Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
Invest Ophthalmol Vis Sci ; 61(6): 46, 2020 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-32572457

RESUMO

Purpose: Diabetic retinopathy (DR) is a leading cause of visual impairment. Müller cells in DR are dysfunctional due to downregulation of the inwardly rectifying potassium channel Kir4.1. Metformin, a commonly used oral antidiabetic drug, is known to elicit its action through 5' adenosine monophosphate-activated protein kinase (AMPK), a cellular metabolic regulator; however, its effect on Kir4.1 channels is unknown. For this study, we hypothesized that metformin treatment would correct circadian rhythm disruption and Kir4.1 channel dysfunction in db/db mice. Methods: Metformin was given orally to db/db mice. Wheel-running activity, retinal levels of Kir4.1, and AMPK phosphorylation were determined at study termination. In parallel, rat retinal Müller cell line (rMC-1) cells were treated using metformin and 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) to assess the effect of AMPK activation on the Kir4.1 channel. Results: The wheel-running activity of the db/db mice was improved following the metformin treatment. The Kir4.1 level in Müller cells was corrected after metformin treatment. Metformin treatment led to an upregulation of clock regulatory genes such as melanopsin (Opn4) and aralkylamine N-acetyltransferase (Aanat). In rMC-1 cells, AMPK activation via AICAR and metformin resulted in increased Kir4.1 and intermediate core clock component Bmal-1 protein expression. The silencing of Prkaa1 (gene for AMPKα1) led to decreased Kir4.1 and Bmal-1 protein expression. Conclusions: Our findings demonstrate that metformin corrects abnormal circadian rhythm and Kir4.1 channels in db/db mouse a model of type 2 diabetes. Metformin could represent a critical pharmacological agent for preventing Müller cell dysfunction observed in human DR.


Assuntos
Ritmo Circadiano/fisiologia , Diabetes Mellitus Experimental/tratamento farmacológico , Retinopatia Diabética/tratamento farmacológico , Regulação da Expressão Gênica , Metformina/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização/genética , Células Ganglionares da Retina/metabolismo , Animais , Células Cultivadas , Ritmo Circadiano/efeitos dos fármacos , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Retinopatia Diabética/genética , Retinopatia Diabética/metabolismo , Modelos Animais de Doenças , Hipoglicemiantes/farmacologia , Masculino , Camundongos , Camundongos Transgênicos , Canais de Potássio Corretores do Fluxo de Internalização/biossíntese , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/patologia
2.
Stem Cell Reports ; 13(1): 147-162, 2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31204303

RESUMO

Mutations in the gene encoding the type II transmembrane protease 3 (TMPRSS3) cause human hearing loss, although the underlying mechanisms that result in TMPRSS3-related hearing loss are still unclear. We combined the use of stem cell-derived inner ear organoids with single-cell RNA sequencing to investigate the role of TMPRSS3. Defective Tmprss3 leads to hair cell apoptosis without altering the development of hair cells and the formation of the mechanotransduction apparatus. Prior to degeneration, Tmprss3-KO hair cells demonstrate reduced numbers of BK channels and lower expressions of genes encoding calcium ion-binding proteins, suggesting a disruption in intracellular homeostasis. A proteolytically active TMPRSS3 was detected on cell membranes in addition to ER of cells in inner ear organoids. Our in vitro model recapitulated salient features of genetically associated inner ear abnormalities and will serve as a powerful tool for studying inner ear disorders.


Assuntos
Orelha Interna/patologia , Células Ciliadas Auditivas Internas/patologia , Proteínas de Membrana/genética , Organoides/citologia , Serina Proteases/genética , Animais , Apoptose/genética , Membrana Celular/metabolismo , Códon sem Sentido , Técnicas de Inativação de Genes , Homeostase/genética , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Mecanotransdução Celular , Proteínas de Membrana/metabolismo , Camundongos , Análise de Sequência de RNA , Serina Proteases/metabolismo , Análise de Célula Única
3.
Invest Ophthalmol Vis Sci ; 60(6): 1928-1936, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31042800

RESUMO

Purpose: Diabetes leads to the downregulation of the retinal Kir4.1 channels and Müller cell dysfunction. The insulin receptor substrate-1 (IRS-1) is a critical regulator of insulin signaling in Müller cells. Circadian rhythms play an integral role in normal physiology; however, diabetes leads to a circadian dysrhythmia. We hypothesize that diabetes will result in a circadian dysrhythmia of IRS-1 and Kir4.1 and disturbed clock gene function will have a critical role in regulating Kir4.1 channels. Methods: We assessed a diurnal rhythm of retinal IRS-1 and Kir4.1 in db/db mice. The Kir4.1 function was evaluated using a whole-cell recording of Müller cells. The rat Müller cells (rMC-1) were used to undertake in vitro studies using a siRNA. Results: The IRS-1 exhibited a diurnal rhythm in control mice; however, with diabetes, this natural rhythm was lost. The Kir4.1 levels peaked and troughed at times similar to the IRS-1 rhythm. The IRS-1 silencing in the rMC-1 led to a decrease in Kir4.1 and BMAL1. The insulin treatment of retinal explants upregulated Kir4.1 possibly via upregulation of BMAL1 and phosphorylation of IRS-1 and Akt-1. Conclusions: Our studies highlight that IRS-1, by regulating BMAL1, is an important regulator of Kir4.1 in Müller cells and the dysfunctional signaling mediated by IRS-1 may be detrimental to Kir4.1.


Assuntos
Fatores de Transcrição ARNTL/genética , Ritmo Circadiano/fisiologia , Retinopatia Diabética/genética , Células Ependimogliais/metabolismo , Regulação da Expressão Gênica , Proteínas Substratos do Receptor de Insulina/genética , Canais de Potássio Corretores do Fluxo de Internalização/genética , Fatores de Transcrição ARNTL/biossíntese , Animais , Western Blotting , Células Cultivadas , Diabetes Mellitus Experimental , Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Células Ependimogliais/patologia , Humanos , Proteínas Substratos do Receptor de Insulina/biossíntese , Camundongos , Reação em Cadeia da Polimerase , Canais de Potássio Corretores do Fluxo de Internalização/biossíntese , RNA/genética , Ratos
4.
Am J Pathol ; 187(6): 1426-1435, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28432873

RESUMO

The brain and muscle aryl hydrocarbon receptor nuclear translocator-like protein (BMAL)-1 constitutes a major transcriptional regulator of the circadian clock. Here, we explored the impact of conditional deletion of Bmal1 in endothelium and hematopoietic cells in murine models of microvascular and macrovascular injury. We used two models of Bmal1fx/fx;Tek-Cre mice, a retinal ischemia/reperfusion model and a neointimal hyperplasia model of the femoral artery. Eyes were enumerated for acellular capillaries and were stained for oxidative damage markers using nitrotyrosine immunohistochemistry. LSK (lineage-negative, stem cell antigen-1-positive, c-Kit-positive) cells were quantified and proliferation assessed. Hematopoiesis is influenced by innervation to the bone marrow, which we assessed using IHC analysis. The number of acellular capillaries increased threefold, and nitrotyrosine staining increased 1.5-fold, in the retinas of Bmal1fx/fx;Tek-Cre mice. The number of LSK cells from the Bmal1fx/fx;Tek-Cre mice decreased by 1.5-fold and was accompanied by a profound decrease in proliferative potential. Bmal1fx/fx;Tek-Cre mice also exhibited evidence of bone marrow denervation, demonstrating a loss of neurofilament-200 staining. Injured femoral arteries showed a 20% increase in neointimal hyperplasia compared with similarly injured wild-type controls. Our study highlights the importance of the circadian clock in maintaining vascular homeostasis and demonstrates that specific deletion of BMAL1 in endothelial and hematopoietic cells results in phenotypic features similar to those of diabetes.


Assuntos
Fatores de Transcrição ARNTL/fisiologia , Neointima/patologia , Traumatismo por Reperfusão/metabolismo , Vasos Retinianos/metabolismo , Fatores de Transcrição ARNTL/deficiência , Fatores de Transcrição ARNTL/genética , Animais , Medula Óssea/metabolismo , Medula Óssea/patologia , Capilares/patologia , Proliferação de Células , Ritmo Circadiano/fisiologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Artéria Femoral/lesões , Artéria Femoral/patologia , Deleção de Genes , Células-Tronco Hematopoéticas/patologia , Hiperplasia , Antígenos Comuns de Leucócito/análise , Contagem de Leucócitos , Camundongos Transgênicos , Óxido Nítrico Sintase Tipo III/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Traumatismo por Reperfusão/patologia , Retina/metabolismo , Vasos Retinianos/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA