Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
EMBO Mol Med ; 14(1): e14511, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-34779136

RESUMO

In the course of our studies aiming to discover vascular bed-specific endothelial cell (EC) mitogens, we identified leukemia inhibitory factor (LIF) as a mitogen for bovine choroidal EC (BCE), although LIF has been mainly characterized as an EC growth inhibitor and an anti-angiogenic molecule. LIF stimulated growth of BCE while it inhibited, as previously reported, bovine aortic EC (BAE) growth. The JAK-STAT3 pathway mediated LIF actions in both BCE and BAE cells, but a caspase-independent proapoptotic signal mediated by cathepsins was triggered in BAE but not in BCE. LIF administration directly promoted activation of STAT3 and increased blood vessel density in mouse eyes. LIF also had protective effects on the choriocapillaris in a model of oxidative retinal injury. Analysis of available single-cell transcriptomic datasets shows strong expression of the specific LIF receptor in mouse and human choroidal EC. Our data suggest that LIF administration may be an innovative approach to prevent atrophy associated with AMD, through protection of the choriocapillaris.


Assuntos
Atrofia Geográfica , Fator Inibidor de Leucemia , Mitógenos , Animais , Corioide/irrigação sanguínea , Corioide/metabolismo , Células Endoteliais/metabolismo , Atrofia Geográfica/metabolismo , Janus Quinases/metabolismo , Fator Inibidor de Leucemia/metabolismo , Fator Inibidor de Leucemia/farmacologia , Camundongos , Mitógenos/metabolismo , Mitógenos/farmacologia , Fator de Transcrição STAT3/metabolismo
2.
Diabetes ; 70(3): 665-679, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33303689

RESUMO

The contribution of altered mitochondrial Ca2+ handling to metabolic and functional defects in type 2 diabetic (T2D) mouse hearts is not well understood. In this study, we show that the T2D heart is metabolically inflexible and almost exclusively dependent on mitochondrial fatty acid oxidation as a consequence of mitochondrial calcium uniporter complex (MCUC) inhibitory subunit MCUb overexpression. Using a recombinant endonuclease-deficient Cas9-based gene promoter pulldown approach coupled with mass spectrometry, we found that MCUb is upregulated in the T2D heart due to loss of glucose homeostasis regulator nuclear receptor corepressor 2 repression, and chromatin immunoprecipitation assays identified peroxisome proliferator-activated receptor α as a mediator of MCUb gene expression in T2D cardiomyocytes. Upregulation of MCUb limits mitochondrial matrix Ca2+ uptake and impairs mitochondrial energy production via glucose oxidation by depressing pyruvate dehydrogenase complex activity. Gene therapy displacement of endogenous MCUb with a dominant-negative MCUb transgene (MCUbW246R/V251E) in vivo rescued T2D cardiomyocytes from metabolic inflexibility and stimulated cardiac contractile function and adrenergic responsiveness by enhancing phospholamban phosphorylation via protein kinase A. We conclude that MCUb represents one newly discovered molecular effector at the interface of metabolism and cardiac function, and its repression improves the outcome of the chronically stressed diabetic heart.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Mitocondriais/metabolismo , Correpressor 2 de Receptor Nuclear/metabolismo , PPAR alfa/metabolismo , Animais , Cálcio/metabolismo , Diabetes Mellitus Tipo 2/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Correpressor 2 de Receptor Nuclear/genética , Oxirredução , Espectrometria de Massas em Tandem
3.
Proc Natl Acad Sci U S A ; 115(21): 5468-5473, 2018 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-29735674

RESUMO

Stargardt macular dystrophy 3 (STGD3) is caused by dominant mutations in the ELOVL4 gene. Like other macular degenerations, pathogenesis within the retinal pigment epithelium (RPE) appears to contribute to the loss of photoreceptors from the central retina. However, the RPE does not express ELOVL4, suggesting photoreceptor cell loss in STGD3 occurs through two cell nonautonomous events: mutant photoreceptors first affect RPE cell pathogenesis, and then, second, RPE dysfunction leads to photoreceptor cell death. Here, we have investigated how the RPE pathology occurs, using a STGD3 mouse model in which mutant human ELOVL4 is expressed in the photoreceptors. We found that the mutant protein was aberrantly localized to the photoreceptor outer segment (POS), and that resulting POS phagosomes were degraded more slowly in the RPE. In cell culture, the mutant POSs are ingested by primary RPE cells normally, but the phagosomes are processed inefficiently, even by wild-type RPE. The mutant phagosomes excessively sequester RAB7A and dynein, and have impaired motility. We propose that the abnormal presence of ELOVL4 protein in POSs results in phagosomes that are defective in recruiting appropriate motor protein linkers, thus contributing to slower degradation because their altered motility results in slower basal migration and fewer productive encounters with endolysosomes. In the transgenic mouse retinas, the RPE accumulated abnormal-looking phagosomes and oxidative stress adducts; these pathological changes were followed by pathology in the neural retina. Our results indicate inefficient phagosome degradation as a key component of the first cell nonautonomous event underlying retinal degeneration due to mutant ELOVL4.


Assuntos
Modelos Animais de Doenças , Proteínas do Olho/fisiologia , Degeneração Macular/patologia , Proteínas de Membrana/fisiologia , Mutação , Fagossomos/patologia , Células Fotorreceptoras/patologia , Epitélio Pigmentado da Retina/patologia , Animais , Movimento Celular , Células Cultivadas , Genes Dominantes , Humanos , Degeneração Macular/genética , Degeneração Macular/metabolismo , Camundongos , Camundongos Transgênicos , Fagossomos/metabolismo , Células Fotorreceptoras/metabolismo , Epitélio Pigmentado da Retina/metabolismo
4.
J Biol Chem ; 293(21): 8182-8195, 2018 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-29626093

RESUMO

Diabetes mellitus is a growing health care problem, resulting in significant cardiovascular morbidity and mortality. Diabetes also increases the risk for heart failure (HF) and decreased cardiac myocyte function, which are linked to changes in cardiac mitochondrial energy metabolism. The free mitochondrial calcium level ([Ca2+] m ) is fundamental in activating the mitochondrial respiratory chain complexes and ATP production and is also known to regulate pyruvate dehydrogenase complex (PDC) activity. The mitochondrial calcium uniporter (MCU) complex (MCUC) plays a major role in mediating mitochondrial Ca2+ import, and its expression and function therefore have a marked impact on cardiac myocyte metabolism and function. Here, we investigated MCU's role in mitochondrial Ca2+ handling, mitochondrial function, glucose oxidation, and cardiac function in the heart of diabetic mice. We found that diabetic mouse hearts exhibit altered expression of MCU and MCUC members and a resulting decrease in [Ca2+] m , mitochondrial Ca2+ uptake, mitochondrial energetic function, and cardiac function. Adeno-associated virus-based normalization of MCU levels in these hearts restored mitochondrial Ca2+ handling, reduced PDC phosphorylation levels, and increased PDC activity. These changes were associated with cardiac metabolic reprogramming toward normal physiological glucose oxidation. This reprogramming likely contributed to the restoration of both cardiac myocyte and heart function to nondiabetic levels without any observed detrimental effects. These findings support the hypothesis that abnormal mitochondrial Ca2+ handling and its negative consequences can be ameliorated in diabetes by restoring MCU levels via adeno-associated virus-based MCU transgene expression.


Assuntos
Canais de Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/metabolismo , Diabetes Mellitus Experimental/fisiopatologia , Coração/fisiologia , Mitocôndrias/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Células Cultivadas , Metabolismo Energético , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/citologia
5.
eNeuro ; 4(4)2017.
Artigo em Inglês | MEDLINE | ID: mdl-28828400

RESUMO

Circadian rhythms of mammalian physiology and behavior are coordinated by the suprachiasmatic nucleus (SCN) in the hypothalamus. Within SCN neurons, various aspects of cell physiology exhibit circadian oscillations, including circadian clock gene expression, levels of intracellular Ca2+ ([Ca2+]i), and neuronal firing rate. [Ca2+]i oscillates in SCN neurons even in the absence of neuronal firing. To determine the causal relationship between circadian clock gene expression and [Ca2+]i rhythms in the SCN, as well as the SCN neuronal network dependence of [Ca2+]i rhythms, we introduced GCaMP3, a genetically encoded fluorescent Ca2+ indicator, into SCN neurons from PER2::LUC knock-in reporter mice. Then, PER2 and [Ca2+]i were imaged in SCN dispersed and organotypic slice cultures. In dispersed cells, PER2 and [Ca2+]i both exhibited cell autonomous circadian rhythms, but [Ca2+]i rhythms were typically weaker than PER2 rhythms. This result matches the predictions of a detailed mathematical model in which clock gene rhythms drive [Ca2+]i rhythms. As predicted by the model, PER2 and [Ca2+]i rhythms were both stronger in SCN slices than in dispersed cells and were weakened by blocking neuronal firing in slices but not in dispersed cells. The phase relationship between [Ca2+]i and PER2 rhythms was more variable in cells within slices than in dispersed cells. Both PER2 and [Ca2+]i rhythms were abolished in SCN cells deficient in the essential clock gene Bmal1. These results suggest that the circadian rhythm of [Ca2+]i in SCN neurons is cell autonomous and dependent on clock gene rhythms, but reinforced and modulated by a synchronized SCN neuronal network.


Assuntos
Cálcio/metabolismo , Ritmo Circadiano/fisiologia , Rede Nervosa/fisiologia , Neurônios/fisiologia , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/fisiologia , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Animais , Ritmo Circadiano/efeitos dos fármacos , Ritmo Circadiano/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Técnicas In Vitro , Luciferases/genética , Luciferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Teóricos , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/farmacologia , Transdução Genética , Glicoproteínas da Zona Pelúcida/genética , Glicoproteínas da Zona Pelúcida/metabolismo
6.
Neuroscience ; 357: 1-11, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28576728

RESUMO

In mammals, the master circadian clock resides in the suprachiasmatic nucleus (SCN). The SCN is characterized by robust circadian oscillations of clock gene expression and neuronal firing. The synchronization of circadian oscillations among individual cells in the SCN is attributed to intercellular coupling. Previous studies have shown that gap junctions, specifically those composed of connexin-36 (Cx36) subunits, are required for coupling of electrical firing among SCN neurons at a time scale of milliseconds. However, it remains unknown whether Cx36 gap junctions also contribute to coupling of circadian (∼24h) rhythms of clock gene expression. Here, we investigated circadian expression patterns of the clock gene Period 2 (Per2) in the SCN of Cx36-deficient mice using luminometry and single-cell bioluminescence imaging. Surprisingly, we found that synchronization of circadian PER2 expression rhythms is maintained in SCN explants from Cx36-deficient mice. Since Cx36 expression levels change with age, we also tested circadian running-wheel behavior of juvenile (3-4weeks old) and adult (9-30weeks old) Cx36-deficient mice. We found that impact of connexin-36 expression on circadian behavior changes greatly during postnatal development. However, consistent with the intact synchrony among SCN cells in cultured explants, Cx36-deficient mice had intact locomotor circadian rhythms, although adults displayed a lengthened period in constant darkness. Our data indicate that even though Cx36 may be required for electrical coupling of SCN cells, it does not affect coupling of molecular clock gene rhythms. Thus, electrical coupling of neurons and coupling of circadian clock gene oscillations can be regulated independently in the SCN.


Assuntos
Relógios Circadianos/fisiologia , Ritmo Circadiano/fisiologia , Conexinas/deficiência , Proteínas Circadianas Period/metabolismo , Núcleo Supraquiasmático/metabolismo , Adaptação Fisiológica/fisiologia , Animais , Conexinas/genética , Feminino , Masculino , Camundongos Transgênicos , Núcleo Supraquiasmático/crescimento & desenvolvimento , Técnicas de Cultura de Tecidos , Proteína delta-2 de Junções Comunicantes
7.
J Biol Chem ; 291(51): 26515-26528, 2016 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-27816939

RESUMO

mtDNA damage in cardiac myocytes resulting from increased oxidative stress is emerging as an important factor in the pathogenesis of diabetic cardiomyopathy. A prevalent lesion that occurs in mtDNA damage is the formation of 8-hydroxy-2'-deoxyguanosine (8-OHdG), which can cause mutations when not repaired properly by 8-oxoguanine DNA glycosylase (Ogg1). Although the mtDNA repair machinery has been described in cardiac myocytes, the regulation of this repair has been incompletely investigated. Here we report that the hearts of type 1 diabetic mice, despite having increased Ogg1 protein levels, had significantly lower Ogg1 activity than the hearts of control, non-type 1 diabetic mice. In diabetic hearts, we further observed increased levels of 8-OHdG and an increased amount of mtDNA damage. Interestingly, Ogg1 was found to be highly O-GlcNAcylated in diabetic mice compared with controls. In vitro experiments demonstrated that O-GlcNAcylation inhibits Ogg1 activity, which could explain the mtDNA lesion accumulation observed in vivo Reducing Ogg1 O-GlcNAcylation in vivo by introducing a dominant negative O-GlcNAc transferase mutant (F460A) restored Ogg1 enzymatic activity and, consequently, reduced 8-OHdG and mtDNA damage despite the adverse hyperglycemic milieu. Taken together, our results implicate hyperglycemia-induced O-GlcNAcylation of Ogg1 in increased mtDNA damage and, therefore, provide a new plausible biochemical mechanism for diabetic cardiomyopathy.


Assuntos
Dano ao DNA , DNA Glicosilases/metabolismo , DNA Mitocondrial/metabolismo , Diabetes Mellitus Experimental/metabolismo , Cardiomiopatias Diabéticas/metabolismo , Mitocôndrias Cardíacas/metabolismo , Miócitos Cardíacos/metabolismo , Substituição de Aminoácidos , Animais , DNA Glicosilases/genética , DNA Mitocondrial/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Cardiomiopatias Diabéticas/genética , Cardiomiopatias Diabéticas/patologia , Hiperglicemia/genética , Hiperglicemia/metabolismo , Hiperglicemia/patologia , Masculino , Camundongos , Mitocôndrias Cardíacas/genética , Mutação de Sentido Incorreto
8.
Am J Physiol Cell Physiol ; 311(6): C1005-C1013, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27681178

RESUMO

Diabetic cardiomyopathy is associated with metabolic changes, including decreased glucose oxidation (Gox) and increased fatty acid oxidation (FAox), which result in cardiac energetic deficiency. Diabetic hyperglycemia is a pathophysiological mechanism that triggers multiple maladaptive phenomena. The mitochondrial Ca2+ uniporter (MCU) is the channel responsible for Ca2+ uptake in mitochondria, and free mitochondrial Ca2+ concentration ([Ca2+]m) regulates mitochondrial metabolism. Experiments with cardiac myocytes (CM) exposed to simulated hyperglycemia revealed reduced [Ca2+]m and MCU protein levels. Therefore, we investigated whether returning [Ca2+]m to normal levels in CM by MCU expression could lead to normalization of Gox and FAox with no detrimental effects. Mouse neonatal CM were exposed for 72 h to normal glucose [5.5 mM glucose + 19.5 mM mannitol (NG)], high glucose [25 mM glucose (HG)], or HG + adenoviral MCU expression. Gox and FAox, [Ca2+]m, MCU levels, pyruvate dehydrogenase (PDH) activity, oxidative stress, mitochondrial membrane potential, and apoptosis were assessed. [Ca2+]m and MCU protein levels were reduced after 72 h of HG. Gox was decreased and FAox was increased in HG, PDH activity was decreased, phosphorylated PDH levels were increased, and mitochondrial membrane potential was reduced. MCU expression returned these parameters toward NG levels. Moreover, increased oxidative stress and apoptosis were reduced in HG by MCU expression. We also observed reduced MCU protein levels and [Ca2+]m in hearts from type 1 diabetic mice. Thus we conclude that HG-induced metabolic alterations can be reversed by restoration of MCU levels, resulting in return of [Ca2+]m to normal levels.


Assuntos
Canais de Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/metabolismo , Hiperglicemia/metabolismo , Mitocôndrias/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Proteínas de Transporte de Cátions/metabolismo , Diabetes Mellitus Experimental/metabolismo , Glucose/metabolismo , Potencial da Membrana Mitocondrial/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo/fisiologia
9.
Neurosci Lett ; 619: 49-53, 2016 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-26930624

RESUMO

Lithium is widely used as a treatment of bipolar disorder, a neuropsychiatric disorder associated with disrupted circadian rhythms. Lithium is known to lengthen period and increase amplitude of circadian rhythms. One possible pathway for these effects involves inhibition of glycogen synthase kinase-3ß (GSK-3ß), which regulates degradation of CRY2, a canonical clock protein determining circadian period. CRY1 is also known to play important roles in regulating circadian period and phase, although there is no evidence that it is similarly phosphorylated by GSK-3ß. In this paper, we tested the hypothesis that lithium affects circadian rhythms through CRYs. We cultured fibroblasts and slices of the suprachiasmatic nucleus (SCN), the master circadian pacemaker of the brain, from Cry1-/-, Cry2-/-, or wild-type (WT) mice bearing the PER2:LUC circadian reporter. Lithium was applied in the culture medium, and circadian rhythms of PER2 expression were measured. In WT and Cry2-/- fibroblasts, 10mM lithium increased PER2 expression and rhythm amplitude but not period, and 1mM lithium did not affect either period or amplitude. In non-rhythmic Cry1-/- fibroblasts, 10mM lithium increased PER2 expression. In SCN slices, 1mM lithium lengthened period ∼1h in all genotypes, but did not affect amplitude except in Cry2-/- SCN. Thus, the amplitude-enhancing effect of lithium in WT fibroblasts was unaffected by Cry2 knockout and occurred in the absence of period-lengthening, whereas the period-lengthening effect of lithium in WT SCN was unaffected by Cry1 or Cry2 knockout and occurred in the absence of rhythm amplification, suggesting that these two effects of lithium on circadian rhythms are independent of CRYs and of each other.


Assuntos
Antimaníacos/farmacologia , Ritmo Circadiano/efeitos dos fármacos , Criptocromos/genética , Fibroblastos/efeitos dos fármacos , Cloreto de Lítio/farmacologia , Núcleo Supraquiasmático/efeitos dos fármacos , Animais , Antimaníacos/efeitos adversos , Células Cultivadas , Ritmo Circadiano/genética , Fibroblastos/fisiologia , Técnicas In Vitro , Cloreto de Lítio/efeitos adversos , Camundongos Knockout , Proteínas Circadianas Period/metabolismo , Núcleo Supraquiasmático/fisiologia
10.
PLoS Genet ; 12(2): e1005882, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26895328

RESUMO

Heterodimers of CLOCK and BMAL1 are the major transcriptional activators of the mammalian circadian clock. Because the paralog NPAS2 can substitute for CLOCK in the suprachiasmatic nucleus (SCN), the master circadian pacemaker, CLOCK-deficient mice maintain circadian rhythms in behavior and in tissues in vivo. However, when isolated from the SCN, CLOCK-deficient peripheral tissues are reportedly arrhythmic, suggesting a fundamental difference in circadian clock function between SCN and peripheral tissues. Surprisingly, however, using luminometry and single-cell bioluminescence imaging of PER2 expression, we now find that CLOCK-deficient dispersed SCN neurons and peripheral cells exhibit similarly stable, autonomous circadian rhythms in vitro. In CLOCK-deficient fibroblasts, knockdown of Npas2 leads to arrhythmicity, suggesting that NPAS2 can compensate for loss of CLOCK in peripheral cells as well as in SCN. Our data overturn the notion of an SCN-specific role for NPAS2 in the molecular circadian clock, and instead indicate that, at the cellular level, the core loops of SCN neuron and peripheral cell circadian clocks are fundamentally similar.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas CLOCK/deficiência , Relógios Circadianos , Proteínas do Tecido Nervoso/metabolismo , Animais , Proteínas CLOCK/metabolismo , Fibroblastos/metabolismo , Deleção de Genes , Técnicas de Silenciamento de Genes , Camundongos Knockout , Neurônios/metabolismo , Transdução de Sinais , Núcleo Supraquiasmático/metabolismo
11.
Adv Exp Med Biol ; 854: 751-5, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26427485

RESUMO

The retinal pigment epithelium (RPE) is a post-mitotic epithelial monolayer situated between the light-sensitive photoreceptors and the choriocapillaris. Given its vital functions for healthy vision, the RPE is a primary target for insults that result in blinding diseases, including age-related macular degeneration (AMD). One such function is the phagocytosis and digestion of shed photoreceptor outer segments. In the present study, we examined the process of trafficking of outer segment disk membranes in live cultures of primary mouse RPE, using high speed spinning disk confocal microscopy. This approach has enabled us to track phagosomes, and determine parameters of their motility, which are important for their efficient degradation.


Assuntos
Microscopia Confocal/métodos , Fagossomos/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Imagem com Lapso de Tempo/métodos , Animais , Células Cultivadas , Cinética , Camundongos , Fagocitose , Cultura Primária de Células , Segmento Externo das Células Fotorreceptoras da Retina/metabolismo , Epitélio Pigmentado da Retina/citologia
12.
J Cell Biol ; 210(4): 595-611, 2015 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-26261180

RESUMO

The degradation of phagosomes, derived from the ingestion of photoreceptor outer segment (POS) disk membranes, is a major role of the retinal pigment epithelium (RPE). Here, POS phagosomes were observed to associate with myosin-7a, and then kinesin-1, as they moved from the apical region of the RPE. Live-cell imaging showed that the phagosomes moved bidirectionally along microtubules in RPE cells, with kinesin-1 light chain 1 (KLC1) remaining associated in both directions and during pauses. Lack of KLC1 did not inhibit phagosome speed, but run length was decreased, and phagosome localization and degradation were impaired. In old mice, lack of KLC1 resulted in RPE pathogenesis that was strikingly comparable to aspects of age-related macular degeneration (AMD), with an excessive accumulation of RPE and sub-RPE deposits, as well as oxidative and inflammatory stress responses. These results elucidate mechanisms of POS phagosome transport in relation to degradation, and demonstrate that defective microtubule motor transport in the RPE leads to phenotypes associated with AMD.


Assuntos
Degeneração Macular/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Fagossomos/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Animais , Transporte Biológico , Células Cultivadas , Ativação do Complemento , Cinesinas , Degeneração Macular/genética , Degeneração Macular/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Miosina VIIa , Miosinas/metabolismo , Estresse Oxidativo , Segmento Externo das Células Fotorreceptoras da Retina/metabolismo , Segmento Externo das Células Fotorreceptoras da Retina/patologia
13.
Adv Exp Med Biol ; 801: 725-31, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24664764

RESUMO

Usher syndrome type 1B, which is characterized by congenital deafness and progressive retinal degeneration, is caused by the loss of the function of MYO7A. Prevention of the retinal degeneration should be possible by delivering functional MYO7A to retinal cells. Although this approach has been used successfully in clinical trials for Leber congenital amaurosis (LCA2), it remains a challenge for Usher 1B because of the large size of the MYO7A cDNA. Different viral vectors have been tested for use in MYO7A gene therapy. Here, we review approaches with lentiviruses, which can accommodate larger genes, as well as attempts to use adeno-associated virus (AAV), which has a smaller packaging capacity. In conclusion, both types of viral vector appear to be effective. Despite concerns about the ability of lentiviruses to access the photoreceptor cells, a phenotype of the photoreceptors of Myo7a-mutant mice can be corrected. And although MYO7A cDNA is significantly larger than the nominal carrying capacity of AAV, AAV-MYO7A in single vectors also corrected Myo7a-mutant phenotypes in photoreceptor and RPE cells. Interestingly, however, a dual AAV vector approach was found to be much less effective.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Lentivirus/genética , Miosinas/genética , Síndromes de Usher/terapia , Animais , Vetores Genéticos , Humanos , Camundongos , Camundongos Knockout , Miosina VIIa , Síndromes de Usher/genética
14.
Hum Mol Genet ; 19(21): 4229-38, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20709808

RESUMO

Age-related macular degeneration (AMD) is characterized by the loss or dysfunction of retinal pigment epithelium (RPE) and is the most common cause of vision loss among the elderly. Stem-cell-based strategies, using human embryonic stem cells (hESCs) or human-induced pluripotent stem cells (hiPSCs), may provide an abundant donor source for generating RPE cells in cell replacement therapies. Despite a significant amount of research on deriving functional RPE cells from various stem cell sources, it is still unclear whether stem-cell-derived RPE cells fully mimic primary RPE cells. In this report, we demonstrate that functional RPE cells can be derived from multiple lines of hESCs and hiPSCs with varying efficiencies. Stem-cell-derived RPE cells exhibit cobblestone-like morphology, transcripts, proteins and phagocytic function similar to human fetal RPE (fRPE) cells. In addition, we performed global gene expression profiling of stem-cell-derived RPE cells, native and cultured fRPE cells, undifferentiated hESCs and fibroblasts to determine the differentiation state of stem-cell-derived RPE cells. Our data indicate that hESC-derived RPE cells closely resemble human fRPE cells, whereas hiPSC-derived RPE cells are in a unique differentiation state. Furthermore, we identified a set of 87 signature genes that are unique to human fRPE and a majority of these signature genes are shared by stem-cell-derived RPE cells. These results establish a panel of molecular markers for evaluating the fidelity of human pluripotent stem cell to RPE conversion. This study contributes to our understanding of the utility of hESC/hiPSC-derived RPE in AMD therapy.


Assuntos
Epitélio Pigmentado da Retina/metabolismo , Células-Tronco/metabolismo , Envelhecimento/genética , Western Blotting , Linhagem Celular , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Degeneração Macular/genética , Análise de Sequência com Séries de Oligonucleotídeos , Fagocitose , Epitélio Pigmentado da Retina/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/citologia
15.
Invest Ophthalmol Vis Sci ; 51(2): 1130-5, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19643958

RESUMO

PURPOSE: To investigate the function of MYO7A in human RPE cells and to test the validity of using shaker1 RPE in preclinical studies on therapies for Usher syndrome 1B by comparing human and mouse cells. METHODS: MYO7A was localized by immunofluorescence. Primary cultures of human and mouse RPE cells were used to measure melanosome motility and rod outer segment (ROS) phagocytosis and digestion. MYO7A was knocked down in the human RPE cells by RNAi to test for a mutant phenotype in melanosome motility. RESULTS: The distribution of MYO7A in the RPE of human and mouse was found to be comparable, both in vivo and in primary cultures. Primary cultures of human RPE cells phagocytosed and digested ROSs with kinetics comparable to that of primary cultures of mouse RPE cells. Melanosome motility was also comparable, and, after RNAi knockdown, consisted of longer-range fast movements characteristic of melanosomes in shaker1 RPE. CONCLUSIONS: The localization and function of MYO7A in human RPE cells is comparable to that in mouse RPE cells. Although shaker1 retinas do not undergo degeneration, correction of mutant phenotypes in the shaker1 RPE represents a valid preclinical test for potential therapeutic treatments.


Assuntos
Modelos Animais de Doenças , Miosinas/fisiologia , Epitélio Pigmentado da Retina/metabolismo , Síndromes de Usher/metabolismo , Idoso de 80 Anos ou mais , Animais , Western Blotting , Células Cultivadas , Técnica Indireta de Fluorescência para Anticorpo , Inativação Gênica , Humanos , Masculino , Melanossomas/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Microscopia Confocal , Miosina VIIa , Fagocitose/fisiologia , Interferência de RNA , Segmento Externo da Célula Bastonete/fisiologia , Transfecção
16.
Aging Cell ; 8(6): 765-8, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19793063

RESUMO

Reduced dietary intake increases lifespan in a wide variety of organisms. It also retards disease progression. We tested whether dietary supplementation of citric acid cycle metabolites could mimic this lifespan effect. We report that oxaloacetate supplementation increased lifespan in Caenorhabditis elegans. The increase was dependent on the transcription factor, FOXO/DAF-16, and the energy sensor, AMP-activated protein kinase, indicating involvement of a pathway that is also required for lifespan extension through dietary restriction. These results demonstrate that supplementation of the citric acid cycle metabolite, oxaloacetate, influences a longevity pathway, and suggest a tractable means of introducing the health-related benefits of dietary restriction.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Longevidade , Oxaloacetatos/metabolismo , Transdução de Sinais , Proteínas Quinases Ativadas por AMP/genética , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Fatores de Transcrição Forkhead/genética
17.
J Biol Chem ; 284(16): 10877-89, 2009 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-19240024

RESUMO

Melanoregulin (MREG), the product of the Mreg(dsu) gene, is a small highly charged protein, hypothesized to play a role in organelle biogenesis due to its effect on pigmentation in dilute, ashen, and leaden mutant mice. Here we provide evidence that MREG is required in lysosome-dependent phagosome degradation. In the Mreg(-/-) mouse, we show that loss of MREG function results in phagosome accumulation due to delayed degradation of engulfed material. Over time, the Mreg(-/-) mouse retinal pigment epithelial cells accumulate the lipofuscin component, A2E. MREG-deficient human and mouse retinal pigment epithelial cells exhibit diminished activity of the lysosomal hydrolase, cathepsin D, due to defective processing. Moreover, MREG localizes to small intracellular vesicles and associates with the endosomal phosphoinositide, phosphatidylinositol 3,5-biphosphate. Collectively, these studies suggest that MREG is required for lysosome maturation and support a role for MREG in intracellular trafficking.


Assuntos
Proteínas de Transporte/metabolismo , Células Epiteliais/metabolismo , Lisossomos/metabolismo , Epitélio Pigmentado Ocular/citologia , Proteínas Adaptadoras de Transporte Vesicular , Animais , Proteínas de Transporte/genética , Catepsina D/metabolismo , Linhagem Celular , Células Epiteliais/citologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Lipofuscina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose/fisiologia , Fosfatidiletanolaminas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Compostos de Piridínio/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Retinaldeído/análogos & derivados , Retinaldeído/metabolismo , Retinoides/metabolismo
18.
Bioorg Chem ; 36(6): 261-4, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18667222

RESUMO

The reported presence of a coenzyme B12-dependent methylmalonyl-CoA mutase in potatoes has been reexamined. The enzyme converting methylmalonyl-CoA was purified to electrophoretic homogeneity. Examination of the reaction product by 1H, 31P NMR and mass spectrometry revealed that it was methylmalonyl-3'-dephospho-CoA. The phosphatase enzyme needs neither coenzyme B12 nor S-adenosylmethionine as a cofactor.


Assuntos
Metilmalonil-CoA Mutase/química , Metilmalonil-CoA Mutase/isolamento & purificação , Monoéster Fosfórico Hidrolases/química , Solanum tuberosum/enzimologia , Cobamidas/química , Espectroscopia de Ressonância Magnética , Espectrometria de Massas/métodos , Modelos Químicos , Espectrofotometria Ultravioleta/métodos , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...