powerful neuroprotective agent especially for RGCs. Exogenous application of BDNF to the retina and increased BDNF expression in retinal neurons using viral vector systems are both effective in protecting RGCs from damage. Furthermore, induction of BDNF expression by agents such as valproic acid has also been beneficial in promoting RGC survival. In this review, we discuss the therapeutic potential of neurotrophic factors in retinal diseases and focus on the differential roles of glial and neuronal TrkB in neuroprotection. We also discuss the role of neurotrophic factors in neuroregeneration.

14.
J Neurosci ; 32(1): 264-74, 2012 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-22219288

RESUMO

Dock3, a new member of the guanine nucleotide exchange factors, causes cellular morphological changes by activating the small GTPase Rac1. Overexpression of Dock3 in neural cells promotes axonal outgrowth downstream of brain-derived neurotrophic factor (BDNF) signaling. We previously showed that Dock3 forms a complex with Fyn and WASP (Wiskott-Aldrich syndrome protein) family verprolin-homologous (WAVE) proteins at the plasma membrane, and subsequent Rac1 activation promotes actin polymerization. Here we show that Dock3 binds to and inactivates glycogen synthase kinase-3ß (GSK-3ß) at the plasma membrane, thereby increasing the nonphosphorylated active form of collapsin response mediator protein-2 (CRMP-2), which promotes axon branching and microtubule assembly. Exogenously applied BDNF induced the phosphorylation of GSK-3ß and dephosphorylation of CRMP-2 in hippocampal neurons. Moreover, increased phosphorylation of GSK-3ß was detected in the regenerating axons of transgenic mice overexpressing Dock3 after optic nerve injury. These results suggest that Dock3 plays important roles downstream of BDNF signaling in the CNS, where it regulates cell polarity and promotes axonal outgrowth by stimulating dual pathways: actin polymerization and microtubule assembly.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/fisiologia , Proteínas de Transporte/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Cones de Crescimento/metabolismo , Hipocampo/metabolismo , Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Células COS , Chlorocebus aethiops , Glicogênio Sintase Quinase 3 beta , Cones de Crescimento/efeitos dos fármacos , Cones de Crescimento/ultraestrutura , Fatores de Troca do Nucleotídeo Guanina , Células HEK293 , Hipocampo/citologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microtúbulos/efeitos dos fármacos , Microtúbulos/ultraestrutura
15.
Genes Cells ; 17(8): 688-97, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22734669

RESUMO

Dock3, a new member of the guanine nucleotide exchange factor family, causes cellular morphological changes by activating the small GTPase Rac1. Overexpression of Dock3 in neural cells promotes neurite outgrowth through the formation of a protein complex with Fyn and WAVE downstream of brain-derived neurotrophic factor (BDNF) signaling. Here, we report a novel Dock3-mediated BDNF pathway for neurite outgrowth. We show that Dock3 forms a complex with Elmo and activated RhoG downstream of BDNF-TrkB signaling and induces neurite outgrowth via Rac1 activation in PC12 cells. We also show the importance of Dock3 phosphorylation in Rac1 activation and show two key events that are necessary for efficient Dock3 phosphorylation: membrane recruitment of Dock3 and interaction of Dock3 with Elmo. These results suggest that Dock3 plays important roles downstream of BDNF signaling in the central nervous system where it stimulates actin polymerization by multiple pathways.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuritos/metabolismo , Receptor trkB/metabolismo , Transdução de Sinais , Fatores de Complexo Ternário/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Células COS , Proteínas de Transporte/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Chlorocebus aethiops , Ativação Enzimática , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo , Fatores de Troca do Nucleotídeo Guanina , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Células PC12 , Fosforilação , Mapeamento de Interação de Proteínas , Transporte Proteico , Ratos , Receptor trkB/genética , Fatores de Complexo Ternário/genética , Transfecção , Proteínas rac de Ligação ao GTP/genética , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP , Proteínas rho de Ligação ao GTP
16.
Proc Natl Acad Sci U S A ; 107(16): 7586-91, 2010 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-20368433

RESUMO

Atypical Rho-guanine nucleotide exchange factors (Rho-GEFs) that contain Dock homology regions (DHR-1 and DHR-2) are expressed in a variety of tissues; however, their functions and mechanisms of action remain unclear. We identify key conserved amino acids in the DHR-2 domain that are critical for the catalytic activity of Dock-GEFs (Dock1-4). We further demonstrate that Dock-GEFs directly associate with WASP family verprolin-homologous (WAVE) proteins through the DHR-1 domain. Brain-derived neurotrophic factor (BDNF)-TrkB signaling recruits the Dock3/WAVE1 complex to the plasma membrane, whereupon Dock3 activates Rac and dissociates from the WAVE complex in a phosphorylation-dependent manner. BDNF induces axonal sprouting through Dock-dependent Rac activation, and adult transgenic mice overexpressing Dock3 exhibit enhanced optic nerve regeneration after injury without affecting WAVE expression levels. Our results highlight a unique mechanism through which Dock-GEFs achieve spatial and temporal restriction of WAVE signaling, and identify Dock-GEF activity as a potential therapeutic target for axonal regeneration.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Família de Proteínas da Síndrome de Wiskott-Aldrich/metabolismo , Sequência de Aminoácidos , Animais , Axônios/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células COS , Chlorocebus aethiops , Hipocampo/metabolismo , Humanos , Camundongos , Dados de Sequência Molecular , Regeneração Nervosa , Neurônios/metabolismo , Nervo Óptico/metabolismo , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho , Homologia de Sequência de Aminoácidos , Transdução de Sinais
17.
Neurosci Lett ; 799: 137124, 2023 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-36780941

RESUMO

Visual disturbance after optic nerve injury is a serious problem. Attempts have been made to enhance the intrinsic ability of retinal ganglion cells (RGCs) to regenerate their axons, and the importance of PI3K/Akt and RAF/MEK/ERK signal activation has been suggested. Since these signals are shared with oncogenic signaling cascades, in this study, we focused on a constitutively active form of K-Ras, K-RasV12, to determine if overexpression of this molecule could stimulate axon regeneration. We confirmed that K-RasV12 phosphorylated Akt and ERK in vitro. Intravitreal delivery of AAV2-K-RasV12 increased the number of surviving RGCs and promoted 1.0 mm of axon regeneration one week after optic nerve injury without inducing abnormal proliferative effects in the RGCs. In addition, AAV2-K-RasV12 induced robust RGC axon regeneration, reaching as far as approximately 2.5 mm from the injury site, in eight weeks. Our findings suggest that AAV2-K-RasV12 could provide a good model for speedy and efficient analysis of the mechanism underlying axon regeneration in vivo.


Assuntos
Traumatismos do Nervo Óptico , Humanos , Axônios/fisiologia , Proteínas Proto-Oncogênicas c-akt , Fosfatidilinositol 3-Quinases , Regeneração Nervosa/fisiologia
18.
Cell Death Discov ; 9(1): 166, 2023 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-37188749

RESUMO

Dedicator of cytokinesis 3 (DOCK3) is an atypical member of the guanine nucleotide exchange factors (GEFs) and plays important roles in neurite outgrowth. DOCK3 forms a complex with Engulfment and cell motility protein 1 (Elmo1) and effectively activates Rac1 and actin dynamics. In this study, we screened 462,169 low-molecular-weight compounds and identified the hit compounds that stimulate the interaction between DOCK3 and Elmo1, and neurite outgrowth in vitro. Some of the derivatives from the hit compound stimulated neuroprotection and axon regeneration in a mouse model of optic nerve injury. Our findings suggest that the low-molecular-weight DOCK3 activators could be a potential therapeutic candidate for treating axonal injury and neurodegenerative diseases including glaucoma.

19.
Biochem Biophys Rep ; 29: 101197, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35028438

RESUMO

Lighting conditions may affect the development of retinal degenerative diseases such as macular degeneration. In this study, to determine whether the lighting environment affects the progression of degeneration of retinal ganglion cells (RGCs), we examined glutamate/aspartate transporter (GLAST) heterozygous (GLAST+/-) mice, a mouse model of normal tension glaucoma. GLAST+/- mice were reared under a 12-h light-dark cycle (Light/Dark) or complete darkness (Dark/Dark) condition after birth. The total RGC number in the Dark/Dark group was significantly decreased compared with the Light/Dark group at 3 weeks old, while the number of osteopontin-positive αRGCs were similar in both groups. At 6 and 12 weeks old, the total RGC number were not significantly different in both conditions. In addition, the retinal function examined by multifocal electroretinogram were similar at 12 weeks old. These results suggest that lighting conditions may regulate the progression of RGC degeneration in some types of glaucoma.

20.
J Diabetes Investig ; 13(4): 628-637, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34693664

RESUMO

AIMS/INTRODUCTION: Inhibition of peroxisome proliferator-activated receptor gamma (PPARγ) phosphorylation mediated by cyclin-dependent kinase 5 (Cdk5) is one of the main mechanisms of action of antidiabetic drugs. In this study, we analyzed the ocular expression and activation of Cdk5 in patients with proliferative diabetic retinopathy (PDR). MATERIALS AND METHODS: The concentrations of PPARγ, Cdk5 and its activating subunit (p35) were determined in the vitreous body of 24 PDR and 63 control eyes by enzyme-linked immunosorbent assay. In addition, the messenger ribonucleic acid and protein expression levels of PPARγ, Cdk5 and p35 were measured in proliferative neovascular membranes from seven PDR eyes and non-neovascular epiretinal membranes from five control eyes by quantitative real-time polymerase chain reaction and immunohistochemical analysis. RESULTS: PPARγ, Cdk5 and p35 concentrations in the vitreous body were significantly higher in the PDR group compared with the control group. There was also a positive significant correlation of Cdk5 with PPARγ and p35 in the PDR group. Furthermore, the messenger ribonucleic acid expression levels of PPARγ, Cdk5 and p35 in proliferative neovascular membranes were significantly higher in the PDR group compared with the control group. Immunostaining showed increased protein expression levels of PPARγ, Cdk5 and p35 in proliferative neovascular membranes in the PDR group compared with the control group. CONCLUSIONS: Cdk5 activation is involved in PDR pathogenesis through PPARγ expression, and inhibition of Cdk5-mediated PPARγ phosphorylation might be a new therapeutic target for treatment of PDR.


Assuntos
Quinase 5 Dependente de Ciclina/metabolismo , Diabetes Mellitus , Retinopatia Diabética , Diabetes Mellitus/metabolismo , Retinopatia Diabética/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , PPAR gama/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Corpo Vítreo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA