Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
J Cell Biol ; 223(12)2024 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-39352499

RESUMO

Successful axonal regeneration following injury requires the effective allocation of energy. How axons withstand the initial disruption in mitochondrial energy production caused by the injury and subsequently initiate regrowth is poorly understood. Transcriptomic data showed increased expression of glycolytic genes after optic nerve crush in retinal ganglion cells with the co-deletion of Pten and Socs3. Using retinal cultures in a multicompartment microfluidic device, we observed increased regrowth and enhanced mitochondrial trafficking in the axons of Pten and Socs3 co-deleted neurons. While wild-type axons relied on mitochondrial metabolism, after injury, in the absence of Pten and Socs3, energy production was supported by local glycolysis. Specific inhibition of lactate production hindered injury survival and the initiation of regrowth while slowing down glycolysis upstream impaired regrowth initiation, axonal elongation, and energy production. Together, these observations reveal that glycolytic ATP, combined with sustained mitochondrial transport, is essential for injury-induced axonal regrowth, providing new insights into the metabolic underpinnings of axonal regeneration.


Assuntos
Axônios , Glicólise , Mitocôndrias , Regeneração Nervosa , Células Ganglionares da Retina , Animais , Axônios/metabolismo , Regeneração Nervosa/genética , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Mitocôndrias/metabolismo , Mitocôndrias/genética , Camundongos , Traumatismos do Nervo Óptico/metabolismo , Traumatismos do Nervo Óptico/patologia , Traumatismos do Nervo Óptico/genética , PTEN Fosfo-Hidrolase/metabolismo , PTEN Fosfo-Hidrolase/genética , Camundongos Endogâmicos C57BL , Trifosfato de Adenosina/metabolismo , Metabolismo Energético/genética
2.
Exp Eye Res ; 246: 110017, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39097072

RESUMO

Loss of retinal ganglion cells (RGCs) is central to the pathogenesis of optic neuropathies such as glaucoma. Increased RGC cAMP signaling is neuroprotective. We have shown that displacement of the cAMP-specific phosphodiesterase PDE4D3 from an RGC perinuclear compartment by expression of the modified PDE4D3 N-terminal peptide 4D3(E) increases perinuclear cAMP and protein kinase A activity in cultured neurons and in vivo RGC survival after optic nerve crush (ONC) injury. To explore mechanisms by which PDE4D3 displacement promotes neuroprotection, in this study mice intravitreally injected with an adeno-associated virus to express an mCherry-tagged 4D3(E) peptide were subjected to ONC injury and analyzed by single cell RNA-sequencing (scRNA-seq). 4D3(E)-mCherry expression was associated with an attenuation of injury-induced changes in gene expression, thereby supporting the hypothesis that enhanced perinuclear PKA signaling promotes neuroprotective RGC gene expression.


Assuntos
Camundongos Endogâmicos C57BL , Traumatismos do Nervo Óptico , Células Ganglionares da Retina , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Células Ganglionares da Retina/efeitos dos fármacos , Animais , Camundongos , Traumatismos do Nervo Óptico/metabolismo , Traumatismos do Nervo Óptico/tratamento farmacológico , Traumatismos do Nervo Óptico/genética , Regulação da Expressão Gênica , Modelos Animais de Doenças , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/genética , Compressão Nervosa , Sobrevivência Celular , Injeções Intravítreas , Transdução de Sinais , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Masculino , Células Cultivadas
3.
Exp Eye Res ; 235: 109627, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37619829

RESUMO

The main purpose of this study is to analyze the effects of unilateral optic nerve crush in the gene expression of pro- and anti-inflammatory mediators, and gliosis markers in injured and contralateral retinas. Retinas from intact, unilaterally optic nerve injured or sham-operated C57BL/6J mice were analyzed 1, 3, 9 and 30 days after the surgery (n = 5/group and time point) and the relative expression of TGF-ß1, IL-1ß, TNF-α, Iba1, AQP4, GFAP, MHCII, and TSPO was analyzed in injured and contralateral using qPCR. The results indicated that compared with intact retinas, sham-operated animals showed an early (day 1) upregulation of IL-1ß, TNF-α and TSPO and a late (day 30) upregulation of TNF-α. In sham-contralateral retinas, TNF-α and TSPO mRNA expression were upregulated and day 30 while GFAP, Iba1, AQP4 and MHCII downregulated at day 9. Compared with sham-operated animals, in retinas affected by optic nerve crush GFAP and TSPO upregulated at day 1 and TNF-α, Iba1, AQP4 and MHCII at day 3. In the crushed-contralateral retinas, TGF-ß1, TNF-α, Iba1 and MHCII were upregulated at day 1. TSPO was upregulated up to day 30 whereas TGF-ß1 and Iba1 downregulated after day 9. In conclusion, both sham surgery and optic nerve crush changed the profile of inflammatory and gliosis markers in the injured and contralateral retinas, changes that were more pronounced for optic nerve crush when compared to sham.


Assuntos
Traumatismos do Nervo Óptico , Fator de Crescimento Transformador beta1 , Camundongos , Animais , Fator de Crescimento Transformador beta1/farmacologia , Células Ganglionares da Retina/metabolismo , Gliose/metabolismo , Traumatismos do Nervo Óptico/genética , Traumatismos do Nervo Óptico/metabolismo , Doenças Neuroinflamatórias , Fator de Necrose Tumoral alfa/metabolismo , Camundongos Endogâmicos C57BL , Retina/metabolismo , Nervo Óptico/metabolismo , Compressão Nervosa/métodos
5.
Development ; 150(8)2023 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-37039265

RESUMO

Central nervous system projection neurons fail to spontaneously regenerate injured axons. Targeting developmentally regulated genes in order to reactivate embryonic intrinsic axon growth capacity or targeting pro-growth tumor suppressor genes such as Pten promotes long-distance axon regeneration in only a small subset of injured retinal ganglion cells (RGCs), despite many RGCs regenerating short-distance axons. A recent study identified αRGCs as the primary type that regenerates short-distance axons in response to Pten inhibition, but the rare types which regenerate long-distance axons, and cellular features that enable such response, remained unknown. Here, we used a new method for capturing specifically the rare long-distance axon-regenerating RGCs, and also compared their transcriptomes with embryonic RGCs, in order to answer these questions. We found the existence of adult non-α intrinsically photosensitive M1 RGC subtypes that retained features of embryonic cell state, and showed that these subtypes partially dedifferentiated towards an embryonic state and regenerated long-distance axons in response to Pten inhibition. We also identified Pten inhibition-upregulated mitochondria-associated genes, Dynlt1a and Lars2, which promote axon regeneration on their own, and thus present novel therapeutic targets.


Assuntos
Aminoacil-tRNA Sintetases , Traumatismos do Nervo Óptico , Aminoacil-tRNA Sintetases/metabolismo , Axônios/fisiologia , Mitocôndrias , Regeneração Nervosa/fisiologia , Traumatismos do Nervo Óptico/genética , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Células Ganglionares da Retina/metabolismo
6.
Proc Natl Acad Sci U S A ; 119(15): e2113751119, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35394873

RESUMO

Although mammalian retinal ganglion cells (RGCs) normally cannot regenerate axons nor survive after optic nerve injury, this failure is partially reversed by inducing sterile inflammation in the eye. Infiltrative myeloid cells express the axogenic protein oncomodulin (Ocm) but additional, as-yet-unidentified, factors are also required. We show here that infiltrative macrophages express stromal cell­derived factor 1 (SDF1, CXCL12), which plays a central role in this regard. Among many growth factors tested in culture, only SDF1 enhances Ocm activity, an effect mediated through intracellular cyclic AMP (cAMP) elevation and phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) activation. SDF1 deficiency in myeloid cells (CXCL12flx/flxLysM-Cre−/+ mice) or deletion of the SDF1 receptor CXCR4 in RGCs (intraocular AAV2-Cre in CXCR4flx/flx mice) or SDF1 antagonist AMD3100 greatly suppresses inflammation-induced regeneration and decreases RGC survival to baseline levels. Conversely, SDF1 induces optic nerve regeneration and RGC survival, and, when combined with Ocm/cAMP, SDF1 increases axon regeneration to levels similar to those induced by intraocular inflammation. In contrast to deletion of phosphatase and tensin homolog (Pten), which promotes regeneration selectively from αRGCs, SDF1 promotes regeneration from non-αRGCs and enables the latter cells to respond robustly to Pten deletion; however, SDF1 surprisingly diminishes the response of αRGCs to Pten deletion. When combined with inflammation and Pten deletion, SDF1 enables many RGCs to regenerate axons the entire length of the optic nerve. Thus, SDF1 complements the effects of Ocm in mediating inflammation-induced regeneration and enables different RGC subtypes to respond to Pten deletion.


Assuntos
Traumatismos do Nervo Óptico , Células Ganglionares da Retina , Axônios/metabolismo , Quimiocina CXCL12/genética , Monócitos/metabolismo , Regeneração Nervosa/fisiologia , Traumatismos do Nervo Óptico/genética , Traumatismos do Nervo Óptico/metabolismo , PTEN Fosfo-Hidrolase/genética , Células Ganglionares da Retina/fisiologia
7.
Cells ; 10(7)2021 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-34359839

RESUMO

Neuroinflammation has been put forward as a mechanism triggering axonal regrowth in the mammalian central nervous system (CNS), yet little is known about the underlying cellular and molecular players connecting these two processes. In this study, we provide evidence that MMP2 is an essential factor linking inflammation to axonal regeneration by using an in vivo mouse model of inflammation-induced axonal regeneration in the optic nerve. We show that infiltrating myeloid cells abundantly express MMP2 and that MMP2 deficiency results in reduced long-distance axonal regeneration. However, this phenotype can be rescued by restoring MMP2 expression in myeloid cells via a heterologous bone marrow transplantation. Furthermore, while MMP2 deficiency does not affect the number of infiltrating myeloid cells, it does determine the coordinated expression of pro- and anti-inflammatory molecules. Altogether, in addition to its role in axonal regeneration via resolution of the glial scar, here, we reveal a new mechanism via which MMP2 facilitates axonal regeneration, namely orchestrating the expression of pro- and anti-inflammatory molecules by infiltrating innate immune cells.


Assuntos
Axônios/imunologia , Transplante de Medula Óssea , Metaloproteinase 2 da Matriz/genética , Regeneração Nervosa/imunologia , Traumatismos do Nervo Óptico/imunologia , Nervo Óptico/imunologia , Animais , Antígenos Ly/genética , Antígenos Ly/imunologia , Axônios/ultraestrutura , Receptor 1 de Quimiocina CX3C/genética , Receptor 1 de Quimiocina CX3C/imunologia , Movimento Celular , Proteína GAP-43/genética , Proteína GAP-43/imunologia , Regulação da Expressão Gênica , Imunidade Inata , Inflamação , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/imunologia , Metaloproteinase 2 da Matriz/deficiência , Metaloproteinase 2 da Matriz/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/citologia , Células Mieloides/imunologia , Regeneração Nervosa/genética , Nervo Óptico/metabolismo , Traumatismos do Nervo Óptico/genética , Traumatismos do Nervo Óptico/patologia , Retina/imunologia , Retina/lesões , Retina/metabolismo , Transplante Heterólogo , Irradiação Corporal Total
8.
Invest Ophthalmol Vis Sci ; 62(10): 14, 2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34398198

RESUMO

Purpose: Inhibition or targeted deletion of histone deacetylase 3 (HDAC3) is neuroprotective in a variety neurodegenerative conditions, including retinal ganglion cells (RGCs) after acute optic nerve damage. Consistent with this, induced HDAC3 expression in cultured cells shows selective toxicity to neurons. Despite an established role for HDAC3 in neuronal pathology, little is known regarding the mechanism of this pathology. Methods: Induced expression of an HDAC3-mCherry fusion protein in mouse RGCs was accomplished by transduction with AAV2/2-Pgk-HDAC3-mCherry. Increased susceptibility to optic nerve damage in HDAC3-mCherry expressing RGCs was evaluated in transduced mice that received acute optic nerve crush surgery. Expression of HDAC3-FLAG or HDAC3-mCherry was induced by nucleofection or transfection of plasmids into differentiated or undifferentiated 661W tissue culture cells. Immunostaining for cleaved caspase 3, localization of a GFP-BAX fusion protein, and quantitative RT-PCR was used to evaluate HDAC3-induced damage. Results: Induced expression of exogenous HDAC3 in RGCs by viral-mediated gene transfer resulted in modest levels of cell death but significantly increased the sensitivity of these neurons to axonal damage. Undifferentiated 661W retinal precursor cells were resilient to induced HDAC3 expression, but after differentiation, HDAC3 induced GFP-BAX recruitment to the mitochondria and BAX/BAK dependent activation of caspase 3. This was accompanied by an increase in accumulation of transcripts for the JNK2/3 kinases and the p53-regulated BH3-only gene Bbc3/Puma. Cell cycle arrest of undifferentiated 661W cells did not increase their sensitivity to HDAC3 expression. Conclusions: Collectively, these results indicate that HDAC3-induced toxicity to neurons is mediated by the intrinsic apoptotic pathway.


Assuntos
Apoptose/genética , Regulação da Expressão Gênica , Histona Desacetilases/genética , Neurônios/metabolismo , Traumatismos do Nervo Óptico/genética , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Histona Desacetilases/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/patologia , Traumatismos do Nervo Óptico/metabolismo , Traumatismos do Nervo Óptico/patologia , Células Ganglionares da Retina/patologia , Transdução de Sinais
9.
Exp Cell Res ; 406(1): 112737, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34324864

RESUMO

The retina is the innermost part of the eye of most vertebrates and it is essential for vision. The development, maintenance, and function of this laminated structure is tightly regulated by numerous genes. Deficiencies in the expression of these genes as well as deregulation of various molecular mechanisms can cause retinopathies and blindness. MicroRNAs (miRNAs) are one of the most important and effective molecular regulatory mechanisms that underlie the biology of the retina. miRNAs have specific functional roles in the development and maintenance of different retinal layers and retinal cell types. While previous studies have reported a large number of miRNAs linked to development, maintenance and diseases of the retina, no comprehensive study has properly discussed and integrated data from these studies. Given the particular importance of miR-204 in retinal biology, we intend to critically discuss the expression and functional significance of this miRNA in the development, maintenance, and pathologies of the retina. Moreover, we explore biological processes through which miR-204 influences retinal pathophysiology. This review highlights the crucial functions of miR-204 in the retina and suggests the putative mechanism of miR-204 action in retinal biology.


Assuntos
Retinopatia Diabética/genética , Glaucoma/genética , Degeneração Macular/genética , MicroRNAs/genética , Traumatismos do Nervo Óptico/genética , Retinoblastoma/genética , Animais , Sequência de Bases , Sequência Conservada , Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Proteína 1 Homóloga a Discs-Large/genética , Proteína 1 Homóloga a Discs-Large/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Glaucoma/metabolismo , Glaucoma/patologia , Humanos , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , MicroRNAs/metabolismo , Traumatismos do Nervo Óptico/metabolismo , Traumatismos do Nervo Óptico/patologia , Proteína Companheira de mTOR Insensível à Rapamicina/genética , Proteína Companheira de mTOR Insensível à Rapamicina/metabolismo , Retina/metabolismo , Retina/patologia , Retinoblastoma/metabolismo , Retinoblastoma/patologia , Transdução de Sinais
10.
Cell Rep ; 34(9): 108777, 2021 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-33657370

RESUMO

Adult mammalian central nervous system (CNS) trauma interrupts neural networks and, because axonal regeneration is minimal, neurological deficits persist. Repair via axonal growth is limited by extracellular inhibitors and cell-autonomous factors. Based on results from a screen in vitro, we evaluate nearly 400 genes through a large-scale in vivo regeneration screen. Suppression of 40 genes using viral-driven short hairpin RNAs (shRNAs) promotes retinal ganglion cell (RGC) axon regeneration after optic nerve crush (ONC), and most are validated by separate CRISPR-Cas9 editing experiments. Expression of these axon-regeneration-suppressing genes is not significantly altered by axotomy. Among regeneration-limiting genes, loss of the interleukin 22 (IL-22) cytokine allows an early, yet transient, inflammatory response in the retina after injury. Reduced IL-22 drives concurrent activation of signal transducer and activator of transcription 3 (Stat3) and dual leucine zipper kinase (DLK) pathways and upregulation of multiple neuron-intrinsic regeneration-associated genes (RAGs). Including IL-22, our screen identifies dozens of genes that limit CNS regeneration. Suppression of these genes in the context of axonal damage could support improved neural repair.


Assuntos
Regeneração Nervosa/genética , Neurogênese/genética , Traumatismos do Nervo Óptico/genética , Nervo Óptico/metabolismo , Animais , Axônios/metabolismo , Axônios/patologia , Sistemas CRISPR-Cas , Dependovirus/genética , Feminino , Edição de Genes , Regulação da Expressão Gênica , Estudos de Associação Genética , Células HEK293 , Humanos , Interleucinas/genética , Interleucinas/metabolismo , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Nervo Óptico/patologia , Nervo Óptico/fisiopatologia , Traumatismos do Nervo Óptico/metabolismo , Traumatismos do Nervo Óptico/patologia , Traumatismos do Nervo Óptico/fisiopatologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Interleucina 22
11.
Cell Death Dis ; 12(2): 213, 2021 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-33637688

RESUMO

Axonal damage is an early step in traumatic and neurodegenerative disorders of the central nervous system (CNS). Damaged axons are not able to regenerate sufficiently in the adult mammalian CNS, leading to permanent neurological deficits. Recently, we showed that inhibition of the autophagic protein ULK1 promotes neuroprotection in different models of neurodegeneration. Moreover, we demonstrated previously that axonal protection improves regeneration of lesioned axons. However, whether axonal protection mediated by ULK1 inhibition could also improve axonal regeneration is unknown. Here, we used an adeno-associated viral (AAV) vector to express a dominant-negative form of ULK1 (AAV.ULK1.DN) and investigated its effects on axonal regeneration in the CNS. We show that AAV.ULK1.DN fosters axonal regeneration and enhances neurite outgrowth in vitro. In addition, AAV.ULK1.DN increases neuronal survival and enhances axonal regeneration after optic nerve lesion, and promotes long-term axonal protection after spinal cord injury (SCI) in vivo. Interestingly, AAV.ULK1.DN also increases serotonergic and dopaminergic axon sprouting after SCI. Mechanistically, AAV.ULK1.DN leads to increased ERK1 activation and reduced expression of RhoA and ROCK2. Our findings outline ULK1 as a key regulator of axonal degeneration and regeneration, and define ULK1 as a promising target to promote neuroprotection and regeneration in the CNS.


Assuntos
Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Axônios/metabolismo , Dependovirus/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Regeneração Nervosa , Traumatismos do Nervo Óptico/terapia , Nervo Óptico/metabolismo , Traumatismos da Medula Espinal/terapia , Medula Espinal/metabolismo , Animais , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/genética , Axônios/patologia , Células Cultivadas , Modelos Animais de Doenças , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Regulação para Baixo , Feminino , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Crescimento Neuronal , Nervo Óptico/patologia , Traumatismos do Nervo Óptico/genética , Traumatismos do Nervo Óptico/metabolismo , Traumatismos do Nervo Óptico/patologia , Ratos Wistar , Neurônios Serotoninérgicos/metabolismo , Neurônios Serotoninérgicos/patologia , Medula Espinal/patologia , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Fatores de Tempo , Proteínas rho de Ligação ao GTP/metabolismo , Quinases Associadas a rho/metabolismo
12.
Nature ; 588(7836): 124-129, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33268865

RESUMO

Ageing is a degenerative process that leads to tissue dysfunction and death. A proposed cause of ageing is the accumulation of epigenetic noise that disrupts gene expression patterns, leading to decreases in tissue function and regenerative capacity1-3. Changes to DNA methylation patterns over time form the basis of ageing clocks4, but whether older individuals retain the information needed to restore these patterns-and, if so, whether this could improve tissue function-is not known. Over time, the central nervous system (CNS) loses function and regenerative capacity5-7. Using the eye as a model CNS tissue, here we show that ectopic expression of Oct4 (also known as Pou5f1), Sox2 and Klf4 genes (OSK) in mouse retinal ganglion cells restores youthful DNA methylation patterns and transcriptomes, promotes axon regeneration after injury, and reverses vision loss in a mouse model of glaucoma and in aged mice. The beneficial effects of OSK-induced reprogramming in axon regeneration and vision require the DNA demethylases TET1 and TET2. These data indicate that mammalian tissues retain a record of youthful epigenetic information-encoded in part by DNA methylation-that can be accessed to improve tissue function and promote regeneration in vivo.


Assuntos
Envelhecimento/genética , Reprogramação Celular/genética , Metilação de DNA , Epigênese Genética , Olho , Regeneração Nervosa/genética , Visão Ocular/genética , Visão Ocular/fisiologia , Envelhecimento/fisiologia , Animais , Axônios/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular , Proteínas de Ligação a DNA/genética , Dependovirus/genética , Dioxigenases , Modelos Animais de Doenças , Olho/citologia , Olho/inervação , Olho/patologia , Feminino , Vetores Genéticos/genética , Glaucoma/genética , Glaucoma/patologia , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Camundongos Endogâmicos C57BL , Fator 3 de Transcrição de Octâmero/genética , Traumatismos do Nervo Óptico/genética , Proteínas Proto-Oncogênicas/genética , Células Ganglionares da Retina/citologia , Fatores de Transcrição SOXB1/genética , Transcriptoma/genética
13.
Int J Mol Sci ; 21(19)2020 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-33007835

RESUMO

Glaucoma is a progressive chronic retinal degenerative disease and a leading cause of global irreversible blindness, characterized by optic nerve damage and retinal ganglion cell (RGC) death. Elevated intraocular pressure (IOP) is a main risk factor of glaucoma. Neuroinflammation plays an important role in glaucoma. We have been demonstrating that elevated pressure triggers microglia reactivity that contribute to the loss of RGCs. Adenosine, acting on adenosine receptors, is a crucial modulator of microglia phenotype. Microglia express all adenosine receptors. Previously, we demonstrated that the activation of adenosine A3 receptor (A3R) affords protection to the retina, including RGCs, unveiling the possibility for a new strategy for glaucoma treatment. Since microglial cells express A3R, we now studied the ability of a selective A3R agonist (2-Cl-IB-MECA) in controlling microglia reactivity induced by elevated hydrostatic pressure (EHP), used to mimic elevated IOP. The activation of A3R reduced EHP-induced inducible nitric oxide synthase (iNOS) expression, microglia migration and phagocytosis in BV-2 cells. In retinal microglia, proliferation and phagocytosis elicited by EHP were also decreased by A3R activation. This work demonstrates that 2-Cl-IB-MECA, the selective agonist of A3R, is able to hinder microglia reactivity, suggesting that A3R agonists could afford protection against glaucomatous degeneration through the control of neuroinflammation.


Assuntos
Agonistas do Receptor A3 de Adenosina/farmacologia , Adenosina/análogos & derivados , Glaucoma/tratamento farmacológico , Receptor A3 de Adenosina/genética , Adenosina/genética , Adenosina/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Glaucoma/genética , Glaucoma/patologia , Humanos , Pressão Intraocular/efeitos dos fármacos , Microglia/efeitos dos fármacos , Nervo Óptico/efeitos dos fármacos , Nervo Óptico/patologia , Traumatismos do Nervo Óptico/tratamento farmacológico , Traumatismos do Nervo Óptico/genética , Traumatismos do Nervo Óptico/patologia , Fagocitose/efeitos dos fármacos , Ratos , Degeneração Retiniana/tratamento farmacológico , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/patologia
14.
Exp Cell Res ; 396(1): 112159, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32652081

RESUMO

Traumatic optic neuropathy is a common clinical problem. Damage to the optic nerve leads to shear stress and triggers secondary swelling within the optic canal. The study aims to explore the role of the inflammatory response following optic nerve injury (ONI) in toll-like receptor-9 knockout mice (TLR-9-/-) compared to wild-type mice (WT). At first, TLR-9-/- and WT mice were subjected to ONI. We then found that ONI significantly up-regulated TLR-9 expression levels in retinal tissues of WT mice. The retinal degeneration after ONI was alleviated in TLR-9-/- mice, as evidenced by the increased number of retinal ganglion cells (RGCs) and thickness of inner retinal layer (IRL). TUNEL staining and immunofluorescence staining of BRN3A indicated that TLR-9 knockout effectively improved the survival of RGCs. ONI-enhanced expression of Iba-1 and TMEM119 was markedly reduced in TLR-9-/- mice, indicating the suppression of microglial activation. Moreover, production of pro-inflammatory regulators, including inducible nitric oxide synthase (iNOS), macrophage chemo-attractant protein (MCP)-1, cyclooxygenase-2 (COX-2), interleukin (IL)-1ß, IL-18 and tumor necrosis factor-α (TNF-α), was significantly decreased in TLR-9-/- mice following ONI. TLR-9 knockout-attenuated inflammation was mainly through repressing myeloid differentiation factor 88 (MyD88) and IL-1 receptor-associated kinase 4 (IRAK4). Furthermore, ONI greatly up-regulated the protein expression levels of phosphorylated (p)-IKKα, p-IκBα and p-nuclear factor (NF)-κB, whereas being repressed in TLR-9-/- mice. The effects of TLR-9 on ONI were verified in lipopolysaccharide (LPS)-stimulated retinal microglial cells transfected with small interfering RNA TLR-9 (siTLR-9). As expected, promoting TLR-9 with its agonist markedly restored inflammation in TLR-9 knockdown cells stimulated by LPS. Therefore, all findings above suggested that suppressing TLR-9 showed neuroprotective effects against ONI through reducing inflammatory response, and TILR-9 might be a promising therapeutic target to develop effective strategies for the treatment of optic neuropathies.


Assuntos
Microglia/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Traumatismos do Nervo Óptico/genética , Nervo Óptico/metabolismo , Células Ganglionares da Retina/metabolismo , Receptor Toll-Like 9/genética , Animais , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Contagem de Células , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Regulação da Expressão Gênica , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Inflamação , Quinases Associadas a Receptores de Interleucina-1/genética , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Interleucina-18/genética , Interleucina-18/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Microglia/patologia , Fator 88 de Diferenciação Mieloide/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Nervo Óptico/patologia , Traumatismos do Nervo Óptico/metabolismo , Traumatismos do Nervo Óptico/patologia , Células Ganglionares da Retina/patologia , Transdução de Sinais , Receptor Toll-Like 9/deficiência , Fator de Transcrição Brn-3A/genética , Fator de Transcrição Brn-3A/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
15.
J Recept Signal Transduct Res ; 40(2): 97-108, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32019426

RESUMO

Background: Cisplatin (CiSP), a chemotherapeutic agent, is widely used to treat several types of cancers. However, its clinical use is limited due to adverse side effects caused by excessive production of reactive oxygen species (ROS) and death of neurons. The transient receptor potential (TRP) melastatin 2 (TRPM2) cation channel is activated by ADP-ribose (ADPR) and ROS. The protective effect of curcumin (CURCU) against CiSP-induced apoptosis and mitochondrial ROS through inhibition of TRP channels in several types of neuron except optic nerve, was recently reported. The aim of the current study is to clarify the protective effect of CURCU on CiSP-induced mitochondrial oxidative injury and TRPM2 activation in the mice optic nerve and SH-SY5Y human derived neuronal cells.Material and methods: The SH-SY5Y cells and mice were divided into four groups: Control, CURCU, CiSP, and CURCU + CiSP. The mice were treated for 14 days and the cells were incubated with CiSP and CURCU for 24 h.Results: CURCU and PARP-1 inhibitor (PJ34) treatments ameliorated CiSP-induced mitochondrial membrane depolarization, mitochondrial and cytosolic ROS levels and neuronal death in the optic nerve. In the patch-clamp of SH-SY5Y cells and laser confocal microscopy experiments of optic nerve, CURCU and TRPM2 blocker treatments also decreased ADPR-induced TRPM2 currents and cytosolic free calcium ion (Ca2+) concentration, suggesting a suppression of Ca2+ influx and neuronal death.Conclusion: CURCU prevents CiSP-induced optic nerve oxidative injury and cell death by suppressing mitochondrial ROS production via regulating TRPM2 signaling pathways. CURCU may serve as a potential therapeutic target against CiSP-induced toxicity in the optic nerve of CiSP-treated patients.


Assuntos
Curcumina/farmacologia , Traumatismos do Nervo Óptico/tratamento farmacológico , Nervo Óptico/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Canais de Cátion TRPM/genética , Adenosina Difosfato Ribose/genética , Animais , Apoptose/efeitos dos fármacos , Cisplatino/toxicidade , Humanos , Camundongos , Mitocôndrias/efeitos dos fármacos , Neoplasias/complicações , Neoplasias/tratamento farmacológico , Neoplasias/genética , Nervo Óptico/patologia , Traumatismos do Nervo Óptico/induzido quimicamente , Traumatismos do Nervo Óptico/genética , Traumatismos do Nervo Óptico/patologia , Técnicas de Patch-Clamp , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
16.
Restor Neurol Neurosci ; 37(6): 545-552, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31839616

RESUMO

BACKGROUND: Permanent loss of vital functions after central nervous system (CNS) injury occurs in part because axons in the adult mammalian CNS do not regenerate after injury. PTEN was identified as a prominent intrinsic inhibitor of CNS axon regeneration about 10 years ago. The PTEN negatively regulated PI3K-AKT-mTOR pathway, which has been intensively explored in diverse models of axon injury and diseases and its mechanism for axon regeneration is becoming clearer. OBJECTIVE: It is timely to summarize current knowledge about the PTEN/AKT/mTOR pathway and discuss future directions of translational regenerative research for neural injury and neurodegenerative diseases. METHODS: Using mouse optic nerve crush as an in vivo retinal ganglion cell axon injury model, we have conducted an extensive molecular dissection of the PI3K-AKT-mTORC1/mTORC2 pathway to illuminate the cross-regulating mechanisms in axon regeneration. RESULTS: AKT is the nodal point that coordinates both positive (PI3K-PDK1-pAKT-T308) and negative (PI3K-mTORC2-pAKT-S473) signals to regulate adult CNS axon regeneration through two parallel pathways, activating mTORC1 and inhibiting GSK3ß. However, mTORC1/S6K1-mediated feedback inhibition after PTEN deletion prevents potent AKT activation. CONCLUSIONS: A key permissive signal from an unidentified AKT-independent pathway is required for stimulating the neuron-intrinsic growth machinery. Future studies into this complex neuron-intrinsic balancing mechanism involving necessary and permissive signals for axon regeneration is likely to lead to safe and effective regenerative strategies for CNS repair.


Assuntos
Regeneração Nervosa/fisiologia , Traumatismos do Nervo Óptico/metabolismo , PTEN Fosfo-Hidrolase/deficiência , Transdução de Sinais/fisiologia , Animais , Camundongos , Nervo Óptico/metabolismo , Nervo Óptico/patologia , Traumatismos do Nervo Óptico/genética , Traumatismos do Nervo Óptico/patologia , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
17.
Invest Ophthalmol Vis Sci ; 60(4): 965-977, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30884523

RESUMO

Purpose: Retinal ischemia, a common cause of several vision-threatening diseases, contributes to the death of retinal neurons, particularly retinal ganglion cells (RGCs). Heat shock transcription factor 1 (HSF1), a stress-responsive protein, has been shown to be important in response to cellular stress stimuli, including ischemia. This study is to investigate whether HSF1 has a role in retinal neuronal injury in a mouse model of retinal ischemia-reperfusion (IR). Methods: IR was induced by inserting an infusion needle into the anterior chamber of the right eye and elevating a saline reservoir connected to the needle to raise the intraocular pressure to 110 mm Hg for 45 minutes. HSF1, Hsp70, molecules in the endoplasmic reticulum (ER) stress branches, tau phosphorylation, inflammatory molecules, and RGC injury were determined by immunohistochemistry, Western blot, or quantitative PCR. Results: HSF1 expression was significantly increased in the retina 6 hours after IR. Using our novel transgenic mice carrying full-length human HSF gene, we demonstrated that IR-induced retinal neuronal apoptosis and necroptosis were abrogated 12 hours after IR. RGCs and their function were preserved in the HSF1 transgenic mice 7 days after IR. Mechanistically, the beneficial effects of HSF1 may be mediated by its induction of chaperone protein Hsp70 and alleviation of ER stress, leading to decreased tau phosphorylation and attenuated inflammatory response 12 to 24 hours after IR. Conclusions: These data provide compelling evidence that HSF1 is neuroprotective against retinal IR injury, and boosting HSF1 expression may be a beneficial strategy to limit neuronal degeneration in retinal diseases.


Assuntos
Regulação da Expressão Gênica/fisiologia , Fatores de Transcrição de Choque Térmico/genética , Traumatismos do Nervo Óptico/genética , Traumatismo por Reperfusão/genética , Doenças Retinianas/genética , Animais , Western Blotting , Retículo Endoplasmático/metabolismo , Proteínas de Choque Térmico HSP70/genética , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Leucostasia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Compressão Nervosa , Neuroproteção/fisiologia , Traumatismos do Nervo Óptico/prevenção & controle , Fosforilação , Reação em Cadeia da Polimerase em Tempo Real , Traumatismo por Reperfusão/prevenção & controle , Doenças Retinianas/prevenção & controle , Tomografia de Coerência Óptica , Proteínas tau/metabolismo
18.
Anat Rec (Hoboken) ; 301(11): 1968-1976, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30299595

RESUMO

Pim-1 is a proto-oncogene which has been discovered to involve in cell proliferation, differentiation, and survival. In this study, we observed the expression of Pim-1 in neonatal and adult rat retina and the changes in rat retina following optic nerve crush (ONC) in order to explore the relationship between Pim-1 and the survival of retinal ganglion cells (RGC). We discovered that Pim-1 was distributed mainly in retinal pigment epithelial cells (RPE) and retinal ganglion cell layer (GCL) in normal newborn rats, and it appeared in RPE, cone rod cell layer and GCL in normal adult rats by immunohistochemistry. Our double immunofluorescent staining of Pim-1 and γ-synuclein further confirmed that Pim-1 was localized in 80% of RGC. Moreover, we found that the amount of Pim-1 mRNA and protein in adult rat retina was transiently increased after ONC and then decreased 2 weeks after ONC, and the expression level was lower than that of neonatal rat retina under all conditions. We also discovered that Pim-1 expression in GCL detected by immunohistochemistry was upregulated at Day 1 and Day 3 after ONC, but downregulated at Day 14 after ONC when the survival of RGC was decreased and the apoptotic cells in GCL were increased by hematoxylin-eosin staining, immunohistochemistry, and TUNEL detection. We suggest that the overexpression of Pim-1 in the RGC is related to the optic nerve repair while the low expression of Pim-1 in RGC may be associated with apoptosis and weak intrinsic regeneration ability of RGC. Anat Rec, 301:1968-1976, 2018. © 2018 Wiley Periodicals, Inc.


Assuntos
Compressão Nervosa/efeitos adversos , Traumatismos do Nervo Óptico/metabolismo , Nervo Óptico/metabolismo , Proteínas Proto-Oncogênicas c-pim-1/biossíntese , Retina/metabolismo , Animais , Animais Recém-Nascidos , Expressão Gênica , Masculino , Nervo Óptico/química , Traumatismos do Nervo Óptico/genética , Proteínas Proto-Oncogênicas c-pim-1/genética , Ratos , Ratos Sprague-Dawley , Retina/química , Células Ganglionares da Retina/química , Células Ganglionares da Retina/metabolismo
19.
Cell Death Dis ; 9(10): 1007, 2018 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-30258047

RESUMO

Previous studies have demonstrated that intravitreal delivery of brain-derived neurotrophic factor (BDNF) by injection of recombinant protein or by gene therapy can alleviate retinal ganglion cell (RGC) loss after optic nerve injury. BDNF gene therapy can improve RGC survival in experimental models of glaucoma, the leading cause of irreversible blindness worldwide. However, the therapeutic efficacy of BDNF supplementation alone is time limited at least in part due to BDNF receptor downregulation. Tropomyosin-related receptor kinase-B (TrkB) downregulation has been reported in many neurological diseases including glaucoma, potentially limiting the effect of sustained or repeated BDNF delivery.Here, we characterize a novel adeno-associated virus (AAV) gene therapy (AAV2 TrkB-2A-mBDNF) that not only increases BDNF production but also improves long-term neuroprotective signaling by increasing expression of the BDNF receptor (TrkB) within the inner retina. This approach leads to significant and sustained elevation of survival signaling pathways ERK and AKT within RGCs over 6 months and avoids the receptor downregulation which we observe with treatment with AAV2 BDNF alone. We validate the neuroprotective efficacy of AAV2 TrkB-2A-mBDNF in a mouse model of optic nerve injury, where it outperforms conventional AAV2 BDNF or AAV2 TrkB therapy, before showing powerful proof of concept neuroprotection of RGCs and axons in a rat model of chronic intraocular pressure (IOP) elevation. We also show that there are no adverse effects of the vector on retinal structure or function as assessed by histology and electroretinography in young or aged animals. Further studies are underway to explore the potential of this vector as a candidate for progression into clinical studies to protect RGCs in patients with glaucoma and progressive visual loss despite conventional IOP-lowering treatment.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Glicoproteínas de Membrana/genética , Neuroproteção/genética , Receptor trkB/genética , Células Ganglionares da Retina/patologia , Transdução de Sinais/genética , Animais , Axônios/patologia , Dependovirus/genética , Modelos Animais de Doenças , Regulação para Baixo/genética , Terapia Genética/métodos , Glaucoma/genética , Glaucoma/patologia , Células HEK293 , Humanos , Pressão Intraocular/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismos do Nervo Óptico/genética , Traumatismos do Nervo Óptico/patologia , Ratos , Ratos Sprague-Dawley , Retina/patologia
20.
Adv Exp Med Biol ; 1074: 387-393, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29721968

RESUMO

Factor XIII-A (FXIII-A), which has become known as cellular transglutaminase, plays important roles in mediating cross-linking reactions in various tissues. FXIII-A acts as one of the regeneration molecules in the fish retina and optic nerve after optic nerve injury and becomes activated at the site of injury within a few hours. Previous research has shown that activated FXIII-A induces neurite outgrowth from injured retinal ganglion cells and supports elongation of the regenerating optic nerve. However, the activation mechanism of FXIII-A remains unknown. Furthermore, the injured tissues do not express thrombin, a known activator of plasma FXIII. Here, we investigated the mRNA expression of FXIII-A based on two different regions, one encoding the activation peptide and the other encoding the enzymatic active site. We found that expression of the region encoding the activation peptide was markedly suppressed compared with the region encoding the active site. An overexpression study with a short-type FXIII-A cDNA lacking the activation peptide revealed induction of long neurite outgrowth in fish retinal explant cultures compared with full-length FXIII-A cDNA. The present findings suggest that alternative splicing may occur in the FXIII-A gene, resulting in deletion of the region encoding the activation peptide and thus allowing direct production of activated FXIII-A protein in the fish retina and optic nerve after optic nerve injury.


Assuntos
Processamento Alternativo , Proteínas do Olho/genética , Fator XIIIa/metabolismo , Traumatismos do Nervo Óptico/genética , RNA Mensageiro/genética , Proteínas de Peixe-Zebra/genética , Animais , Axônios/ultraestrutura , Ativação Enzimática , Proteínas do Olho/biossíntese , Proteínas do Olho/fisiologia , Regulação da Expressão Gênica , Carpa Dourada , Peptídeos e Proteínas de Sinalização Intercelular , Compressão Nervosa , Regeneração Nervosa , Traumatismos do Nervo Óptico/metabolismo , Técnicas de Cultura de Órgãos , Peptídeos/metabolismo , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Recombinantes/metabolismo , Deleção de Sequência , Peixe-Zebra , Proteínas de Peixe-Zebra/biossíntese , Proteínas de Peixe-Zebra/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA