Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cells ; 10(8)2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34440732

RESUMO

The zebrafish provided an excellent platform to study the genetic and molecular approach of cellular phenotype-based cardiac research. We designed a novel protocol to develop the transparent transgenic zebrafish model to study annexin-5 activity in the cardiovascular function by generating homozygous transparent skin Casper(roy-/-,nacre-/-); myl7:RFP; annexin-5:YFP transgenic zebrafish. The skin pigmentation background of any vertebrate model organism is a major obstruction for in vivo confocal imaging to study the transgenic cellular phenotype-based study. By developing Casper(roy-/-,nacre-/-); myl7; annexin-5 transparent transgenic zebrafish strain, we established time-lapse in vivo confocal microscopy to study cellular phenotype/pathologies of cardiomyocytes over time to quantify changes in cardiomyocyte morphology and function over time, comparing control and cardiac injury and cardio-oncology. Casper contributes to the study by integrating a transparent characteristic in adult zebrafish that allows for simpler transparent visualization and observation. The Casper(roy-/-,nacre-/-) transgenic progenies developed through cross-breeding with the transgenic strain of Tg(UAS:SEC-Hsa.ANXA5-YFP,myl7:RFP). Confocal and fluorescent microscopy were being used to obtain accurate, precise imaging and to determine fluorescent protein being activated. This study protocol was conducted under two sections; 1.1: Generation of homozygous Tg(UAS:SEC-Hsa.ANXA5-YFP,myl7:RFP); Casper(roy-/-,nacre-/-) zebrafish (generation F01-F06) and 1.2: Screening and sorting the transparent transgenic progeny and in vivo imaging to validate cardiac morphology through in vivo confocal imaging. We coined the newly developed strain as Tg(UAS:SEC-Hsa.ANXA5-YFP,myl7:RFP); Casper(roy-/-,nacre-/-)gmc1. Thus, the newly developed strain maintains transparency of the skin throughout the entire life of zebrafish and is capable of application of a non-invasive in vivo imaging process. These novel results provide an in vivo whole organism-based platform to design high-throughput screening and establish a new horizon for drug discovery in cardiac cell death and cardio-oncology therapeutics and treatment.


Assuntos
Animais Geneticamente Modificados/genética , Miócitos Cardíacos/metabolismo , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Anexina A5/genética , Anexina A5/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Fator de Transcrição Associado à Microftalmia/deficiência , Fator de Transcrição Associado à Microftalmia/genética , Microscopia Confocal , Modelos Animais , Pigmentação da Pele , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/metabolismo
2.
Sci Rep ; 6: 21208, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26876013

RESUMO

Regeneration of the visual pigment by cells of the retinal pigment epithelium (RPE) is fundamental to vision. Here we show that the microphthalmia-associated transcription factor, MITF, which plays a central role in the development and function of RPE cells, regulates the expression of two visual cycle genes, Rlbp1 which encodes retinaldehyde binding protein-1 (RLBP1), and Rdh5, which encodes retinol dehydrogenase-5 (RDH5). First, we found that Rlbp1 and Rdh5 are downregulated in optic cups and presumptive RPEs of Mitf-deficient mouse embryos. Second, experimental manipulation of MITF levels in human RPE cells in culture leads to corresponding modulations of the endogenous levels of RLBP1 and RDH5. Third, the retinal degeneration associated with the disruption of the visual cycle in Mitf-deficient mice can be partially corrected both structurally and functionally by an exogenous supply of 9-cis-retinal. We conclude that the expression of Rlbp1 and Rdh5 critically depends on functional Mitf in the RPE and suggest that MITF has an important role in controlling retinoid processing in the RPE.


Assuntos
Oxirredutases do Álcool/biossíntese , Proteínas de Transporte/biossíntese , Fator de Transcrição Associado à Microftalmia/genética , Degeneração Retiniana/genética , Visão Ocular/genética , Oxirredutases do Álcool/genética , Animais , Proteínas de Transporte/genética , Regulação da Expressão Gênica , Humanos , Camundongos , Fator de Transcrição Associado à Microftalmia/deficiência , Degeneração Retiniana/patologia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Visão Ocular/fisiologia
3.
Cell Cycle ; 14(8): 1232-41, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25832911

RESUMO

The circadian clock controls the timing of the cell cycle in healthy tissues and clock disruption is known to increase tumourigenesis. Melanoma is one of the most rapidly increasing forms of cancer and the precise molecular circadian changes that occur in a melanoma tumor are unknown. Using a melanoma zebrafish model, we have explored the molecular changes that occur to the circadian clock within tumors. We have found disruptions in melanoma clock gene expression due to a major impairment to the light input pathway, with a parallel loss of light-dependent activation of DNA repair genes. Furthermore, the timing of mitosis in tumors is perturbed, as well as the regulation of certain key cell cycle regulators, such that cells divide arhythmically. The inability to co-ordinate DNA damage repair and cell division is likely to promote further tumourigenesis and accelerate melanoma development.


Assuntos
Relógios Circadianos/fisiologia , Luz , Melanoma/patologia , Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados/metabolismo , Relógios Circadianos/genética , Criptocromos/genética , Criptocromos/metabolismo , Ciclina B1/genética , Ciclina B1/metabolismo , Dano ao DNA , Reparo do DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Melanoma/metabolismo , Fator de Transcrição Associado à Microftalmia/deficiência , Fator de Transcrição Associado à Microftalmia/genética , Fator de Transcrição Associado à Microftalmia/metabolismo , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Fase S , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
5.
Mol Biosyst ; 8(11): 2924-31, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22898827

RESUMO

Microphthalmia-associated transcription factor (MITF) is a master regulator in melanocyte proliferation, development, survival and melanoma formation. In melanocyte dysfunction disease, it is observed that the expressions of MITF, tyrosinase (TYR), tyrosinase related protein 1 (TYRP1) and tyrosinase related protein 2 (TYRP2)/dopachrome tautomerase (DCT) are changed, the consequence of which remains unclear. In this study, we focused on the change of microRNA (miRNA) profiles and Tyrosinase Related Proteins (TRPs) in MITF knocked down melanocytes. For the first time, we assayed the MITF-KD miRNA profiles using a miRNA microarray and found that hsa-miR-1225-3p, hsa-miR-634, hsa-miR-197, hsa-miR-766, hsa-miR-574-5p and hsa-miR-328 were upregulated, and hsa-miR-720 and hsa-miR-1308 were downregulated in MITF knocked down melanocytes. These miRNAs were validated by miRNA real time qPCR. These miRNA potential targets, especially the TRPs, were analyzed according to the miRNA database (Sanger Center). By TargetScan prediction, the hsa-miR-634 and hsa-miR-328 have poorly conserved sites on TYR and hsa-miR-197 have poorly conserved sites on TYR1. Through qPCR and western blotting we found that the expression of TYR and TYRP1 were dramatically decreased and the expression of TYRP2 was increased in MITF knocked down melanocytes (MITF-KD). These results suggested that the miRNAs may be involved in MITF regulation of TYR, TYRP1 and TYRP2, which provides a new clue for understanding the role of miRNAs in melanocyte dysfunctional disease.


Assuntos
Melanócitos/metabolismo , MicroRNAs/genética , Fator de Transcrição Associado à Microftalmia/deficiência , Fator de Transcrição Associado à Microftalmia/genética , Western Blotting , Células Cultivadas , Humanos , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/metabolismo , Monofenol Mono-Oxigenase/genética , Monofenol Mono-Oxigenase/metabolismo , Oxirredutases/genética , Oxirredutases/metabolismo , Reação em Cadeia da Polimerase , Interferência de RNA/fisiologia
6.
Nat Neurosci ; 14(9): 1150-9, 2011 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-21857656

RESUMO

In a neural integrator, the variability and topographical organization of neuronal firing-rate persistence can provide information about the circuit's functional architecture. We used optical recording to measure the time constant of decay of persistent firing (persistence time) across a population of neurons comprising the larval zebrafish oculomotor velocity-to-position neural integrator. We found extensive persistence time variation (tenfold; coefficients of variation = 0.58-1.20) across cells in individual larvae. We also found that the similarity in firing between two neurons decreased as the distance between them increased and that a gradient in persistence time was mapped along the rostrocaudal and dorsoventral axes. This topography is consistent with the emergence of persistence time heterogeneity from a circuit architecture in which nearby neurons are more strongly interconnected than distant ones. Integrator circuit models characterized by multiple dimensions of slow firing-rate dynamics can account for our results.


Assuntos
Movimentos Oculares/fisiologia , Rede Nervosa/fisiologia , Neurônios/fisiologia , Dinâmica não Linear , Potenciais de Ação , Animais , Animais Geneticamente Modificados , Tronco Encefálico/citologia , Cálcio/metabolismo , Simulação por Computador , Movimentos Oculares/genética , Lateralidade Funcional , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Larva , Luz , Fator de Transcrição Associado à Microftalmia/deficiência , Modelos Neurológicos , Estimulação Luminosa/métodos , Fatores de Tempo , Peixe-Zebra , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética
7.
Cancer Res ; 70(9): 3813-22, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20388797

RESUMO

Apoptosis and senescence are cellular failsafe programs that counteract excessive mitogenic signaling observed in cancer cells. Melanoma is known for its notorious resistance to apoptotic processes; therefore, senescence, which remains poorly understood in melanomas, can be viewed as a therapeutic alternative. Microphthalmia-associated transcription factor (MITF), in which its M transcript is specifically expressed in melanocyte cells, plays a critical role in melanoma proliferation, and its specific inhibition is associated with G(0)-G(1) growth arrest. Interestingly, decreased MITF expression has been described in senescent melanocytes, and we have observed an inhibition of MITF expression in melanoma cells exposed to chemotherapeutic drugs that induce their senescence. All these observations thereby question the role of MITF in controlling senescence in melanoma cells. Here, we report that long-term depletion of MITF in melanoma cells triggers a senescence program characterized by typical morphologic and biochemical changes associated with a sustained growth arrest. Further, we show that MITF-silenced cells engage a DNA damage response (DDR) signaling pathway, leading to p53 upregulation, which is critically required for senescence entry. This study uncovers the existence of a lineage-restricted DDR/p53 signaling pathway that is inhibited by MITF to prevent senescence and favor melanoma cell proliferation.


Assuntos
Dano ao DNA , Melanoma/genética , Fator de Transcrição Associado à Microftalmia/deficiência , Animais , Linhagem Celular Tumoral , Linhagem da Célula/fisiologia , Senescência Celular/fisiologia , Humanos , Melanoma/metabolismo , Melanoma/patologia , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Fator de Transcrição Associado à Microftalmia/genética , Fator de Transcrição Associado à Microftalmia/metabolismo , Mitose/genética , Transdução de Sinais , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
8.
J Biol Chem ; 283(18): 12635-42, 2008 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-18281284

RESUMO

Melanosomes are lysosome-related organelles specialized in melanin synthesis and transport. In this study, we show that microphthalmia-associated transcription factor (MITF) silencing induces melanosome gathering around the nucleus and causes the relocalization of Rab27A, Slac2a-Mlph, and Myo5a that control the transport of melanosomes on the actin network. In an attempt to elucidate the mechanism by which MITF controls melanosome distribution, we identify RAB27A as a new MITF target gene. Indeed, MITF silencing leads to a dramatic decrease in Rab27A expression and blocks the stimulation of Rab27A expression evoked by cAMP. Further, forced expression of MITF increases Rab27A expression, indicating that MITF is required and sufficient for Rab27A expression in melanoma cells. MITF binds to two E-boxes in the proximal region of the Rab27A promoter and stimulates its transcriptional activity. Finally, re-expression of Rab27A, in MITF-depleted cells, restores the transport of melanosomes to the cell periphery. These results show that RAB27A is a new direct transcriptional target of MITF and link MITF to melanosome transport, another key parameter of melanocyte differentiation and skin pigmentation. Interestingly, Rab27A is involved in other fundamental physiological functions, such as the transport of lytic granules and insulin secretion. Thus our results, deciphering the mechanism of Rab27A transcriptional regulation, have an interest that goes beyond the skin pigmentation field.


Assuntos
Regulação Neoplásica da Expressão Gênica , Melanossomas/metabolismo , Fator de Transcrição Associado à Microftalmia/metabolismo , Proteínas rab de Ligação ao GTP/genética , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Sequência de Bases , Transporte Biológico , Linhagem Celular Tumoral , Elementos E-Box/genética , Inativação Gênica , Humanos , Camundongos , Fator de Transcrição Associado à Microftalmia/deficiência , Dados de Sequência Molecular , Miosina Tipo V/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , Transcrição Gênica , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab27 de Ligação ao GTP
9.
Nat Neurosci ; 11(9): 1083-90, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19160507

RESUMO

Sensory circuits frequently integrate converging inputs while maintaining precise functional relationships between them. For example, in mammals with stereopsis, neurons at the first stages of binocular visual processing show a close alignment of receptive-field properties for each eye. Still, basic questions about the global wiring mechanisms that enable this functional alignment remain unanswered, including whether the addition of a second retinal input to an otherwise monocular neural circuit is sufficient for the emergence of these binocular properties. We addressed this question by inducing a de novo binocular retinal projection to the larval zebrafish optic tectum and examining recipient neuronal populations using in vivo two-photon calcium imaging. Notably, neurons in rewired tecta were predominantly binocular and showed matching direction selectivity for each eye. We found that a model based on local inhibitory circuitry that computes direction selectivity using the topographic structure of both retinal inputs can account for the emergence of this binocular feature.


Assuntos
Lateralidade Funcional/fisiologia , Células Receptoras Sensoriais/fisiologia , Colículos Superiores/fisiologia , Visão Binocular/fisiologia , Campos Visuais/fisiologia , Aminoácidos , Animais , Animais Geneticamente Modificados , Cálcio/metabolismo , Embrião não Mamífero , Lateralidade Funcional/genética , Técnicas In Vitro , Larva , Fator de Transcrição Associado à Microftalmia/deficiência , Estimulação Luminosa/métodos , Privação Sensorial , Células Receptoras Sensoriais/classificação , Fatores de Tempo , Visão Binocular/genética , Campos Visuais/genética , Vias Visuais/fisiologia , Peixe-Zebra , Proteínas de Peixe-Zebra/deficiência
10.
J Biol Chem ; 282(21): 15921-9, 2007 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-17403683

RESUMO

Transcription factors NFATc1, PU.1, and MITF collaborate to regulate specific genes in response to colony-stimulating factor-1 (CSF-1) and receptor activator of NF-kappaB ligand (RANKL) signaling during osteoclast differentiation. However, molecular details concerning timing and mechanism of specific events remain ill-defined. In bone marrow-derived precursors, CSF-1 alone promoted assembly of MITF-PU.1 complexes at osteoclast target gene promoters like cathepsin K and acid 5 phosphatase without increasing gene expression. The combination of RANKL and CSF-1 concurrently increased the levels of MAPK-phosphorylated forms of MITF, p38 MAPK, and SWI/SNF chromatin-remodeling complexes bound to these target promoters and markedly increased expression of the genes. NFATc1 was subsequently recruited to complexes at the promoters during terminal stages of osteoclast differentiation. Genetic analysis of Mitf and Pu.1 in mouse models supported the critical interaction of these genes in osteoclast differentiation. The results define MITF and PU.1 as nuclear effectors that integrate CSF-1/RANKL signals during osteoclast differentiation to initiate expression of target genes, whereas a complex that includes NFATc1 may act to maintain target gene expression in differentiated cells.


Assuntos
Diferenciação Celular/fisiologia , Fator de Transcrição Associado à Microftalmia/metabolismo , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/fisiologia , Transativadores/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Fosfatase Ácida/biossíntese , Animais , Catepsina K , Catepsinas/biossíntese , Células Cultivadas , Montagem e Desmontagem da Cromatina/fisiologia , Proteínas Cromossômicas não Histona/metabolismo , Isoenzimas/biossíntese , Fator Estimulador de Colônias de Macrófagos/metabolismo , Camundongos , Camundongos Mutantes , Fator de Transcrição Associado à Microftalmia/deficiência , Osteoclastos/citologia , Regiões Promotoras Genéticas/fisiologia , Ligante RANK/metabolismo , Fosfatase Ácida Resistente a Tartarato , Fatores de Transcrição/metabolismo
11.
Int Immunol ; 17(11): 1495-503, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16186160

RESUMO

The microphthalmic (mi) mouse exhibits deficiencies in the development of osteoclasts, melanocytes, mast cells and marrow B cells. Previously, we demonstrated that the marrow of such mice over-express receptor activator of nuclear factor kappaB (RANK) ligand (RANKL). RANKL has been shown to induce the production of IFN-beta, a type I IFN. Additionally, maturing B cells have been shown to undergo apoptosis in response to type I IFNs including IFN-beta during differentiation. We hypothesized that the loss of B cells in the marrow of mi mice was due to the over-expression of IFN-beta as a result of heightened RANK-RANKL signaling. Creating a mouse with the mi genotype that was non-responsive to IFN-beta (lacking the type I IFNR) allowed us to test this hypothesis. These mice demonstrated an elevated number of marrow B cells and marrow precursor cells compared with mi animals possessing the type I IFNR. Intriguingly, type I IFNR-deficient wild-type animals also demonstrated an increased number of precursor cells in the marrow, but not an expansion of B220-positive pre-B cells, compared with wild type, suggesting that modulation of type I IFN responses directly controls the development of marrow constituents.


Assuntos
Linfócitos B/imunologia , Células da Medula Óssea/imunologia , Interferon beta/imunologia , Fator de Transcrição Associado à Microftalmia/imunologia , Microftalmia/imunologia , Osteopetrose/imunologia , Animais , Linfócitos B/patologia , Células da Medula Óssea/patologia , Proteínas de Transporte/imunologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Células Cultivadas , Interferon beta/deficiência , Antígenos Comuns de Leucócito/imunologia , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Fator de Transcrição Associado à Microftalmia/deficiência , Microftalmia/genética , Microftalmia/patologia , Osteopetrose/genética , Osteopetrose/patologia , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Transdução de Sinais/genética , Transdução de Sinais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA