Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Am Heart Assoc ; 7(1)2018 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-29306897

RESUMO

BACKGROUND: Sudden unexplained nocturnal death syndrome (SUNDS) remains an autopsy negative entity with unclear etiology. Arrhythmia has been implicated in SUNDS. Mutations/deficiencies in intercalated disc components have been shown to cause arrhythmias. Human cardiomyopathy-associated 1 (XIRP1) and 3 (XIRP2) are intercalated disc-associated, Xin repeats-containing proteins. Mouse Xirp1 is necessary for the integrity of intercalated disc and for the surface expression of transient outward and delayed rectifier K+ channels, whereas mouse Xirp2 is required for Xirp1 intercalated disc localization. Thus, XIRP1 and XIRP2 may be potentially causal genes for SUNDS. METHODS AND RESULTS: We genetically screened XIRP genes in 134 sporadic SUNDS victims and 22 Brugada syndrome (BrS) cases in a Chinese Han population. We identified 16 rare variants (6 were in silico predicted as deleterious) in SUNDS victims, including a novel variant, XIRP2-E215K. There were also four rare variants (2 were in silico predicted as deleterious) detected in BrS cases, including a novel variant, XIRP2-L2718P. Interestingly, among these 20 variants, we detected 2 likely pathogenic variants: a nonsense variant (XIRP2-Q2875*) and a frameshift variant (XIRP2-T2238QfsX7). Analyzing available Xirp2 knockout mice, we further found that mouse hearts without Xirp2 exhibited prolonged PR and QT intervals, slow conduction velocity, atrioventricular conduction block, and an abnormal infranodal ventricular conduction system. Whole-cell patch-clamp detected altered ionic currents in Xirp2-/- cardiomyocytes, consistent with the observed association between Xirp2 and Nav1.5/Kv1.5 in co-immunoprecipitation. CONCLUSIONS: This is the first report identifying likely pathogenic XIRP rare variants in arrhythmogenic disorders such as SUNDS and Brugada syndrome, and showing critical roles of Xirp2 in cardiac conduction.


Assuntos
Síndrome de Brugada/etnologia , Proteínas de Ligação a DNA/genética , Proteínas com Domínio LIM/genética , Mutação , Proteínas Nucleares/genética , Polimorfismo de Nucleotídeo Único , Potenciais de Ação , Adolescente , Adulto , Animais , Povo Asiático/genética , Bloqueio Atrioventricular/genética , Bloqueio Atrioventricular/metabolismo , Bloqueio Atrioventricular/fisiopatologia , Síndrome de Brugada/genética , Síndrome de Brugada/metabolismo , Síndrome de Brugada/fisiopatologia , China/epidemiologia , Proteínas de Ligação a DNA/metabolismo , Morte Súbita Cardíaca/etnologia , Feminino , Predisposição Genética para Doença , Sistema de Condução Cardíaco/metabolismo , Sistema de Condução Cardíaco/fisiopatologia , Frequência Cardíaca , Humanos , Canal de Potássio Kv1.5/metabolismo , Proteínas com Domínio LIM/metabolismo , Masculino , Camundongos Knockout , Pessoa de Meia-Idade , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Proteínas Nucleares/metabolismo , Fenótipo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Fatores de Risco , Adulto Jovem
2.
Cell Rep ; 10(11): 1811-8, 2015 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-25772365

RESUMO

Hair cells of the inner ear are mechanoreceptors for hearing and balance, and proteins highly enriched in hair cells may have specific roles in the development and maintenance of the mechanotransduction apparatus. We identified XIRP2/mXinß as an enriched protein likely to be essential for hair cells. We found that different isoforms of this protein are expressed and differentially located: short splice forms (also called XEPLIN) are targeted more to stereocilia, whereas two long isoforms containing a XIN-repeat domain are in both stereocilia and cuticular plates. Mice lacking the Xirp2 gene developed normal stereocilia bundles, but these degenerated with time: stereocilia were lost and long membranous protrusions emanated from the nearby apical surfaces. At an ultrastructural level, the paracrystalline actin filaments became disorganized. XIRP2 is apparently involved in the maintenance of actin structures in stereocilia and cuticular plates of hair cells, and perhaps in other organs where it is expressed.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Células Ciliadas Auditivas/metabolismo , Proteínas com Domínio LIM/metabolismo , Proteínas Nucleares/metabolismo , Estereocílios/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestrutura , Animais , Proteínas do Citoesqueleto , Proteínas de Ligação a DNA/genética , Células Ciliadas Auditivas/ultraestrutura , Proteínas com Domínio LIM/genética , Camundongos , Proteínas Nucleares/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico
3.
Int Rev Cell Mol Biol ; 310: 89-128, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24725425

RESUMO

Since the discovery of Xin repeat-containing proteins in 1996, the importance of Xin proteins in muscle development, function, regeneration, and disease has been continuously implicated. Most Xin proteins are localized to myotendinous junctions of the skeletal muscle and also to intercalated discs (ICDs) of the heart. The Xin gene is only found in vertebrates, which are characterized by a true chambered heart. This suggests that the evolutionary origin of the Xin gene may have played a key role in vertebrate origins. Diverse vertebrates including mammals possess two paralogous genes, Xinα (or Xirp1) and Xinß (or Xirp2), and this review focuses on the role of their encoded proteins in cardiac muscles. Complete loss of mouse Xinß (mXinß) results in the failure of forming ICD, severe growth retardation, and early postnatal lethality. Deletion of mouse Xinα (mXinα) leads to late-onset cardiomyopathy with conduction defects. Molecular studies have identified three classes of mXinα-interacting proteins: catenins, actin regulators/modulators, and ion-channel subunits. Thus, mXinα acts as a scaffolding protein modulating the N-cadherin-mediated adhesion and ion-channel surface expression. Xin expression is significantly upregulated in early stages of stressed hearts, whereas Xin expression is downregulated in failing hearts from various human cardiomyopathies. Thus, mutations in these Xin loci may lead to diverse cardiomyopathies and heart failure.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Proteínas com Domínio LIM/metabolismo , Miocárdio/metabolismo , Proteínas Nucleares/metabolismo , Actinas/metabolismo , Animais , Cardiomiopatias/metabolismo , Cortactina/metabolismo , Proteínas do Citoesqueleto , Citoesqueleto/metabolismo , Regulação para Baixo , Coração/fisiologia , Insuficiência Cardíaca/metabolismo , Humanos , Camundongos , Mutação , Canais de Potássio/metabolismo , Estrutura Terciária de Proteína , Fatores de Tempo , beta Catenina/metabolismo
4.
Mol Cell Proteomics ; 12(11): 3094-107, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23882026

RESUMO

Pressure-overload stress to the heart causes pathological cardiac hypertrophy, which increases the risk of cardiac morbidity and mortality. However, the detailed signaling pathways induced by pressure overload remain unclear. Here we used phosphoproteomics to delineate signaling pathways in the myocardium responding to acute pressure overload and chronic hypertrophy in mice. Myocardial samples at 4 time points (10, 30, 60 min and 2 weeks) after transverse aortic banding (TAB) in mice underwent quantitative phosphoproteomics assay. Temporal phosphoproteomics profiles showed 360 phosphorylation sites with significant regulation after TAB. Multiple mechanical stress sensors were activated after acute pressure overload. Gene ontology analysis revealed differential phosphorylation between hearts with acute pressure overload and chronic hypertrophy. Most interestingly, analysis of the cardiac hypertrophy pathway revealed phosphorylation of the mitochondrial fission protein dynamin-related protein 1 (DRP1) by prohypertrophic kinases. Phosphorylation of DRP1 S622 was confirmed in TAB-treated mouse hearts and phenylephrine (PE)-treated rat neonatal cardiomyocytes. TAB-treated mouse hearts showed phosphorylation-mediated mitochondrial translocation of DRP1. Inhibition of DRP1 with the small-molecule inhibitor mdivi-1 reduced the TAB-induced hypertrophic responses. Mdivi-1 also prevented PE-induced hypertrophic growth and oxygen consumption in rat neonatal cardiomyocytes. We reveal the signaling responses of the heart to pressure stress in vivo and in vitro. DRP1 may be important in the development of cardiac hypertrophy.


Assuntos
Cardiomegalia/metabolismo , Dinaminas/metabolismo , Animais , Cardiomegalia/etiologia , Células Cultivadas , Dinaminas/química , Hipertrofia Ventricular Esquerda/etiologia , Hipertrofia Ventricular Esquerda/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Consumo de Oxigênio/efeitos dos fármacos , Fenilefrina/farmacologia , Fosforilação , Proteômica , Ratos , Transdução de Sinais
5.
Arch Biochem Biophys ; 535(1): 91-100, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23296090

RESUMO

The Xin repeat-containing proteins, Xinα (Xirp1) and Xinß (Xirp2), localize to the intercalated discs (ICDs) of mammalian hearts. Mouse Xinα (mXinα) directly interacts with ß-catenin and actin filaments, potentially coupling the N-cadherin/ß-catenin complexes to the underlying actin cytoskeleton and modulating ICD integrity and function. Supporting this possibility, mXinα-null hearts develop ICD structural defects and cardiomyopathy with conduction defects. However, the underlying mechanisms leading to these defects remain unclear. Here, we showed that mXinα also interacted with p120-catenin and cortactin. Different from the ß-catenin binding domain, there existed multiple p120-catenin binding sites on mXinα, while only the extreme N-terminus of mXinα containing a SH3-binding motif could interact with cortactin. In mouse heart, a significant fraction of cortactin was co-localized with N-cadherin to ICDs, whereas in mXinα-null heart, this fraction of cortactin was drastically reduced. Therefore, mXinα may modulate ICD integrity and function through its interactions with catenins and cortactin. Analyses of the in vivo consequence of p120-catenin and mXinα interaction revealed that force-expressed mXinα or its fragments significantly suppressed the p120-catenin-induced branching phenotypes. It is known that p120-catenin directly regulates Rho GTPases, leading to the branching phenotype. Thus, mXinα may sequester the p120-catenin from inhibiting RhoA activity and/or from activating Rac1 activity.


Assuntos
Junções Aderentes/metabolismo , Cateninas/metabolismo , Forma Celular/fisiologia , Cortactina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/fisiologia , Junções Aderentes/fisiologia , Animais , Sítios de Ligação , Células CHO , Caderinas/metabolismo , Adesão Celular , Cricetinae , Proteínas do Citoesqueleto , DNA Complementar/genética , DNA Complementar/metabolismo , Coração/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Fenótipo , Plasmídeos/genética , Plasmídeos/metabolismo , Mapeamento de Interação de Proteínas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfecção , Domínios de Homologia de src , delta Catenina
6.
Dev Biol ; 374(2): 264-80, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23261932

RESUMO

The intercalated disc (ICD) is a unique structure to the heart and plays vital roles in communication and signaling among cardiomyocytes. ICDs are formed and matured during postnatal development through a profound redistribution of the intercellular junctions, as well as recruitment and assembly of more than 200 proteins at the termini of cardiomyocytes. The molecular mechanism underlying this process is not completely understood. The mouse orthologs (mXinα and mXinß) of human cardiomyopathy-associated (CMYA)/Xin actin-binding repeat-containing protein (XIRP) genes (CMYA1/XIRP1 and CMYA3/XIRP2, respectively) encode proteins localized to ICDs. Ablation of mXinα results in adult late-onset cardiomyopathy with conduction defects and up-regulation of mXinß. ICD structural defects are found in adult but not juvenile mXinα-null hearts. On the other hand, loss of mXinß leads to ICD defects at postnatal day 16.5, a developmental stage when the heart is forming ICDs, suggesting mXinß is required for ICD formation. Using quantitative Western blot, we showed in this study that mXinß but not mXinα was uniquely up-regulated during the redistribution of intercellular junction from the lateral membrane of cardiomyocytes to their termini. In the absence of mXinß, the intercellular junctions failed to be restricted to the termini of the cells, and the onset of such defect correlated with the peak expression of mXinß. Immunofluorescence staining and subcellular fractionation showed that mXinß preferentially associated with the forming ICDs, further suggesting that mXinß functioned locally to promote ICD maturation. In contrast, the spatiotemporal expression profile of mXinα and the lack of more severe ICD defects in mXinα-/-;mXinß-/- double knockout hearts than in mXinß-/- hearts suggested that mXinα was not essential for the postnatal formation of ICDs. A two-step model for the development of ICD is proposed where mXinß is essential for the redistribution of intercellular junction components from the lateral puncta to the cell termini.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Coração/crescimento & desenvolvimento , Proteínas com Domínio LIM/metabolismo , Miocárdio/metabolismo , Proteínas Nucleares/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Proteínas do Citoesqueleto , Proteínas de Ligação a DNA/genética , Feminino , Coração/embriologia , Imuno-Histoquímica , Junções Intercelulares/metabolismo , Proteínas com Domínio LIM/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Miocárdio/citologia , Miócitos Cardíacos/metabolismo , Proteínas Nucleares/genética
7.
J Cardiol Clin Res ; 1(2): 1011, 2013 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-24734257
8.
Front Biosci (Elite Ed) ; 3(4): 1425-42, 2011 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-21622147

RESUMO

Mouse Xin-alpha (mXin-alpha) encodes a Xin repeat-containing, actin-binding protein localized to the intercalated disc (ICD). Ablation of mXin-alpha progressively leads to disrupted ICD structure, cardiac hypertrophy and cardiomyopathy with conduction defects during adulthood. Such conduction defects could be due to ICD structural defects and/or cell electrophysiological property changes. Here, we showed that despite the normal ICD structure, juvenile mXina-null cardiomyocytes (from 3~4-week-old mice) exhibited a significant reduction in the transient outward K+ current (ITO), similar to adult mutant cells. Juvenile but not adult mutant cardiomyocytes also had a significant reduction in the delayed rectifier K+ current. In contrast, the mutant adult ventricular myocytes had a significant reduction in the inward rectifier K+ current (IK1) on hyperpolarization. These together could account for the prolongation of action potential duration (APD) and the ease of developing early afterdepolarization observed in juvenile mXin-alpha-null cells. Interestingly, juvenile mXin-alpha-null cardiomyocytes had a notable decrease in the amplitude of intracellular Ca2+ transient and no change in the L-type Ca2+ current, suggesting that the prolonged APD did not promote an increase in intracellular Ca2+ for cardiac hypertrophy. Juvenile mXin-alpha-null ventricles had reduced levels of membrane-associated Kv channel interacting protein 2, an auxiliary subunit of ITO, and filamin, an actin cross-linking protein. We further showed that mXin-alpha interacted with both proteins, providing a novel mechanism for ITO surface expression.


Assuntos
Ventrículos do Coração/metabolismo , Proteínas Musculares/metabolismo , Animais , Sequência de Bases , Células Cultivadas , Primers do DNA , Ventrículos do Coração/citologia , Ventrículos do Coração/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Proteínas Musculares/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Técnicas do Sistema de Duplo-Híbrido
9.
Am J Physiol Cell Physiol ; 300(6): C1356-65, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21289288

RESUMO

Tropomyosin (Tm) is known to be an important gatekeeper of actin function. Tm isoforms are encoded by four genes, and each gene produces several variants by alternative splicing, which have been proposed to play roles in motility, proliferation, and apoptosis. Smooth muscle studies have focused on gizzard smooth muscle, where a heterodimer of Tm from the α-gene (Tmsm-α) and from the ß-gene (Tmsm-ß) is associated with contractile filaments. In this study we examined Tm in differentiated mammalian vascular smooth muscle (dVSM). Liquid chromatography-tandem mass spectrometry (LC MS/MS) analysis and Western blot screening with variant-specific antibodies revealed that at least five different Tm proteins are expressed in this tissue: Tm6 (Tmsm-α) and Tm2 from the α-gene, Tm1 (Tmsm-ß) from the ß-gene, Tm5NM1 from the γ-gene, and Tm4 from the δ-gene. Tm6 is by far most abundant in dVSM followed by Tm1, Tm2, Tm5NM1, and Tm4. Coimmunoprecipitation and coimmunofluorescence studies demonstrate that Tm1 and Tm6 coassociate with different actin isoforms and display different intracellular localizations. Using an antibody specific for cytoplasmic γ-actin, we report here the presence of a γ-actin cortical cytoskeleton in dVSM cells. Tm1 colocalizes with cortical cytoplasmic γ-actin and coprecipitates with γ-actin. Tm6, on the other hand, is located on contractile bundles. These data indicate that Tm1 and Tm6 do not form a classical heterodimer in dVSM but rather describe different functional cellular compartments.


Assuntos
Diferenciação Celular/fisiologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/fisiologia , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Tropomiosina/química , Tropomiosina/metabolismo , Actinas/genética , Actinas/metabolismo , Sequência de Aminoácidos , Animais , Galinhas , Furões , Humanos , Dados de Sequência Molecular , Miócitos de Músculo Liso/citologia , Ligação Proteica , Isoformas de Proteínas/genética , Alinhamento de Sequência , Tropomiosina/genética
10.
Circ Res ; 106(9): 1468-78, 2010 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-20360251

RESUMO

RATIONALE: The Xin repeat-containing proteins mXinalpha and mXinbeta localize to the intercalated disc of mouse heart and are implicated in cardiac development and function. The mXinalpha directly interacts with beta-catenin, p120-catenin, and actin filaments. Ablation of mXinalpha results in adult late-onset cardiomyopathy with conduction defects. An upregulation of the mXinbeta in mXinalpha-deficient hearts suggests a partial compensation. OBJECTIVE: The essential roles of mXinbeta in cardiac development and intercalated disc maturation were investigated. METHODS AND RESULTS: Ablation of mXinbeta led to abnormal heart shape, ventricular septal defects, severe growth retardation, and postnatal lethality with no upregulation of the mXinalpha. Postnatal upregulation of mXinbeta in wild-type hearts, as well as altered apoptosis and proliferation in mXinbeta-null hearts, suggests that mXinbeta is required for postnatal heart remodeling. The mXinbeta-null hearts exhibited a misorganized myocardium as detected by histological and electron microscopic studies and an impaired diastolic function, as suggested by echocardiography and a delay in switching off the slow skeletal troponin I. Loss of mXinbeta resulted in the failure of forming mature intercalated discs and the mislocalization of mXinalpha and N-cadherin. The mXinbeta-null hearts showed upregulation of active Stat3 (signal transducer and activator of transcription 3) and downregulation of the activities of Rac1, insulin-like growth factor 1 receptor, protein kinase B, and extracellular signal-regulated kinases 1 and 2. CONCLUSIONS: These findings identify not only an essential role of mXinbeta in the intercalated disc maturation but also mechanisms of mXinbeta modulating N-cadherin-mediated adhesion signaling and its crosstalk signaling for postnatal heart growth and animal survival.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Coração/crescimento & desenvolvimento , Coração/fisiopatologia , Proteínas Nucleares/metabolismo , Animais , Animais Recém-Nascidos , Proliferação de Células , Sobrevivência Celular , Proteínas do Citoesqueleto , Proteínas de Ligação a DNA/deficiência , Proteínas com Domínio LIM , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/deficiência
11.
Int Rev Cell Mol Biol ; 274: 1-68, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19349035

RESUMO

Caldesmon (CaD) is a multimodular protein that regulates contractility and actin cytoskeleton remodeling in smooth muscle and nonmuscle cells. A single gene (CALD1) encodes high molecular mass CaD (h-CaD) and low molecular mass CaD (l-CaD) by alternative splicings. The h-CaD exclusively expresses in smooth muscle, whereas the l-CaD ubiquitously expresses in all cell types except skeletal muscle. The h-CaD/l-CaD ratio could be a marker for monitoring differentiating and pathological states of smooth muscles. The l-CaD associates with stress fibers and membrane ruffles in nonmuscle cells and with the actin core of podosomes in highly motile/invasive cells. Together with tropomyosin, CaD stabilizes actin filaments and inhibits actin-tropomyosin-activated myosin ATPase activity. This inhibition can be effectively released by Ca(2+)-calmodulin and/or by phosphorylation with various kinases. Through its interactions with a spectrum of actin-binding proteins, CaD modulates dynamics of cortical actin networks and stress fibers, which are essential to cell motility and cytoskeleton rearrangement. Regulation of CaD level and its activity may provide a novel strategy for gene therapy.


Assuntos
Citoesqueleto de Actina/fisiologia , Proteínas de Ligação a Calmodulina/fisiologia , Movimento Celular/fisiologia , Citoesqueleto/fisiologia , Sequência de Aminoácidos , Animais , Humanos , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos
12.
Anat Rec (Hoboken) ; 291(12): 1574-86, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18951515

RESUMO

The cardiac-specific -497 bp promoter of rat cardiac troponin T (cTnT) contains two similar modules, D and F, each of which possesses TCTG(G/C) direct repeats and A/T-rich sites. To identify cis-elements critical for cardiac specificity, a -249 bp promoter containing only module F and its site-directed mutations were used to generate transgenic mice. Transgene expression of the -249 bp promoter remained cardiac-specific, despite low and nonuniform expression. The nonuniform expression pattern of the transgene coincided with differential expression of HMGB1, which appeared to be the predominant form of HMGB family proteins in the heart. The HMGB1 binds to the A/T-rich/MEF2-like sites of the cTnT promoter, as determined by chromatin immunoprecipitation assays. Mice carrying the -249 bp promoter with point mutations disrupting the direct repeats expressed transgene at lower levels in the heart and ectopically in the brain. Ectopic expression of transgene was also observed in developing limbs and head. These results suggest an important role for the direct repeat in determining the cardiac specificity. Furthermore, mice carrying a mutant promoter simultaneously disrupting the direct repeats and overlapping GATA site failed to express the transgene in any tissues tested. Therefore, the direct repeat and overlapping GATA site are critical for the expression level and cardiac specificity. The F module controls one level of cardiac specificity. For a uniform and high level of cardiac-specific expression, the upstream element (-497 to -250 bp) is further required, possibly through the D enhancer module and the combination of Nkx2.5 and GATA sites.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Coração/embriologia , Miocárdio/metabolismo , Regiões Promotoras Genéticas/genética , Sequências Repetitivas de Ácido Nucleico/genética , Troponina T/genética , Animais , Sequência de Bases/genética , Padronização Corporal/genética , Sinalização do Cálcio/genética , Diferenciação Celular/genética , Elementos Facilitadores Genéticos/genética , Fatores de Transcrição GATA/genética , Camundongos , Camundongos Transgênicos , Contração Muscular/genética , Mutagênese Sítio-Dirigida , Miocárdio/ultraestrutura , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/ultraestrutura , Mutação Puntual/genética , Elementos Reguladores de Transcrição/genética , Transcrição Gênica/genética , Transgenes/genética , Troponina T/biossíntese
13.
Biomed Chromatogr ; 22(12): 1374-84, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18814195

RESUMO

Simultaneous extraction, separation and quantitation of reduced nicotinamide adenine dinucleotide (NADH), reduced nicotinamide adenine dinucleotide phosphate (NADPH), flavin adenine dinucleotide (FAD) and flavin mononucleotide (FMN) in Chinese Hamster Ovary (CHO) cells were investigated. The separation of flavins and nicotinamide cofactors was performed by capillary electrophoresis with laser-induced fluorescence detection at the excitation wavelength of 325 nm. The separation protocol was established by investigating the excitation wavelength, high voltage and effects of buffer nature, pH and concentration. All endogenous fluorophores riboflavin, FAD, FMN, NADH and NADPH show wide linear range of quantitation. The limits of detection for the five compounds ranged from 4.5 to 23 nM. Extraction conditions were optimized for high-efficiency recovery of all endogenous fluorophores from CHO cells. To account for the complex matrix of cell extracts, a standard addition method was used to quantify FAD, FMN, NADH and NADPH in CHO cells. The quantitative results should be useful to reveal the metabolic status of cells. The protocols for extraction, separation and quantitation are readily adaptable to normal and cancer cell lines for the analysis of endogenous fluorophores.


Assuntos
Eletroforese Capilar/métodos , Flavinas/isolamento & purificação , Niacinamida/isolamento & purificação , Animais , Células CHO , Cricetinae , Cricetulus , NAD/isolamento & purificação , NADP/isolamento & purificação , Reprodutibilidade dos Testes , Espectrometria de Fluorescência
14.
PLoS One ; 3(8): e2857, 2008 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-18682726

RESUMO

The mouse Xin repeat-containing proteins (mXinalpha and mXinbeta) localize to the intercalated disc in the heart. mXinalpha is able to bundle actin filaments and to interact with beta-catenin, suggesting a role in linking the actin cytoskeleton to N-cadherin/beta-catenin adhesion. mXinalpha-null mouse hearts display progressively ultrastructural alterations at the intercalated discs, and develop cardiac hypertrophy and cardiomyopathy with conduction defects. The up-regulation of mXinbeta in mXinalpha-deficient mice suggests a partial compensation for the loss of mXinalpha. To elucidate the evolutionary relationship between these proteins and to identify the origin of Xin, a phylogenetic analysis was done with 40 vertebrate Xins. Our results show that the ancestral Xin originated prior to the emergence of lamprey and subsequently underwent gene duplication early in the vertebrate lineage. A subsequent teleost-specific genome duplication resulted in most teleosts encoding at least three genes. All Xins contain a highly conserved beta-catenin-binding domain within the Xin repeat region. Similar to mouse Xins, chicken, frog and zebrafish Xins also co-localized with beta-catenin to structures that appear to be the intercalated disc. A putative DNA-binding domain in the N-terminus of all Xins is strongly conserved, whereas the previously characterized Mena/VASP-binding domain is a derived trait found only in Xinalphas from placental mammals. In the C-terminus, Xinalphas and Xinbetas are more divergent relative to each other but each isoform from mammals shows a high degree of within-isoform sequence identity. This suggests different but conserved functions for mammalian Xinalpha and Xinbeta. Interestingly, the origin of Xin ca. 550 million years ago coincides with the genesis of heart chambers with complete endothelial and myocardial layers. We postulate that the emergence of the Xin paralogs and their functional differentiation may have played a key role in the evolutionary development of the heart.


Assuntos
Proteínas de Ligação a DNA/genética , Coração/fisiologia , Proteínas Nucleares/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação , Sequência Conservada , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/fisiologia , Evolução Molecular , Humanos , Camundongos , Dados de Sequência Molecular , Proteínas Nucleares/química , Proteínas Nucleares/fisiologia , Filogenia , RNA Mensageiro/genética , Sequências Repetitivas de Ácido Nucleico , Tempo , beta Catenina/química , beta Catenina/fisiologia
15.
Life Sci ; 83(7-8): 272-83, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18644388

RESUMO

We have previously shown that left atrial-pulmonary vein tissue (LA-PV) can generate reentrant arrhythmias (atrial fibrillation, AF) in wild-type (mXinalpha+/+) but not in mXinalpha-null (mXinalpha-/-) mice. With the present experiments, we investigated the arrhythmogenic activity and the underlying mechanisms in mXinalpha+/+ vs. mXinalpha-/- LA-PV. Electrical activity and conduction velocity (CV) were recorded in LA-PV by means of a MED64 system. CV was significantly faster in mXinalpha+/+ than in mXinalpha-/- LA-PV and it was increased by 1 muM isoproterenol (ISO). AF could be induced by fast pacing in the mXinalpha+/+ but not in mXinalpha-/- LA-PV where automatic rhythms could occur. ISO increased the incidence of AF in Xinalpha+/+ whereas it increased that of automatic rhythms in mXinalpha-/- LA-PV. In LA-PV with the right atrium attached (RA-LA-PV), automatic rhythms occurred in all preparations. In mXinalpha+/+ RA-LA-PV simultaneously treated with ISO, strophanthidin and atropine, the incidence of the automatic rhythm was about the same, but AF increased significantly. In contrast, in mXinalpha-/- RA-LA-PV under the same condition, the automatic rhythm was markedly enhanced, but still no AF occurred. Conventional microelectrode techniques showed a longer APD(90) and a less negative maximum diastolic potential (MDP) in mXinalpha-/- than mXinalpha+/+ LA-PV tissues. Whole-cell current clamp experiments also showed a less negative MDP in mXinalpha-/- vs. mXinalpha+/+ LA-PV cardiomyocytes. The fact that AF could be induced by fast pacing under several conditions in mXinalpha+/+ but not in mXinalpha-/- LA-PV preparations appears to be due to a slower CV, a prolonged APD(90), a less negative MDP and possibly larger areas of conduction block in mXinalpha-/- myocardial cells. In contrast, the non-impairment of automatic and triggered rhythms in mXinalpha-/- preparations may be due to the fact that the mechanisms underlying these rhythms do not involve cell-to-cell conduction.


Assuntos
Fibrilação Atrial/fisiopatologia , Comunicação Celular , Proteínas de Ligação a DNA , Miocárdio , Proteínas Nucleares , Veias Pulmonares/fisiopatologia , Animais , Antiarrítmicos/farmacologia , Fibrilação Atrial/genética , Atropina/farmacologia , Cardiotônicos/farmacologia , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/genética , Proteínas de Ligação a DNA/genética , Condutividade Elétrica , Técnicas Eletrofisiológicas Cardíacas/métodos , Isoproterenol/farmacologia , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Estrofantidina/farmacologia
16.
Adv Exp Med Biol ; 644: 201-22, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19209824

RESUMO

Over the past two decades, extensive molecular studies have identified multiple tropomyosin isoforms existing in all mammalian cells and tissues. In humans, tropomyosins are encoded by TPM1 (alpha-Tm, 15q22.1), TPM2 (beta-Tm, 9p13.2-p13.1), TPM3 (gamma-Tm, 1q21.2) and TPM4 (delta-Tm, 19p13.1) genes. Through the use of different promoters, alternatively spliced exons and different sites of poly(A) addition signals, at least 22 different tropomyosin cDNAs with full-length open reading frame have been cloned. Compelling evidence suggests that these isoforms play important determinants for actin cytoskeleton functions, such as intracellular vesicle movement, cell migration, cytokinesis, cell proliferation and apoptosis. In vitro biochemical studies and in vivo localization studies suggest that different tropomyosin isoforms have differences in their actin-binding properties and their effects on other actin-binding protein functions and thus, in their specification ofactin microfilaments. In this chapter, we will review what has been learned from experimental studies on human tropomyosin isoforms about the mechanisms for differential localization and functions of tropomyosin. First, we summarize current information concerning human tropomyosin isoforms and relate this to the functions of structural homologues in rodents. We will discuss general strategies for differential localization oftropomyosin isoforms, particularly focusing on differential protein turnover and differential isoform effects on other actin binding protein functions. We will then review tropomyosin functions in regulating cell motility and in modulating the anti-angiogenic activity of cleaved high molecular weight kininogen (HKa) and discuss future directions in this area.


Assuntos
Citoesqueleto/metabolismo , Tropomiosina/química , Tropomiosina/fisiologia , Animais , Apoptose , Proliferação de Células , Humanos , Cininogênios/metabolismo , Camundongos , Músculos/metabolismo , Neovascularização Patológica , Regiões Promotoras Genéticas , Isoformas de Proteínas , Estrutura Terciária de Proteína , Ratos , Transdução de Sinais , Tropomiosina/genética
17.
Cell Motil Cytoskeleton ; 65(3): 197-215, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18044718

RESUMO

Myosin Va, an actin-based motor protein that transports intracellular cargos, can bundle actin in vitro. Whether myosin Va regulates cellular actin dynamics or cell migration remains unclear. To address this, we compared Chinese Hamster Ovary (CHO) cells that stably express GFP fused to either full length mouse myosin Va (GFP-M5) or heavy meromyosin Va (GFP-M5Delta). GFP-M5 and GFP-M5Delta co-immunoprecipitate with CHO myosin Va and serve as overexpression of wild-type and dominant negative mutants of myosin Va. Compared to non-expressing control cells, GFP-M5-overexpressing cells have peripheral endocytic vesicles, spread slowly after plating, as well as produce robust interior actin stress fibers, myosin II bundles, and focal adhesions. However, these cells display normal cell migration and lamellipodial dynamics. In contrast, GFP-M5Delta-expressing cells have perinuclear endocytic vesicles, produce thin interior actin and myosin bundles and contain no interior focal adhesions. In addition, these cells spread rapidly, migrate slowly and display reduced lamellipodial dynamics. Similarly, neurite outgrowth is compromised in neurons cultured from transgenic Drosophila that express M5Delta-dsRed and in neurons cultured from Drosophila that produce a tailless version of endogenous myosin V. Together, these data suggest that myosin Va overexpression induces actin bundles in vivo whereas the tailless version fails to bundle actin and disrupts cell motility.


Assuntos
Movimento Celular , Citoesqueleto/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Subfragmentos de Miosina/metabolismo , Miosina Tipo V/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Vesículas Transportadoras/metabolismo , Actinas/metabolismo , Animais , Células CHO , Divisão Celular , Proliferação de Células , Forma Celular , Cricetinae , Cricetulus , Drosophila melanogaster , Endocitose , Proteínas de Fluorescência Verde/metabolismo , Imunoprecipitação , Camundongos , Proteínas Mutantes/metabolismo , Miosina Tipo II/metabolismo , Transporte Proteico , Proteínas Recombinantes de Fusão/metabolismo
18.
Anadolu Kardiyol Derg ; 7 Suppl 1: 90-2, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17584692

RESUMO

OBJECTIVE: mXin alpha, a downstream target gene of Nkx2.5 transcription factor, was shown to encode a proline-rich and Xin repeats-containing protein which localizes to the intercalated disc of adult hearts. Our previous voltage-clamp studies have shown that the ventricular myocytes of mXin alpha -deficient mice exhibited a significant reduction in K+ currents (Ito and IK1), L-type Ca2+ currents, and maximum diastolic potential, leading to the development of early afterdepolarization (EAD) and arrhythmias. However, changes in cationic inward currents could also contribute to the genesis of EAD and arrhythmias in mXin alpha -deficient mice. METHODS: The present study aims to characterize changes in Na+ currents on depolarization and transient inward currents (Iti) on repolarization. Conduction velocity (CV) on the frontal surface of ventricles were also measured and compared. RESULTS: Results of optical mapping on the Langendorff-perfused hearts at 37oC revealed a 36% reduction of CV in mXin alpha -/- ventricle. Pacing (3 Hz)-induced tachyarrhythmias were more frequently found and ventricular fibrillation (VF, 21 Hz for 5 min) occurred in one out of 8 mXin alpha-/- heart. When perfused at 30 degrees C, no VF was observed in both types of preparations. Voltage-clamp study on isolated ventricular myocytes at 37 degrees C shows increase in INa and Iti in mXin alpha -/- cardiomyocytes thus could explain the occurrence of re-entrant triggered arrhythmias. CONCLUSION: The present results revealed that the CV was slower, but INa and Iti were increased in mXin alpha -/-cardiomyocytes thus were prone to reentrant triggered arrhythmias. Hypothermia could reduce the occurrence of arrhythmias.


Assuntos
Arritmias Cardíacas/fisiopatologia , Proteínas de Ligação a DNA , Hipertrofia Ventricular Esquerda/fisiopatologia , Canais Iônicos/fisiologia , Proteínas Nucleares , Potenciais de Ação , Animais , Masculino , Camundongos , Camundongos Endogâmicos , Células Musculares/fisiologia , Miocárdio/citologia , Técnicas de Patch-Clamp
19.
Methods Mol Biol ; 366: 183-201, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17568125

RESUMO

To understand how cardiac gene expression is regulated, the identification and characterization of cis-regulatory elements and their trans-acting factors by gel mobility shift assay (GMSA) or gel retardation assay are essential and common steps. In addition to providing a general protocol for GMSA, this chapter describes some applications of this assay to characterize cardiac-specific and ubiquitous trans-acting factors bound to regulatory elements [novel TCTG(G/C) direct repeat and A/T-rich region] of the rat cardiac troponin T promoter. In GMSA, the specificity of the binding of trans-acting factor to labeled DNA probe should be verified by the addition of unlabeled probe in the reaction mixture. The migratory property of DNA-protein complexes formed by protein extracts prepared from different tissues can be compared to determine the tissue specificity of trans-acting factors. GMSA, coupled with specific antibody to trans-acting factor (antibody supershift assay), is used to identify proteins present in the DNA-protein complex. The gel-shift competition assay with an unlabeled probe containing a slightly different sequence is a powerful technique used to assess the sequence specificity and relative binding affinity of a DNA-protein interaction. GMSA with SDS-PAGE fractionated proteins allows for the determination of the apparent molecular mass of bound trans-acting factor.


Assuntos
Ensaio de Desvio de Mobilidade Eletroforética/métodos , Miocárdio/metabolismo , Regiões Promotoras Genéticas , Elementos Reguladores de Transcrição/genética , Fatores de Transcrição/genética , Animais , Sequência de Bases , Células Cultivadas , Eletroforese em Gel de Poliacrilamida , Mucosa Gástrica/metabolismo , Fígado/metabolismo , Dados de Sequência Molecular , Músculo Esquelético/metabolismo , Ligação Proteica , Ratos , Homologia de Sequência do Ácido Nucleico , Troponina T/genética
20.
Parasitol Res ; 99(1): 45-54, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16479376

RESUMO

The actin cytoskeleton of host cells has been implicated in Cryptosporidium invasion. However, the underlying mechanism of how actin filaments and associated proteins modulate this process remains unclear. In this study, we use in vitro cultured cell lines, human ileocecal adenocarcinoma HCT-8 and Chinese hamster ovary (CHO), and an in vivo mouse model to investigate the roles of tropomyosin isoforms in Cryptosporidium invasion. Using isoform-specific monoclonal antibodies, we found that the major human tropomyosin (hTM) isoforms expressed in HCT-8 cells are hTM4 and hTM5. HCT-8 cells also express hTM1 at low levels but not hTM2 and hTM3. During Cryptosporidium parvum infection, hTM5 colocalized to the infection sites with a novel parasite membrane protein, CP2. Neither hTM1 nor hTM4 accumulated at infection sites. Similarly, a high level of TM5 and varying amounts of TM4 accumulated at the C. parvum infection sites in CHO cells. CHO cells overexpressing hTM5 exhibit a significantly higher percent of mature meronts early in the infection process relative to CHO cells or CHO cells overexpressing a tropomyosin mutant, chimeric isoform hTM5/3. These results suggest that functional TM5 enhances Cryptosporidium invasion of host cells. In C. parvum-infected mice, accumulation and rearrangement of TM5 and TM4 were detected throughout the infected ileum. Similarly, in the Cryptosporidium muris-infected mice, TM5 accumulated in discrete regions of the epithelial cells of gastric glands and in the oocyst-laden stomach gland lumen. Cryptosporidium infection appears to rearrange and recruit host TM isoforms in both culture cells and in the mouse. Localized accumulation of tropomyosin at the infection sites may facilitate parasite invasion.


Assuntos
Criptosporidiose/metabolismo , Cryptosporidium , Proteínas de Membrana/metabolismo , Proteínas de Protozoários/metabolismo , Tropomiosina/metabolismo , Animais , Western Blotting , Células CHO , Linhagem Celular Tumoral , Cricetinae , Feminino , Imunofluorescência , Mucosa Gástrica/metabolismo , Humanos , Íleo/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Isoformas de Proteínas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...