Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Mol Ther ; 27(2): 424-441, 2019 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-30341011

RESUMO

Glaucoma is characterized by the loss of retinal ganglion cells (RGC), and accordingly the preservation of RGCs and their axons has recently attracted significant attention to improve therapeutic outcomes in the disease. Here, we report that Src homology region 2-containing protein tyrosine phosphatase 2 (Shp2) undergoes activation in the RGCs, in animal model of glaucoma as well as in the human glaucoma tissues and that Shp2 dephosphorylates tropomyosin receptor kinase B (TrkB) receptor, leading to reduced BDNF/TrkB neuroprotective survival signaling. This was elucidated by specifically modulating Shp2 expression in the RGCs in vivo, using adeno-associated virus serotype 2 (AAV2) constructs. Shp2 upregulation promoted endoplasmic reticulum (ER) stress and apoptosis, along with functional and structural deficits in the inner retina. In contrast, loss of Shp2 decelerated the loss of RGCs, preserved their function, and suppressed ER stress and apoptosis in glaucoma. This report constitutes the first identification of Shp2-mediated TrkB regulatory mechanisms in the RGCs that can become a potential therapeutic target in both glaucoma and other neurodegenerative disorders.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Receptor trkB/metabolismo , Células Ganglionares da Retina/metabolismo , Animais , Western Blotting , Fator Neurotrófico Derivado do Encéfalo/genética , Modelos Animais de Doenças , Eletroforese em Gel de Poliacrilamida , Eletrorretinografia , Glaucoma/metabolismo , Glaucoma/patologia , Masculino , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Ratos , Ratos Sprague-Dawley , Receptor trkB/genética , Retina/citologia , Retina/metabolismo , Células Ganglionares da Retina/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
2.
Cell Mol Life Sci ; 73(22): 4279-4297, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27333888

RESUMO

Accumulation of amyloid ß (Aß) and its aggregates in the ageing central nervous system is regarded synonymous to Alzheimer's disease (AD) pathology. Despite unquestionable advances in mechanistic and diagnostic aspects of the disease understanding, the primary cause of Aß accumulation as well as its in vivo roles remains elusive; nonetheless, the majority of the efforts to address pathological mechanisms for therapeutic development are focused towards moderating Aß accumulation in the brain. More recently, Aß deposition has been identified in the eye and is linked with distinct age-related diseases including age-related macular degeneration, glaucoma as well as AD. Awareness of the Aß accumulation in these markedly different degenerative disorders has led to an increasing body of work exploring overlapping mechanisms, a prospective biomarker role for Aß and the potential to use retina as a model for brain related neurodegenerative disorders. Here, we present an integrated view of current understanding of the retinal Aß deposition discussing the accumulation mechanisms, anticipated impacts and outlining ameliorative approaches that can be extrapolated to the retina for potential therapeutic benefits. Further longitudinal investigations in humans and animal models will determine retinal Aß association as a potential pathognomonic, diagnostic or prognostic biomarker.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Encéfalo/patologia , Doenças Neurodegenerativas/metabolismo , Retina/patologia , Animais , Encéfalo/metabolismo , Humanos , Inflamação/patologia , Agregados Proteicos , Retina/metabolismo
3.
Clin Exp Ophthalmol ; 45(9): 911-922, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28516453

RESUMO

BACKGROUND: The DBA/2J mouse has been described as a model for congenital experimental glaucoma. It develops anterior segment anomalies with synechiae and pigment dispersion leading to raised intraocular pressure and glaucomatous damage. However, there are serious practical considerations when using this model in longitudinal studies. METHODS: We followed 118 mice from 12-48 weeks of age in a pharmaceutical trial. Here we report on the findings in control animals (n = 37). Intraocular pressure was measured weekly, electrophysiology and optical coherence tomography every 6 weeks. A subset also had invasive intraocular pressure measurements performed prior to euthanasia. RESULTS: Although intraocular pressure eventually rose by 9 months in most animals, tonometry was complicated by corneal calcification in the majority of animals rendering intraocular pressure measurement unreliable. Invasive intraocular pressure did not correlate with non-invasive measures. Loss of scotopic threshold response and thinning of inner retinal layers on optical coherence tomography was observed over time, suggesting glaucomatous damage, but this occurred in some animals without raised intraocular pressure. Poor pupil dilation significantly affected electrophysiology, optical coherence tomography and fundus imaging; 22% of animals developed major systemic complications leading to high dropout rate. CONCLUSIONS: The DBA/2J experimental glaucoma model shows variability in expression, and its pathological changes cause major difficulties in assessing disease progression. From our experience, the model presents significant challenges for drug studies in glaucoma, as there are many confounding factors: difficulty with accurate intraocular pressure measurement, in vivo imaging, and electrophysiology recording and a high dropout rate. In addition, there may be an underlying neurodegenerative process independent of intraocular pressure.


Assuntos
Glaucoma/diagnóstico , Pressão Intraocular/fisiologia , Retina/patologia , Tomografia de Coerência Óptica/métodos , Animais , Córnea/patologia , Modelos Animais de Doenças , Progressão da Doença , Eletrorretinografia , Feminino , Glaucoma/fisiopatologia , Iris/patologia , Masculino , Camundongos , Camundongos Endogâmicos DBA , Retina/fisiopatologia , Índice de Gravidade de Doença
4.
Proteomes ; 12(1)2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38535505

RESUMO

Neural regeneration and neuroprotection represent strategies for future management of neurodegenerative disorders such as Alzheimer's disease (AD) or glaucoma. However, the complex molecular mechanisms that are involved in neuroprotection are not clearly understood. A promising candidate that maintains neuroprotective signaling networks is neuroserpin (Serpini1), a serine protease inhibitor expressed in neurons which selectively inhibits extracellular tissue-type plasminogen activator (tPA)/plasmin and plays a neuroprotective role during ischemic brain injury. Abnormal function of this protein has been implicated in several conditions including stroke, glaucoma, AD, and familial encephalopathy with neuroserpin inclusion bodies (FENIB). Here, we explore the potential biochemical roles of Serpini1 by comparing proteome changes between neuroserpin-deficient (NS-/-) and control mice, in the retina (RE), optic nerve (ON), frontal cortex (FC), visual cortex (VC), and cerebellum (CB). To achieve this, a multiple-plex quantitative proteomics approach using isobaric tandem mass tag (TMT) technology was employed followed by functional enrichment and protein-protein interaction analysis. We detected around 5000 proteins in each tissue and a pool of 6432 quantified proteins across all regions, resulting in a pool of 1235 differentially expressed proteins (DEPs). Principal component analysis and hierarchical clustering highlighted similarities and differences in the retina compared to various brain regions, as well as differentiating NS-/- proteome signatures from control samples. The visual cortex revealed the highest number of DEPs, followed by cerebellar regions. Pathway analysis unveiled region-specific changes, including visual perception, focal adhesion, apoptosis, glutamate receptor activation, and supramolecular fiber organization in RE, ON, FC, VC, and CB, respectively. These novel findings provide comprehensive insights into the region-specific networking of Serpini1 in the central nervous system, further characterizing its potential role as a neuroprotective agent. Data are available via ProteomeXchange with identifier PXD046873.

5.
Mol Neurobiol ; 60(12): 7222-7237, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37542647

RESUMO

Glaucoma is a leading cause of permanent blindness worldwide and is characterized by neurodegeneration linked to progressive retinal ganglion cell (RGC) death, axonal damage, and neuroinflammation. Glutamate excitotoxicity mediated through N-methyl-D-aspartate (NMDA) receptors plays a crucial role in glaucomatous RGC loss. Sphingosine 1-phosphate receptors (S1PRs) are important mediators of neurodegeneration and neuroinflammation in the brain and the retina. Siponimod is an immunomodulatory drug for multiple sclerosis and is a selective modulator of S1PR subtypes 1 and 5 and has been shown to have beneficial effects on the central nervous system (CNS) in degenerative conditions. Our previous study showed that mice administered orally with siponimod protected inner retinal structure and function against acute NMDA excitotoxicity. To elucidate the molecular mechanisms behind these protective effects, we investigated the inflammatory pathways affected by siponimod treatment in NMDA excitotoxicity model. NMDA excitotoxicity resulted in the activation of glial cells coupled with upregulation of the inflammatory NF-kB pathway and increased expression of TNFα, IL1-ß, and IL-6. Siponimod treatment significantly reduced glial activation and suppressed the pro-inflammatory pathways. Furthermore, NMDA-induced activation of NLRP3 inflammasome and upregulation of neurotoxic inducible nitric oxide synthase (iNOS) were significantly diminished with siponimod treatment. Our data demonstrated that siponimod induces anti-inflammatory effects via suppression of glial activation and inflammatory singling pathways that could protect the retina against acute excitotoxicity conditions. These findings provide insights into the anti-inflammatory effects of siponimod in the CNS and suggest a potential therapeutic strategy for neuroinflammatory conditions.


Assuntos
Glaucoma , N-Metilaspartato , Camundongos , Animais , N-Metilaspartato/metabolismo , Doenças Neuroinflamatórias , Retina/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Glaucoma/metabolismo , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Anti-Inflamatórios/metabolismo
6.
Int J Tryptophan Res ; 15: 11786469221126063, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36329761

RESUMO

Autosomal recessive polycystic kidney disease (ARPKD) is an early onset genetic disorder characterized by numerous renal cysts resulting in end stage renal disease. Our study aimed to determine if metabolic reprogramming and tryptophan (Trp) metabolism via the kynurenine pathway (KP) is a critical dysregulated pathway in PKD. Using the Lewis polycystic kidney (LPK) rat model of PKD and Lewis controls, we profiled temporal trends for KP metabolites in plasma, urine, and kidney tissues from 6- and 12-week-old mixed sex animals using liquid and gas chromatography, minimum n = 5 per cohort. A greater kynurenine (KYN) concentration was observed in LPK kidney and plasma of 12-week rats compared to age matched Lewis controls (P ⩽ .05). LPK kidneys also showed an age effect (P ⩽ .05) with KYN being greater in 12-week versus 6-week LPK. The metabolites xanthurenic acid (XA), 3-hydroxykynurenine (3-HK), and 3-hydroxyanthranilic acid (3-HAA) were significantly greater in the plasma of 12-week LPK rats compared to age matched Lewis controls (P ⩽ .05). Plasma XA and 3-HK also showed an age effect (P ⩽ .05) being greater in 12-week versus 6-week LPK. We further describe a decrease in Trp levels in LPK plasma and kidney (strain effect P ⩽ .05). There were no differences in KP metabolites in urine between cohorts. Using the ratio of product and substrates in the KP, a significant age-strain effect (P ⩽ .05) was observed in the activity of the KYN/Trp ratio (tryptophan-2,3-dioxygenase [TDO] or indoleamine-2,3-dioxygenase [IDO] activity), kynurenine 3-monooxygenase (KMO), KAT A (kynurenine aminotransferase A), KAT B, total KAT, total KYNU (kynureninase), KYNU A, KYNU B, and total KYNU within LPK kidneys, supporting an activated KP. Confirmation of the activation of these enzymes will require verification through orthogonal techniques. In conclusion, we have demonstrated an up-regulation of the KP in alignment with progression of renal impairment in the LPK rat model, suggesting that KP activation may be a critical contributor to the pathobiology of PKD.

7.
Sci Rep ; 7(1): 8412, 2017 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-28827627

RESUMO

Neuroserpin is a serine protease inhibitor that regulates the activity of plasmin and its activators in the neuronal tissues. This study provides novel evidence of regulatory effect of the neuroserpin on plasmin proteolytic activity in the retina in glaucoma. Human retinal and vitreous tissues from control and glaucoma subjects as well as retinas from experimental glaucoma rats were analysed to establish changes in plasmin and neuroserpin activity. Neuroserpin undergoes oxidative inactivation in glaucoma which leads to augmentation of plasmin activity. Neuroserpin contains several methionine residues in addition to a conserved reactive site methionine and our study revealed enhanced oxidation of Met residues in the serpin under glaucoma conditions. Met oxidation was associated with loss of neuroserpin inhibitory activity and similar findings were observed in the retinas of superoxide dismutase (SOD) mutant mice that have increased oxidative stress. Treatment of purified neuroserpin with H2O2 further established that Met oxidation inversely correlated with its plasmin inhibitory activity. Dysregulation of the plasmin proteolytic system associated with increased degradation of the extracellular matrix (ECM) proteins in the retina. Collectively, these findings delineate a novel molecular basis of plasmin activation in glaucoma and potentially for other neuronal disorders with implications in disease associated ECM remodelling.


Assuntos
Fibrinolisina/metabolismo , Fibrinolíticos/metabolismo , Glaucoma/patologia , Neuropeptídeos/metabolismo , Inibidores de Serina Proteinase/metabolismo , Serpinas/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Modelos Animais de Doenças , Humanos , Metionina/metabolismo , Camundongos , Oxirredução , Ratos , Retina/patologia , Neuroserpina
8.
Data Brief ; 6: 776-82, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26909388

RESUMO

TrkB is a high affinity receptor for the brain derived neurotrophic factor (BDNF) and its phosphorylation stimulates activation of several intracellular signalling pathways linked to cellular growth, differentiation and maintenance. Identification of various activators and inhibitors of the TrkB receptor and greater understanding their binding mechanisms is critical to elucidate the biochemical and pharmacological pathways and analyse various protein crystallization studies. The data presented here is related to the research article entitled "Brain Derived neurotrophic factor is involved in the regulation of glycogen synthase kinase 3ß (GSK3ß) signalling" [1]. Cyclotraxin B (CTXB) is a disulphide bridge linked cyclic peptide molecule that interacts with TrkB receptor and inhibits the BDNF/TrkB downstream signalling. This article reports for the first time binding mechanism and interaction parameters of CTXB with the TrkB receptor. The molecular model of CTXB has been generated and it's docking with TrkB domain carried out to determine the critical residues involved in the protein peptide interaction.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA