Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Am J Physiol Heart Circ Physiol ; 324(4): H504-H518, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36800508

RESUMO

Upon myocardial infarction (MI), ischemia-induced cell death triggers an inflammatory response responsible for removing necrotic material and inducing tissue repair. TRPM4 is a Ca2+-activated ion channel permeable to monovalent cations. Although its role in cardiomyocyte-driven hypertrophy and arrhythmia post-MI has been established, no study has yet investigated its role in the inflammatory process orchestrated by endothelial cells, immune cells, and fibroblasts. This study aims to assess the role of TRPM4 in 1) survival and cardiac function, 2) inflammation, and 3) healing post-MI. We performed ligation of the left coronary artery or sham intervention on 154 Trpm4 WT or KO mice under isoflurane anesthesia. Survival and echocardiographic functions were monitored up to 5 wk. We collected serum during the acute post-MI phase to analyze proteomes and performed single-cell RNA sequencing on nonmyocytic cells of hearts after 24 and 72 h. Lastly, we assessed chronic fibrosis and angiogenesis. We observed no significant differences in survival or cardiac function, even though our proteomics data showed significantly decreased tissue injury markers (i.e., creatine kinase M and VE-cadherin) in KO serum after 12 h. On the other hand, inflammation, characterized by serum amyloid P component in the serum, higher number of recruited granulocytes, inflammatory monocytes, and macrophages, as well as expression of proinflammatory genes, was significantly higher in KO. This correlated with increased chronic cardiac fibrosis and angiogenesis. Since inflammation and fibrosis are closely linked to adverse remodeling, future therapeutic attempts at inhibiting TRPM4 will need to assess these parameters carefully before proceeding with translational studies.NEW & NOTEWORTHY Deletion of Trpm4 increases markers of cardiac and systemic inflammation within the first 24 h after MI, while inducing an earlier fibrotic transition at 72 h and more overall chronic fibrosis and angiogenesis at 5 wk. The descriptive, robust, and methodologically broad approach of this study sheds light on an important caveat that will need to be taken into account in all future therapeutic attempts to inhibit TRPM4 post-MI.


Assuntos
Infarto do Miocárdio , Canais de Cátion TRPM , Camundongos , Animais , Células Endoteliais/metabolismo , Multiômica , Miócitos Cardíacos/metabolismo , Inflamação/metabolismo , Fibrose , Camundongos Endogâmicos C57BL , Camundongos Knockout , Remodelação Ventricular , Miocárdio/metabolismo , Modelos Animais de Doenças , Canais de Cátion TRPM/genética
2.
Proc Natl Acad Sci U S A ; 111(11): 4239-44, 2014 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-24591640

RESUMO

Phospholipase Cε (PLCε) has been characterized as a direct effector of Ras in vitro and in cellular systems; however, the role of PLCε in tumorigenesis and its link to Ras in this context remain unclear. To assess the role of PLCε in Ras-driven cancers, we generated two new mouse strains: one carrying a targeted deletion of Plce (Plce(-/-)) and the other carrying mutant alleles of Plce unable to bind to Ras (Plce(RAm/RAm)). The Plce(-/-) and, to a lesser degree, Plce(RAm/RAm) transgenic mice exhibited increased susceptibility to tumor formation in the two-stage skin carcinogenesis protocol, revealing a tumor suppressor function for this PLC. This result also suggests that in this context Ras binding in part regulates functions of PLCε. Although significant differences were not seen in the LSL-Kras(G12D) nonsmall cell lung carcinoma model, down-regulation of PLCε was found in animal tumors and in cellular systems following expression of the oncogenic Ras. An inhibitory impact of PLCε on cell growth requires intact lipase activity and is likely mediated by protein kinase C enzymes. Further cellular studies suggest involvement of histone deacetylase in the mechanism of PLCε down-regulation. Taken together, our results show a previously unidentified tumor suppressor role for this PLC in animal models and, together with observations of marked down-regulation in colorectal, lung, and skin tumors, suggest its use as a biological marker in cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica/fisiologia , Genes Supressores de Tumor/fisiologia , Genes ras/genética , Neoplasias/genética , Fosfoinositídeo Fosfolipase C/fisiologia , Animais , Proliferação de Células , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Transgênicos , Fosfoinositídeo Fosfolipase C/genética , Proteína Quinase C/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
3.
PLoS One ; 19(3): e0298820, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38452156

RESUMO

BACKGROUND: 14-3-3 proteins are ubiquitous proteins that play a role in cardiac physiology (e.g., metabolism, development, and cell cycle). Furthermore, 14-3-3 proteins were proposed to regulate the electrical function of the heart by interacting with several cardiac ion channels, including the voltage-gated sodium channel Nav1.5. Given the many cardiac arrhythmias associated with Nav1.5 dysfunction, understanding its regulation by the protein partners is crucial. AIMS: In this study, we aimed to investigate the role of 14-3-3 proteins in the regulation of the human cardiac sodium channel Nav1.5. METHODS AND RESULTS: Amongst the seven 14-3-3 isoforms, only 14-3-3η (encoded by YWHAH gene) weakly co-immunoprecipitated with Nav1.5 when heterologously co-expressed in tsA201 cells. Total and cell surface expression of Nav1.5 was however not modified by 14-3-3η overexpression or inhibition with difopein, and 14-3-3η did not affect physical interaction between Nav1.5 α-α subunits. The current-voltage relationship and the amplitude of Nav1.5-mediated sodium peak current density were also not changed. CONCLUSIONS: Our findings illustrate that the direct implication of 14-3-3 proteins in regulating Nav1.5 is not evident in a transformed human kidney cell line tsA201.


Assuntos
Proteínas 14-3-3 , Canais de Sódio Disparados por Voltagem , Humanos , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Canais de Sódio Disparados por Voltagem/metabolismo , Miócitos Cardíacos/metabolismo , Linhagem Celular , Arritmias Cardíacas , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo
4.
Physiol Rep ; 11(16): e15783, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37604672

RESUMO

TRPM4 is a calcium-activated, voltage-modulated, nonselective ion channel widely expressed in various cells and tissues. TRPM4 regulates the influx of sodium ions, thus playing a role in regulating the membrane potential. In the heart, TRPM4 is expressed in both cardiomyocytes and cells of the conductive pathways. Clinical studies have linked TRPM4 mutations to several cardiac disorders. While data from experimental studies have demonstrated TRPM4's functional significance in cardiac physiology, its exact roles in the heart have remained unclear. In this study, we investigated the role of TRPM4 in cardiac physiology in a newly generated Trpm4 knockdown mouse model. Male and female Trpm4 knockdown (Trpm4-/- ) and wild-type mice of different ages (5- to 12- week-old (young) and 24-week-old or more (adult)) were characterized using a multimodal approach, encompassing surface electrocardiograms (ECG), echocardiography recordings, ex vivo ECGs in isolated heart, endocardial mappings, Western blots, and mRNA quantifications. The assessment of cardiac electrophysiology by surface ECGs revealed no significant differences between wild-type and Trpm4-/- young (5- to 12-week-old) mice of either sex. Above 24 weeks of age, adult male Trpm4-/- mice showed reduced heart rate and increased heart rate variability. Echocardiography revealed that only adult male Trpm4-/- mice exhibited slight left ventricular hypertrophic alterations compared to controls, illustrated by alterations of the mitral valve pressure halftime, the mitral valve E/A ratio, the isovolumetric relaxation time, and the mitral valve deceleration. In addition, an assessment of the right ventricular systolic function by scanning the pulmonary valve highlighted an alteration in pulmonary valve peak velocity and pressure in adult male Trpm4-/- mice. Endocardial mapping recordings showed that applying 5 µM of the new TRPM4 inhibitor NBA triggered a third-degree atrioventricular block on 40% of wild-type hearts. These results confirm the key role of TRPM4 in the proper structure and electrical function of the heart. It also reveals differences between male and female animals that have never been reported. In addition, the investigation of the effects of NBA on heart function confirms the role of TRPM4 in atrioventricular conduction.


Assuntos
Técnicas Eletrofisiológicas Cardíacas , Canais de Cátion TRPM , Animais , Feminino , Masculino , Camundongos , Eletrofisiologia Cardíaca , Eletrocardiografia , Hemodinâmica , Miócitos Cardíacos , Canais de Cátion TRPM/genética , Técnicas de Silenciamento de Genes
5.
Sci Rep ; 9(1): 19422, 2019 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-31857666

RESUMO

The Cre/lox system is a potent technology to control gene expression in mouse tissues. However, cardiac-specific Cre recombinase expression alone can lead to cardiac alterations when no loxP sites are present, which is not well understood. Many loxP-like sites have been identified in the mouse genome that might be Cre sensitive. One of them is located in the Dmd gene encoding dystrophin, a protein important for the function and stabilization of voltage-gated calcium (Cav1.2) and sodium (Nav1.5) channels, respectively. Here, we investigate whether Cre affects dystrophin expression and function in hearts without loxP sites in the genome. In mice expressing Cre under the alpha-myosin heavy chain (MHC-Cre) or Troponin T (TNT-Cre) promoter, we investigated dystrophin expression, Nav1.5 expression, and Cav1.2 function. Compared to age-matched MHC-Cre- mice, dystrophin protein level was significantly decreased in hearts from MHC-Cre+ mice of more than 12-weeks-old. Quantitative RT-PCR revealed decreased mRNA levels of Dmd gene. Unexpectedly, calcium current (ICaL), but not Nav1.5 protein expression was altered in those mice. Surprisingly, in hearts from 12-week-old and older TNT-Cre+ mice, neither ICaL nor dystrophin and Nav1.5 protein content were altered compared to TNT-Cre-. Cre recombinase unpredictably affects cardiac phenotype, and Cre-expressing mouse models should be carefully investigated before experimental use.


Assuntos
Cálcio/metabolismo , Distrofina/metabolismo , Integrases/metabolismo , Miócitos Cardíacos/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Regiões Promotoras Genéticas , Troponina T/genética , Envelhecimento/metabolismo , Animais , Distrofina/genética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
6.
Front Physiol ; 10: 834, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31333492

RESUMO

Background: In cardiac ventricular muscle cells, the presence of voltage-gated sodium channels Nav1.5 at the lateral membrane depends in part on the interaction between the dystrophin-syntrophin complex and the Nav1.5 C-terminal PDZ-domain-binding sequence Ser-Ile-Val (SIV motif). α1-Syntrophin, a PDZ-domain adaptor protein, mediates the interaction between Nav1.5 and dystrophin at the lateral membrane of cardiac cells. Using the cell-attached patch-clamp approach on cardiomyocytes expressing Nav1.5 in which the SIV motif is deleted (ΔSIV), sodium current (INa) recordings from the lateral membrane revealed a SIV-motif-independent INa. Since immunostaining has suggested that Nav1.5 is expressed in transverse (T-) tubules, this remaining INa might be carried by channels in the T-tubules. Of note, a recent study using heterologous expression systems showed that α1-syntrophin also interacts with the Nav1.5 N-terminus, which may explain the SIV-motif independent INa at the lateral membrane of cardiomyocytes. Aim: To address the role of α1-syntrophin in regulating the INa at the lateral membrane of cardiac cells. Methods and Results: Patch-clamp experiments in cell-attached configuration were performed on the lateral membranes of wild-type, α1-syntrophin knockdown, and ΔSIV ventricular mouse cardiomyocytes. Compared to wild-type, a reduction of the lateral INa was observed in myocytes from α1-syntrophin knockdown hearts. Similar to ΔSIV myocytes, a remaining INa was still recorded. In addition, cell-attached INa recordings from lateral membrane did not differ significantly between non-detubulated and detubulated ΔSIV cardiomyocytes. Lastly, we obtained evidence suggesting that cell-attached patch-clamp experiments on the lateral membrane cannot record currents carried by channels in T-tubules such as calcium channels. Conclusion: Altogether, these results suggest the presence of a sub-pool of sodium channels at the lateral membrane of cardiomyocytes that is independent of α1-syntrophin and the PDZ-binding motif of Nav1.5, located in membrane domains outside of T-tubules. The question of a T-tubular pool of Nav1.5 channels, however, remains open.

7.
Methods Mol Biol ; 337: 185-205, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16929948

RESUMO

Analysis of genetically engineered mice is crucial for our understanding of the in vivo function of genes and proteins in the whole organism. This includes inactivation of a gene or the generation of specific mutations. The development of knockout and transgenic technologies in the mouse, therefore, represents a powerful tool for elucidating gene function, for modeling of human diseases, and potentially for the evaluation of drugs. In particular, conditional gene targeting applying the Cre/loxP-mediated recombination system is increasingly used to evaluate the role of the gene of interest in a cell-type-specific or even inducible manner. The experimental steps start with the characterization of the gene locus, followed by construction of a vector, gene targeting in ES cells, and establishment of mouse lines carrying the desired mutation. These are then bred to transgenic mice expressing Cre recombinase in a tissue-specific manner, thus allowing gene inactivation in a cell type of interest.


Assuntos
Deleção de Genes , Especificidade de Órgãos , Transgenes/genética , Animais , Quimera/genética , Embrião de Mamíferos/citologia , Feminino , Vetores Genéticos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células-Tronco/metabolismo , Transfecção
8.
Elife ; 5: e12994, 2016 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-26765561

RESUMO

Rho-associated kinases 1 and 2 (ROCK1/2) are Rho-GTPase effectors that control key aspects of the actin cytoskeleton, but their role in proliferation and cancer initiation or progression is not known. Here, we provide evidence that ROCK1 and ROCK2 act redundantly to maintain actomyosin contractility and cell proliferation and that their loss leads to cell-cycle arrest and cellular senescence. This phenotype arises from down-regulation of the essential cell-cycle proteins CyclinA, CKS1 and CDK1. Accordingly, while the loss of either Rock1 or Rock2 had no negative impact on tumorigenesis in mouse models of non-small cell lung cancer and melanoma, loss of both blocked tumor formation, as no tumors arise in which both Rock1 and Rock2 have been genetically deleted. Our results reveal an indispensable role for ROCK, yet redundant role for isoforms 1 and 2, in cell cycle progression and tumorigenesis, possibly through the maintenance of cellular contractility.


Assuntos
Carcinogênese , Proliferação de Células , Quinases Associadas a rho/metabolismo , Animais , Carcinoma Pulmonar de Células não Pequenas/patologia , Técnicas de Inativação de Genes , Melanoma/patologia , Camundongos , Quinases Associadas a rho/genética
9.
Curr Biol ; 22(21): 2063-8, 2012 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-23063435

RESUMO

RAL small GTPases, encoded by the Rala and Ralb genes, are members of the RAS superfamily of small GTPases and can act as downstream effectors of RAS [1]. Although highly similar, distinct functions have been identified for RALA and RALB: RALA has been implicated in epithelial cell polarity [2], insulin secretion [3], GLUT4 translocation [4, 5], neurite branching, and neuronal polarity [6, 7], and RALB in tumor cell survival [8], migration/invasion [9-12], TBK1 activation [13], and autophagy [14]. To investigate RAL GTPases in vivo, we generated null and conditional knockout mice. Ralb null mice are viable with no overt phenotype; the Rala null leads to exencephaly and embryonic lethality. The exencephaly phenotype is exacerbated in Rala(-/-);Ralb(+/-) embryos; embryos null for Rala and Ralb do not live past gastrulation. Using a Kras-driven non-small cell lung carcinoma mouse model, we found that either RALA or RALB is sufficient for tumor growth. However, deletion of both Ral genes blocks tumor formation. Either RALA or RALB is sufficient for cell proliferation, but cells lacking both fail to proliferate. These studies demonstrate functions of RAL proteins in development, tumorigenesis, and cell proliferation and show that RALA and RALB act in a redundant fashion.


Assuntos
Transformação Celular Neoplásica , Desenvolvimento Embrionário , Proteínas ral de Ligação ao GTP/fisiologia , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Polaridade Celular , Proliferação de Células , Células Cultivadas , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tubo Neural/embriologia , Tubo Neural/metabolismo , Defeitos do Tubo Neural/genética , Proteína Oncogênica p21(ras)/metabolismo , Transdução de Sinais , Proteínas ral de Ligação ao GTP/genética
10.
Gene Expr Patterns ; 11(5-6): 299-308, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21397048

RESUMO

In order to fully describe the expression pattern of the transcription factor FoxO1, we have screened the ES cell genetrap repository databases and obtained a clone that contains the ß-geo reporter gene inserted within intron 1 of FoxO1. We then used the ES cell clone to generate a new mouse strain (B6;129P2- Foxo1(Gt(AD0086)Wtsi/JJC)), which expresses ß-geo according to the endogenous FoxO1 pattern, and collected embryo stages from 7.0dpc to 18.5dpc. We show that the expression of FoxO1 is highly dynamic, starting in the neuroepithelium and then extending into the developing vasculature, including all early stages of heart formation. There is a dramatic switch of expression at 11.5dpc in which most vascular expression is abolished and replaced by skeletal muscle expression. In addition FoxO1 is also expressed in several epithelial structures including the olfactory and otic systems, the cornea and at different levels of the gut depending on developmental stage. At later foetal stages, FoxO1 is upregulated again in the same tissues were it is active during early development, including skeletal muscle, vascular system and neuroepithelium.


Assuntos
Embrião de Mamíferos/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Animais , Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias/metabolismo , Feminino , Proteína Forkhead Box O1 , Camundongos , RNA Mensageiro/metabolismo , Regulação para Cima
11.
Dev Biol ; 303(2): 838-47, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17196956

RESUMO

Pigment cells of mammals are characterized by two different developmental origins: cells of the retinal pigment epithelium (RPE) originate from the optic cup of the developing forebrain, whereas melanocytes arise from the neural crest. The pigmentation gene tyrosinase is expressed in all pigment cells but differentially regulated in melanocytes and RPE. The tyrosinase promoter does not confer strong expression in pigment cells in vivo, while inclusion of a distal regulatory element at position -15 kb is necessary and sufficient to provide strong expression in melanocytes. Nevertheless, the regulatory elements responsible for correct spatial and temporal tyrosinase expression in the RPE remained unidentified so far. In this report, we show that a 186 kb BAC containing the tyrosinase gene provides transgene expression in both RPE and melanocytes indicating the presence of regulatory sequences required for expression in the RPE. A deletion analysis of the BAC was performed demonstrating that a RPE-regulatory element resides between -17 and -75 kb. Using multi-species comparative genomic analysis we identified three conserved sequences within this region. When tested in transgenic mice one of these sequences located at -47 kb targeted expression to the RPE. In addition, deletion of this regulatory element within a tyrosinase::lacZ BAC provided evidence that this sequence is not only sufficient but also required for correct spatial and temporal expression in the RPE. The identification of this novel element demonstrates that tyrosinase gene expression is controlled by separate distal regulatory sequences in melanocytes and RPE.


Assuntos
Melanócitos/enzimologia , Monofenol Mono-Oxigenase/genética , Epitélio Pigmentado Ocular/enzimologia , Albinismo Oculocutâneo/enzimologia , Albinismo Oculocutâneo/genética , Animais , Sequência de Bases , Cromossomos Artificiais Bacterianos/genética , Primers do DNA/genética , Elementos Facilitadores Genéticos , Regulação Enzimológica da Expressão Gênica , Genes Reguladores , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Mutação , Fenótipo
12.
Pigment Cell Res ; 20(3): 173-84, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17516925

RESUMO

The terminal differentiation of melanocytes is associated with the transcriptional activation of genes responsible for pigment production such as tyrosinase. Pigment cell-specific transcription factors, such as Mitf, as well as specific proximal and distal regulatory elements (DRE) are implicated in the tight control of tyrosinase expression during development and adulthood. Proper tyrosinase expression in melanocytes depends upon the presence of a DRE that is located at -15 kb and provides enhancer activity via a central element termed core-enhancer. In this report, we show that the transcription factors Sox10, Mitf and USF-1 are able to activate the core-enhancer in luciferase reporter assays. Comparative sequence analysis identified evolutionarily motifs resembling Sox10 binding sites that were required for full enhancer activity in melanoma cells and in tyrosinase::lacZ transgenic mice. Sox10 was able to bind the DRE in vitro and mutation of the conserved motifs abolished the enhancer transactivation mediated by Sox10. In addition, two highly conserved CAGCTG E-box motifs were identified that were also required for enhancer activity and for transactivation by Mitf. The results suggest that Sox10 directly, and Mitf, most likely indirectly, activate the tyrosinase enhancer, underlining the contribution of Sox10 to tyrosinase gene regulation in melanocytes.


Assuntos
Elementos Facilitadores Genéticos , Regulação Enzimológica da Expressão Gênica , Proteínas de Grupo de Alta Mobilidade/fisiologia , Fator de Transcrição Associado à Microftalmia/fisiologia , Monofenol Mono-Oxigenase/metabolismo , Fatores de Transcrição/fisiologia , Animais , Sequência de Bases , Sítios de Ligação , Ativação Enzimática , Humanos , Camundongos , Dados de Sequência Molecular , Fatores de Transcrição SOXE , Homologia de Sequência do Ácido Nucleico , Pigmentação da Pele/genética , Ativação Transcricional
13.
Dev Biol ; 298(2): 644-55, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16934245

RESUMO

Pigment cells of mammals originate from two different lineages: melanocytes arise from the neural crest, whereas cells of the retinal pigment epithelium (RPE) originate from the optic cup of the developing forebrain. Previous studies have suggested that pigmentation genes are controlled by different regulatory networks in melanocytes and RPE. The promoter of the tyrosinase-related family gene Tyrp1 has been shown to drive detectable transgene expression only to the RPE, even though the gene is also expressed in melanocytes as evident from Tyrp1-mutant mice. This indicates that the regulatory elements responsible for Tyrp1 gene expression in the RPE are not sufficient for expression in melanocytes. We thus searched for a putative melanocyte-specific regulatory sequence and demonstrate that a bacterial artificial chromosome (BAC) containing the Tyrp1 gene and surrounding sequences is able to target transgenic expression to melanocytes and to rescue the Tyrp1b (brown) phenotype. This BAC contains several highly conserved non-coding sequences that might represent novel regulatory elements. We further focused on a sequence located at -15 kb, which we identified as a melanocyte-specific enhancer as shown by cell culture and transgenic mice experiments. In addition, we show that the transcription factor Sox10 can activate this conserved enhancer. The presence of a distal Tyrp1 regulatory element, which specifies melanocyte-specific expression, supports the idea that separate regulatory sequences can mediate differential gene expression in melanocytes and RPE.


Assuntos
Elementos Facilitadores Genéticos , Melanócitos/metabolismo , Glicoproteínas de Membrana/metabolismo , Oxirredutases/metabolismo , Transcrição Gênica , Animais , Sequência de Bases , Linhagem Celular , Cromossomos Artificiais Bacterianos , Sequência Conservada , Proteínas de Ligação a DNA/genética , Proteínas de Grupo de Alta Mobilidade/genética , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Dados de Sequência Molecular , Oxirredutases/genética , Fenótipo , Epitélio Pigmentado Ocular/citologia , Epitélio Pigmentado Ocular/metabolismo , Fatores de Transcrição SOXE , Análise de Sequência de DNA , Homologia de Sequência do Ácido Nucleico , Fatores de Transcrição/genética , Ativação Transcricional , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA