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.
Biochem Biophys Res Commun ; 586: 14-19, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34823217

RESUMO

Plakophilin3 (PKP3) loss leads to tumor progression and metastasis of colon cancer cells. The goal of this report was to determine if PKP3 loss led to increased disease progression in mice. We generated a colonocyte-specific knockout of PKP3 in APCmin mice, which led to increased adenoma formation, the formation of rectal prolapse, and a significant decrease in survival. The observed increase in rectal prolapse formation and decrease in survival correlated with an increase in the expression of Lipocalin2 (LCN2). Increased disease progression was observed even upon treatment with 5-fluorouracil (5FU). These results suggest that an increase in LCN2 expression might lead to therapy resistance and that LCN2 might serve as a potential therapeutic target in colorectal cancer.


Assuntos
Adenoma/genética , Neoplasias Colorretais/genética , Resistencia a Medicamentos Antineoplásicos/genética , Lipocalina-2/genética , Placofilinas/genética , Prolapso Retal/genética , Adenoma/tratamento farmacológico , Adenoma/mortalidade , Adenoma/patologia , Animais , Antimetabólitos Antineoplásicos/farmacologia , Colo/efeitos dos fármacos , Colo/metabolismo , Colo/patologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Progressão da Doença , Feminino , Fluoruracila/farmacologia , Regulação Neoplásica da Expressão Gênica , Queratina-8/genética , Queratina-8/metabolismo , Lipocalina-2/metabolismo , Masculino , Metaloproteinase 7 da Matriz/genética , Metaloproteinase 7 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Placofilinas/deficiência , Prolapso Retal/tratamento farmacológico , Prolapso Retal/mortalidade , Prolapso Retal/patologia , Transdução de Sinais , Análise de Sobrevida
2.
PLoS One ; 16(6): e0252649, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34086773

RESUMO

Arrhythmogenic right ventricular cardiomyopathy is a hereditary, rare disease with an increased risk for sudden cardiac death. The disease-causing mutations are located within the desmosomal complex and the highest incidence is found in plakophilin2. However, there are other factors playing a role for the disease progression unrelated to the genotype such as inflammation or exercise. Competitive sports have been identified as risk factor, but the type and extend of physical activity as cofactor for arrhythmogenesis remains under debate. We thus studied the effect of light voluntary exercise on cardiac health in a mouse model. Mice with a heterozygous PKP2 loss-of-function mutation were given the option to exercise in a running wheel which was monitored 24 h/d. We analyzed structural and functional development in vivo by echocardiography which revealed that neither the genotype nor the exercise caused any significant structural changes. Ejection fraction and fractional shortening were not influenced by the genotype itself, but exercise did cause a drop in both parameters after 8 weeks, which returned to normal after 16 weeks of training. The electrophysiological analysis revealed that the arrhythmogenic potential was slightly higher in heterozygous animals (50% vs 18% in wt littermates) and that an additional stressor (isoprenaline) did not lead to an increase of arrhythmogenic events pre run or after 8 weeks of running but the vulnerability was increased after 16 weeks. Exercise-induced alterations in Ca handling and contractility of isolated myocytes were mostly abolished in heterozygous animals. No fibrofatty replacements or rearrangement of gap junctions could be observed. Taken together we could show that light voluntary exercise can cause a transient aggravation of the mutation-induced phenotype which is abolished after long term exercise indicating a beneficial effect of long term light exercise.


Assuntos
Condicionamento Físico Animal , Placofilinas/genética , Animais , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patologia , Sinalização do Cálcio , Conexina 43/metabolismo , Modelos Animais de Doenças , Ecocardiografia , Fenômenos Eletrofisiológicos , Junções Comunicantes/metabolismo , Genótipo , Ventrículos do Coração/patologia , Heterozigoto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Miocárdio/metabolismo , Miocárdio/patologia , Fenótipo , Placofilinas/deficiência , Função Ventricular/fisiologia
3.
Cell Mol Life Sci ; 76(17): 3465-3476, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30949721

RESUMO

Plakophilins (Pkp) are desmosomal plaque proteins crucial for desmosomal adhesion and participate in the regulation of desmosomal turnover and signaling. However, direct evidence that Pkps regulate clustering and molecular binding properties of desmosomal cadherins is missing. Here, keratinocytes lacking either Pkp1 or 3 in comparison to wild type (wt) keratinocytes were characterized with regard to their desmoglein (Dsg) 1- and 3-binding properties and their capability to induce Dsg3 clustering. As revealed by atomic force microscopy (AFM), both Pkp-deficient keratinocyte cell lines showed reduced membrane availability and binding frequency of Dsg1 and 3 at cell borders. Extracellular crosslinking and AFM cluster mapping demonstrated that Pkp1 but not Pkp3 is required for Dsg3 clustering. Accordingly, Dsg3 overexpression reconstituted cluster formation in Pkp3- but not Pkp1-deficient keratinocytes as shown by AFM and STED experiments. Taken together, these data demonstrate that both Pkp1 and 3 regulate Dsg membrane availability, whereas Pkp1 but not Pkp3 is required for Dsg3 clustering.


Assuntos
Adesão Celular , Desmogleína 1/metabolismo , Desmogleína 3/metabolismo , Placofilinas/genética , Animais , Anisomicina/farmacologia , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Desmogleína 1/genética , Desmogleína 3/genética , Queratinócitos/citologia , Queratinócitos/metabolismo , Camundongos , Microscopia de Força Atômica , Placofilinas/deficiência , Placofilinas/metabolismo , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
4.
Exp Cell Res ; 369(2): 251-265, 2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-29803740

RESUMO

An increase in tumour formation and metastasis are observed upon plakophilin3 (PKP3) loss. To identify pathways downstream of PKP3 loss that are required for increased tumour formation, a gene expression analysis was performed, which demonstrated that the expression of lipocalin2 (LCN2) was elevated upon PKP3 loss and this is consistent with expression data from human tumour samples suggesting that PKP3 loss correlates with an increase in LCN2 expression. PKP3 loss leads to an increase in invasion, tumour formation and metastasis and these phenotypes were dependent on the increase in LCN2 expression. The increased LCN2 expression was due to an increase in the activation of p38 MAPK in the HCT116 derived PKP3 knockdown clones as LCN2 expression decreased upon inhibition of p38 MAPK. The phosphorylated active form of p38 MAPK is translocated to the nucleus upon PKP3 loss and is dependent on complex formation between p38 MAPK and PKP3. WT PKP3 inhibits LCN2 reporter activity in PKP3 knockdown cells but a PKP3 mutant that fails to form a complex with p38 MAPK cannot suppress LCN2 promoter activity. Further, LCN2 expression is decreased upon loss of p38ß, but not p38α, in the PKP3 knockdown cells. These results suggest that PKP3 loss leads to an increase in the nuclear translocation of p38 MAPK and p38ß MAPK is required for the increase in LCN2 expression.


Assuntos
Lipocalina-2/metabolismo , Neoplasias/metabolismo , Placofilinas/deficiência , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HCT116 , Xenoenxertos , Humanos , Lipocalina-2/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mutação , Neoplasias/etiologia , Neoplasias/genética , Placofilinas/antagonistas & inibidores , Placofilinas/genética , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
Circ Res ; 119(6): 731-50, 2016 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-27470638

RESUMO

RATIONALE: PKP2, encoding plakophilin 2 (PKP2), is the most common causal gene for arrhythmogenic cardiomyopathy. OBJECTIVE: To characterize miRNA expression profile in PKP2-deficient cells. METHODS AND RESULTS: Control and PKP2-knockdown HL-1 (HL-1(Pkp2-shRNA)) cells were screened for 750 miRNAs using low-density microfluidic panels. Fifty-nine miRNAs were differentially expressed. MiR-184 was the most downregulated miRNA. Expression of miR-184 in the heart and cardiac myocyte was developmentally downregulated and was low in mature myocytes. MicroRNA-184 was predominantly expressed in cardiac mesenchymal progenitor cells. Knockdown of Pkp2 in cardiac mesenchymal progenitor cells also reduced miR-184 levels. Expression of miR-184 was transcriptionally regulated by the E2F1 pathway, which was suppressed in PKP2-deficient cells. Activation of E2F1, on overexpression of its activator CCND1 (cyclin D1) or knockdown of its inhibitor retinoblastoma 1, partially rescued miR-184 levels. In addition, DNA methyltransferase-1 was recruited to the promoter region of miR-184, and the CpG sites at the upstream region of miR-184 were hypermethylated. Treatment with 5-aza-2'-deoxycytidine, a demethylation agent, and knockdown of DNA methyltransferase-1 partially rescued miR-184 level. Pathway analysis of paired miR-184:mRNA targets identified cell proliferation, differentiation, and death as the main affected biological processes. Knockdown of miR-184 in HL-1 cells and mesenchymal progenitor cells induced and, conversely, its overexpression attenuated adipogenesis. CONCLUSIONS: PKP2 deficiency leads to suppression of the E2F1 pathway and hypermethylation of the CpG sites at miR-184 promoter, resulting in downregulation of miR-184 levels. Suppression of miR-184 enhances and its activation attenuates adipogenesis in vitro. Thus, miR-184 contributes to the pathogenesis of adipogenesis in PKP2-deficient cells.


Assuntos
Adipogenia/fisiologia , Ilhas de CpG/fisiologia , Metilação de DNA/fisiologia , Fator de Transcrição E2F1/metabolismo , MicroRNAs/metabolismo , Placofilinas/deficiência , Animais , Células Cultivadas , Regulação para Baixo/fisiologia , Fator de Transcrição E2F1/antagonistas & inibidores , Camundongos , Camundongos Knockout , Miócitos Cardíacos/metabolismo , Placofilinas/genética , Transdução de Sinais/fisiologia
6.
J Cell Biol ; 212(4): 425-38, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26858265

RESUMO

Members of the desmosome protein family are integral components of the cardiac area composita, a mixed junctional complex responsible for electromechanical coupling between cardiomyocytes. In this study, we provide evidence that loss of the desmosomal armadillo protein Plakophilin-2 (PKP2) in cardiomyocytes elevates transforming growth factor ß1 (TGF-ß1) and p38 mitogen-activated protein kinase (MAPK) signaling, which together coordinate a transcriptional program that results in increased expression of profibrotic genes. Importantly, we demonstrate that expression of Desmoplakin (DP) is lost upon PKP2 knockdown and that restoration of DP expression rescues the activation of this TGF-ß1/p38 MAPK transcriptional cascade. Tissues from PKP2 heterozygous and DP conditional knockout mouse models also exhibit elevated TGF-ß1/p38 MAPK signaling and induction of fibrotic gene expression in vivo. These data therefore identify PKP2 and DP as central players in coordination of desmosome-dependent TGF-ß1/p38 MAPK signaling in cardiomyocytes, pathways known to play a role in different types of cardiac disease, such as arrhythmogenic or hypertrophic cardiomyopathy.


Assuntos
Cardiomiopatias/enzimologia , Miócitos Cardíacos/enzimologia , Placofilinas/deficiência , Fator de Crescimento Transformador beta1/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Animais Recém-Nascidos , Cardiomiopatias/genética , Cardiomiopatias/patologia , Linhagem Celular , Desmoplaquinas/genética , Desmoplaquinas/metabolismo , Ativação Enzimática , Fibrose , Regulação da Expressão Gênica , Genótipo , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Knockout , Miócitos Cardíacos/patologia , Fenótipo , Fosforilação , Placofilinas/genética , Estabilidade Proteica , Interferência de RNA , Ratos Sprague-Dawley , Transdução de Sinais , Fatores de Tempo , Transcrição Gênica , Transfecção
7.
Cardiovasc Res ; 107(4): 442-52, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26113266

RESUMO

AIMS: Progress in tissue preservation (high-pressure freezing), data acquisition (tomographic electron microscopy, TEM), and analysis (image segmentation and quantification) have greatly improved the level of information extracted from ultrastructural images. Here, we combined these methods and developed analytical tools to provide an in-depth morphometric description of the intercalated disc (ID) in adult murine ventricle. As a point of comparison, we characterized the ultrastructure of the ID in mice heterozygous-null for the desmosomal gene plakophilin-2 (PKP2; mice dubbed PKP2-Hz). METHODS AND RESULTS: Tomographic EM images of thin sections of adult mouse ventricular tissue were processed by image segmentation analysis. Novel morphometric routines allowed us to generate the first quantitative description of the ID intercellular space based on three-dimensional data. We show that complex invaginations of the cell membrane significantly increased the total ID surface area. In addition, PKP2-Hz samples showed increased average intercellular spacing, ID surface area, and membrane tortuosity, as well as reduced number and length of mechanical junctions compared with control. Finally, we observed membranous structures reminiscent of junctional sarcoplasmic reticulum at the ID, which were significantly more abundant in PKP2-Hz hearts. CONCLUSION: We have developed a systematic method to characterize the ultrastructure of the intercellular space in the adult murine ventricle and have provided a quantitative description of the structure of the intercellular membranes and of the intercellular space. We further show that PKP2 deficiency associates with ultrastructural defects. The possible importance of the intercellular space in cardiac behaviour is discussed.


Assuntos
Ventrículos do Coração/ultraestrutura , Miocárdio/ultraestrutura , Miócitos Cardíacos/ultraestrutura , Animais , Desmossomos/metabolismo , Desmossomos/ultraestrutura , Espaço Extracelular/metabolismo , Ventrículos do Coração/metabolismo , Espaço Intracelular/metabolismo , Camundongos , Microscopia Eletrônica/métodos , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Placofilinas/deficiência , Placofilinas/genética , Retículo Sarcoplasmático/ultraestrutura
8.
Cardiovasc Res ; 95(4): 460-8, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22764151

RESUMO

AIMS: The shRNA-mediated loss of expression of the desmosomal protein plakophilin-2 leads to sodium current (I(Na)) dysfunction. Whether pkp2 gene haploinsufficiency leads to I(Na) deficit in vivo remains undefined. Mutations in pkp2 are detected in arrhythmogenic right ventricular cardiomyopathy (ARVC). Ventricular fibrillation and sudden death often occur in the 'concealed phase' of the disease, prior to overt structural damage. The mechanisms responsible for these arrhythmias remain poorly understood. We sought to characterize the morphology, histology, and ultrastructural features of PKP2-heterozygous-null (PKP2-Hz) murine hearts and explore the relation between PKP2 abundance, I(Na) function, and cardiac electrical synchrony. METHODS AND RESULTS: Hearts of PKP2-Hz mice were characterized by multiple methods. We observed ultrastructural but not histological or gross anatomical differences in PKP2-Hz hearts compared with wild-type (WT) littermates. Yet, in myocytes, decreased amplitude and a shift in gating and kinetics of I(Na) were observed. To further unmask I(Na) deficiency, we exposed myocytes, Langendorff-perfused hearts, and anaesthetized animals to a pharmacological challenge (flecainide). In PKP2-Hz hearts, the extent of flecainide-induced I(Na) block, impaired ventricular conduction, and altered electrocardiographic parameters were larger than controls. Flecainide provoked ventricular arrhythmias and death in PKP2-Hz animals, but not in the WT. CONCLUSIONS: PKP2 haploinsufficiency leads to I(Na) deficit in murine hearts. Our data support the notion of a cross-talk between desmosome and sodium channel complex. They also suggest that I(Na) dysfunction may contribute to generation and/or maintenance of arrhythmias in PKP2-deficient hearts. Whether pharmacological challenges could help unveil arrhythmia risk in patients with mutations or variants in PKP2 remains undefined.


Assuntos
Displasia Arritmogênica Ventricular Direita/metabolismo , Haploinsuficiência , Miócitos Cardíacos/metabolismo , Placofilinas/deficiência , Canais de Sódio/metabolismo , Sódio/metabolismo , Potenciais de Ação , Animais , Antiarrítmicos/farmacologia , Displasia Arritmogênica Ventricular Direita/diagnóstico , Displasia Arritmogênica Ventricular Direita/genética , Displasia Arritmogênica Ventricular Direita/patologia , Displasia Arritmogênica Ventricular Direita/fisiopatologia , Modelos Animais de Doenças , Eletrocardiografia , Flecainida/farmacologia , Predisposição Genética para Doença , Ativação do Canal Iônico , Cinética , Camundongos , Camundongos Knockout , Miócitos Cardíacos/ultraestrutura , Perfusão , Fenótipo , Placofilinas/genética , Canais de Sódio/efeitos dos fármacos , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia
9.
PLoS One ; 7(6): e38561, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22701666

RESUMO

The desmosome anchors keratin filaments in epithelial cells leading to the formation of a tissue wide IF network. Loss of the desmosomal plaque protein plakophilin3 (PKP3) in HCT116 cells, leads to an increase in neoplastic progression and metastasis, which was accompanied by an increase in K8 levels. The increase in levels was due to an increase in the protein levels of the Phosphatase of Regenerating Liver 3 (PRL3), which results in a decrease in phosphorylation on K8. The increase in PRL3 and K8 protein levels could be reversed by introduction of an shRNA resistant PKP3 cDNA. Inhibition of K8 expression in the PKP3 knockdown clone S10, led to a decrease in cell migration and lamellipodia formation. Further, the K8 PKP3 double knockdown clones showed a decrease in colony formation in soft agar and decreased tumorigenesis and metastasis in nude mice. These results suggest that a stabilisation of K8 filaments leading to an increase in migration and transformation may be one mechanism by which PKP3 loss leads to tumor progression and metastasis.


Assuntos
Transformação Celular Neoplásica/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Proteínas Imediatamente Precoces/metabolismo , Queratina-8/metabolismo , Metástase Neoplásica/fisiopatologia , Neoplasias/metabolismo , Placofilinas/deficiência , Proteínas Tirosina Fosfatases/metabolismo , Animais , Western Blotting , Desmossomos/metabolismo , Eletroforese em Gel Bidimensional , Transferência Ressonante de Energia de Fluorescência , Técnicas de Silenciamento de Genes , Células HCT116 , Humanos , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Camundongos Nus , Microscopia Confocal , Microscopia de Fluorescência , Oligonucleotídeos/genética , Fosforilação , Placofilinas/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
J Cell Sci ; 125(Pt 4): 1058-67, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22421363

RESUMO

It is generally accepted that the intercalated disc (ICD) required for mechano-electrical coupling in the heart consists of three distinct junctional complexes: adherens junctions, desmosomes and gap junctions. However, recent morphological and molecular data indicate a mixing of adherens junctional and desmosomal components, resulting in a 'hybrid adhering junction' or 'area composita'. The α-catenin family member αT-catenin, part of the N-cadherin-catenin adhesion complex in the heart, is the only α-catenin that interacts with the desmosomal protein plakophilin-2 (PKP2). Thus, it has been postulated that αT-catenin might serve as a molecular integrator of the two adhesion complexes in the area composita. To investigate the role of αT-catenin in the heart, gene targeting technology was used to delete the Ctnna3 gene, encoding αT-catenin, in the mouse. The αT-catenin-null mice are viable and fertile; however, the animals exhibit progressive cardiomyopathy. Adherens junctional and desmosomal proteins were unaffected by loss of αT-catenin, with the exception of the desmosomal protein PKP2. Immunogold labeling at the ICD demonstrated in the αT-catenin-null heart a preferential reduction of PKP2 at the area composita compared with the desmosome. Furthermore, gap junction protein Cx43 was reduced at the ICD, including its colocalization with N-cadherin. Gap junction remodeling in αT-catenin-knockout hearts was associated with an increased incidence of ventricular arrhythmias after acute ischemia. This novel animal model demonstrates for the first time how perturbation in αT-catenin can affect both PKP2 and Cx43 and thereby highlights the importance of understanding the crosstalk between the junctional proteins of the ICD and its implications for arrhythmogenic cardiomyopathy.


Assuntos
Arritmias Cardíacas/fisiopatologia , Cardiomiopatia Dilatada/patologia , Junções Comunicantes/metabolismo , Ventrículos do Coração/fisiopatologia , Isquemia Miocárdica/complicações , Miócitos Cardíacos/metabolismo , alfa Catenina/deficiência , Junções Aderentes/metabolismo , Animais , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/patologia , Caderinas/metabolismo , Cardiomiopatia Dilatada/etiologia , Cardiomiopatia Dilatada/fisiopatologia , Conexina 43/deficiência , Conexina 43/metabolismo , Desmossomos/metabolismo , Modelos Animais de Doenças , Eletrocardiografia , Junções Comunicantes/patologia , Ventrículos do Coração/patologia , Camundongos , Camundongos Knockout , Mutação , Traumatismo por Reperfusão Miocárdica , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/patologia , Placofilinas/deficiência , Placofilinas/metabolismo , alfa Catenina/genética , alfa Catenina/metabolismo
11.
Heart Rhythm ; 8(11): 1740-8, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21723844

RESUMO

BACKGROUND: Loss of expression of the desmosomal protein plakophilin-2 (PKP2) leads to decreased gap junction-mediated (GJ) coupling, and alters the amplitude and kinetics of sodium current in cardiac myocytes. Whether these modifications, alone or in combination, are sufficient to act as arrhythmogenic substrates remains undefined. OBJECTIVE: This study sought to characterize arrhythmia susceptibility and reentry dynamics consequent to loss of PKP2 expression, and to assess the relative contribution of cell uncoupling versus alterations in sodium current in generation of reentry. METHODS: Monolayers of neonatal rat ventricular myocytes were treated with oligonucleotides that either prevented or failed to prevent PKP2 expression. Numerical simulations modeled experimentally observed modifications in I(Na), GJ coupling, or both (models PKP2-Na, PKP2-GJ, and PKP2-KD, respectively). Relative roles of sodium current density versus kinetics were further explored. RESULTS: Loss of PKP2 expression increased incidence of rotors and decreased frequency of rotation. Mathematical simulations revealed that single premature stimuli initiated rotors in models PKP2-Na and PKP2-KD, but not PKP2-GJ. Changes in sodium current kinetics, rather than current density, were key to reentry initiation. Anatomical barriers led to vortex shedding, wavebreaks, and rotors when I(Na) kinetics, but not GJ coupling or I(Na) density, were altered. CONCLUSION: PKP2-dependent changes in sodium current kinetics lead to slow conduction, increased propensity to functional block, and vortex shedding. Changes in GJ or I(Na) density played only a minor role on reentry susceptibility. Changes in electrical properties of the myocyte caused by loss of expression of PKP2 can set the stage for rotors even if anatomical homogeneity is maintained.


Assuntos
Potenciais de Ação/fisiologia , Arritmias Cardíacas/metabolismo , Junções Comunicantes/metabolismo , Líquido Intracelular/metabolismo , Miócitos Cardíacos/metabolismo , Placofilinas/deficiência , Sódio/metabolismo , Animais , Animais Recém-Nascidos , Arritmias Cardíacas/patologia , Arritmias Cardíacas/fisiopatologia , Células Cultivadas , Simulação por Computador , Modelos Animais de Doenças , Miócitos Cardíacos/patologia , Placofilinas/biossíntese , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA