Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
1.
Biomolecules ; 13(12)2023 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-38136565

RESUMO

Calsequestrin (CASQ) is a key intra-sarcoplasmic reticulum Ca2+-handling protein that plays a pivotal role in the contraction of cardiac and skeletal muscles. Its Ca2+-dependent polymerization dynamics shape the translation of electric excitation signals to the Ca2+-induced contraction of the actin-myosin architecture. Mutations in CASQ are linked to life-threatening pathological conditions, including tubular aggregate myopathy, malignant hyperthermia, and Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT). The variability in the penetrance of these phenotypes and the lack of a clear understanding of the disease mechanisms associated with CASQ mutations pose a major challenge to the development of effective therapeutic strategies. In vitro studies have mainly focused on the polymerization and Ca2+-buffering properties of CASQ but have provided little insight into the complex interplay of structural and functional changes that underlie disease. In this review, the biochemical and structural natures of CASQ are explored in-depth, while emphasizing their direct and indirect consequences for muscle Ca2+ physiology. We propose a novel functional classification of CASQ pathological missense mutations based on the structural stability of the monomer, dimer, or linear polymer conformation. We also highlight emerging similarities between polymeric CASQ and polyelectrolyte systems, emphasizing the potential for the use of this paradigm to guide further research.


Assuntos
Calsequestrina , Taquicardia Ventricular , Humanos , Calsequestrina/genética , Calsequestrina/metabolismo , Coração , Taquicardia Ventricular/genética , Taquicardia Ventricular/metabolismo , Retículo Sarcoplasmático/metabolismo , Mutação de Sentido Incorreto , Cálcio/metabolismo
2.
Cells ; 11(16)2022 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-36010545

RESUMO

Calsequestrin 1 (CASQ1) and Ryanodine receptor 1 (RYR1) are two of the main players in excitation-contraction (EC) coupling. CASQ1-knockout mice and mice carrying a mutation in RYR1 (Y522S) linked to human malignant hyperthermia susceptibility (MHS) both suffer lethal hypermetabolic episodes when exposed to halothane (MHS crises) and to environmental heat (heat stroke, HS). The phenotype of Y522S is more severe than that of CASQ1-null mice. As MHS and HS are hypermetabolic responses, we studied the metabolism of adult CASQ1-null and Y522S mice using wild-type (WT) mice as controls. We found that CASQ1-null and Y522S mice have increased food consumption and higher core temperature at rest. By indirect calorimetry, we then verified that CASQ1-null and Y522S mice show an increased oxygen consumption and a lower respiratory quotient (RQ). The accelerated metabolism of CASQ1-null and Y522S mice was also accompanied with a reduction in body fat. Moreover, both mouse models displayed increased oxygen consumption and a higher core temperature during heat stress. The results collected suggest that metabolic rate, oxygen consumption, and body temperature at rest, all more elevated in Y522S than in CASQ1-null mice, could possibly be used as predictors of the level of susceptibility to hyperthermic crises of mice (and possibly humans).


Assuntos
Golpe de Calor , Hipertermia Maligna , Animais , Metabolismo Basal , Proteínas de Ligação ao Cálcio/metabolismo , Calsequestrina/genética , Calsequestrina/metabolismo , Golpe de Calor/genética , Humanos , Hipertermia Maligna/genética , Hipertermia Maligna/metabolismo , Camundongos , Camundongos Knockout , Consumo de Oxigênio , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
3.
Bull Exp Biol Med ; 172(2): 117-120, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34855078

RESUMO

We compared the expression of Са2+-ATPase (SERCA2a), calsequestrin (CASQ2), ryanodine receptors (RyR2) proteins and their genes (ATP2A2, CASQ2, and RYR2) in coronary heart disease (CHD) patients with and without comorbid type 2 diabetes mellitus. All studies were performed on the right atrial appendages resected during coronary bypass surgeries. Expression of SERCA2a and RyR2 proteins and their ATP2A2 (p=0.046) and RYR2 genes in comorbid pathology was significantly (p=0.042) higher (by 1.2 and 2 times; p=0.025). The expression of CASQ2 protein and its gene did not differ significantly between the groups (p=0.82 and p=0.066, respectively). It was concluded that the expression of SERCA2a and RyR2 proteins and their genes (but not CASQ2 and its gene) is elevated in CHD associated with type 2 diabetes mellitus. Expression of the studied proteins correlated with the expression of their genes. Increased expression of CASQ2 protein and its gene can probably prevent imbalance of the Ca2+-transporting systems in cardiomyocytes and contractile dysfunction of the myocardium, even in CHD associated with type 2 diabetes mellitus.


Assuntos
Sinalização do Cálcio/genética , Doença das Coronárias , Diabetes Mellitus Tipo 2 , Miócitos Cardíacos/metabolismo , Retículo Sarcoplasmático/metabolismo , Idoso , Transporte Biológico/genética , Biópsia , Cálcio/metabolismo , Calsequestrina/genética , Calsequestrina/metabolismo , Estudos de Casos e Controles , Doença das Coronárias/complicações , Doença das Coronárias/genética , Doença das Coronárias/metabolismo , Doença das Coronárias/patologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Expressão Gênica , Humanos , Pessoa de Meia-Idade , Miocárdio/metabolismo , Miócitos Cardíacos/patologia , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/patologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo
4.
Mol Ther ; 28(1): 171-179, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31607542

RESUMO

Recessive forms of catecholaminergic polymorphic ventricular tachycardia (CPVT) are induced by mutations in genes encoding triadin or calsequestrin, two proteins that belong to the Ca2+ release complex, responsible for intracellular Ca2+ release triggering cardiac contractions. To better understand the mechanisms of triadin-induced CPVT and to assay multiple therapeutic interventions, we used a triadin knockout mouse model presenting a CPVT-like phenotype associated with a decrease in calsequestrin protein level. We assessed different approaches to rescue protein expression and to correct intracellular Ca2+ release and cardiac function: pharmacological treatment with kifunensine or a viral gene transfer-based approach, using adeno-associated virus serotype 2/9 (AAV2/9) encoding the triadin or calsequestrin. We observed that the levels of triadin and calsequestrin are intimately linked, and that reduction of both proteins contributes to the CPVT phenotype. Different combinations of triadin and calsequestrin expression level were obtained using these therapeutic approaches. A full expression of each is not necessary to correct the phenotype; a fine-tuning of the relative re-expression of both triadin and calsequestrin is required to correct the CPVT phenotype and rescue the cardiac function. AAV-mediated gene delivery of calsequestrin or triadin and treatment with kifunensine are potential treatments for recessive forms of CPVT due to triadin mutations.


Assuntos
Calsequestrina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Musculares/metabolismo , Taquicardia Ventricular/metabolismo , Alcaloides/uso terapêutico , Animais , Arritmias Cardíacas/tratamento farmacológico , Cálcio/metabolismo , Sinalização do Cálcio/genética , Calsequestrina/genética , Dependovirus , Modelos Animais de Doenças , Terapia Genética/métodos , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Camundongos , Camundongos Knockout , Proteínas Musculares/genética , Contração Miocárdica/efeitos dos fármacos , Contração Miocárdica/genética , Miócitos Cardíacos/metabolismo , Parvovirinae/genética , Fenótipo , Ratos , Taquicardia Ventricular/tratamento farmacológico , Taquicardia Ventricular/patologia , Transdução Genética , Transfecção
5.
Poult Sci ; 98(11): 5465-5476, 2019 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-31250001

RESUMO

During recent years, research on meat quality in poultry has aimed to evaluate the presence and consequences of breast myopathies as well as the factors which can affect their occurrence by modifying the growth rate. A total of 900 broiler chickens were reared until slaughter (48 D) to evaluate the effect of 2 genetic lines (A vs. B) and feeding plans (ad libitum [AL], early restricted [ER], from 13 to 23 D of age, and late restricted [LR], from 27 to 37 D of age; restriction rate: 80%) on performance, meat quality, and breast muscle myopathies. Calsequestrin and vascular endothelial growth factor (VEGF) expressions, and muscle fiber degeneration (MFD) were recorded at 22, 36, and 48 D. Chickens in the AL treatment had greater final live (P < 0.01) and carcass weights and proportion of pectoralis major muscle (P = 0.04) compared to chickens in the LR treatment, whereas chickens in the ER treatment had intermediate final live (3,454 g) and carcass weights, and proportion of pectoralis major muscle (25.6%). Chickens of line A were heavier than chickens of line B (P < 0.001), and had a greater feed conversion rate. Chickens of line A also had a greater dressing out percentage (P < 0.001), but a lower proportion of pectoralis major muscle (P = 0.04), as well as a greater meat pH (P < 0.001), meat cooking losses (P < 0.01), and shear force of the pectoralis major muscle (P = 0.03). Calsequestrin and VEGF mRNA were significantly lower in ER and LR chickens compared to AL chickens after feed restriction and during refeeding (P < 0.05). MFD scores increased with chicken age (P < 0.001) and differed between genetic lines (P < 0.001). Neither feeding plan nor genetic line affected the occurrence of white striping or wooden breast condition.


Assuntos
Proteínas Aviárias/genética , Galinhas/fisiologia , Dieta/veterinária , Expressão Gênica , Doenças Musculares/veterinária , Doenças das Aves Domésticas/epidemiologia , Fatores Etários , Ração Animal/análise , Animais , Proteínas Aviárias/metabolismo , Calsequestrina/genética , Calsequestrina/metabolismo , Galinhas/genética , Galinhas/crescimento & desenvolvimento , Masculino , Doenças Musculares/epidemiologia , Doenças Musculares/etiologia , Doenças Musculares/genética , Músculos Peitorais/patologia , Doenças das Aves Domésticas/etiologia , Doenças das Aves Domésticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Distribuição Aleatória , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
Mol Cell Biochem ; 457(1-2): 201-214, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30919218

RESUMO

Adiponectin (ADN) is an abundant protein in serum, secreted by adipocytes, that acts as a signal for fat metabolism. It is marked by a complex molecular structure that results from processes within the secretory pathway, producing a canonical set of multimers. ADN may also be secreted from cardiomyocytes, where a unique sarcomeric endoplasmic/sarcoplasmic reticulum (ER/SR) substructure has been characterized primarily for its Ca handling. We expressed ADN in cultured primary adult cardiomyocytes and nonmuscle (COS) cells. After 48 h of ADN expression by adenovirus treatment, roughly half of synthesized ADN was secreted from cardiomyocytes, and half was still in-transit within inner membrane compartments, similar to COS cells. Cardiomyocytes and COS cells both produced ADN in the three canonical forms: trimers, hexamers, and 18-mers. Higher rates of secretion occurred for higher-molecular weight multimers, especially 18-mers. The highest levels of ADN protein, whether in transit or secreted, were present as trimers and hexamers. In nonmuscle cell lines, ADN trafficked through ER and Golgi compartments as expected. In contrast, ADN in primary adult cardiomyocytes populated ER/SR tubules along the edges of sarcomeres that emanated from nuclear surfaces. Prominent co-localization of ADN occurred with calsequestrin, a marker of junctional SR, the Ca2+-release compartment of the cell. The early steps in ADN trafficking re-trace those recently described for newly made junctional SR proteins, involving a nuclear envelope (NE) translocation into SR tubules that are oriented along sarcolemmal transverse (T)-tubules (NEST pathway).


Assuntos
Adiponectina/metabolismo , Calsequestrina/metabolismo , Miócitos Cardíacos/metabolismo , Multimerização Proteica , Retículo Sarcoplasmático/metabolismo , Animais , Células COS , Chlorocebus aethiops , Células HEK293 , Humanos , Transporte Proteico , Ratos , Ratos Sprague-Dawley
7.
Europace ; 21(6): 981-989, 2019 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-30753421

RESUMO

AIMS: Action potential duration (APD) alternans is an established precursor or arrhythmia and sudden cardiac death. Important differences in fundamental electrophysiological properties relevant to arrhythmia exist between experimental models and the diseased in vivo human heart. To investigate mechanisms of APD alternans using a novel approach combining intact heart and cellular cardiac electrophysiology in human in vivo. METHODS AND RESULTS: We developed a novel approach combining intact heart electrophysiological mapping during cardiac surgery with rapid on-site data analysis to guide myocardial biopsies for laboratory analysis, thereby linking repolarization dynamics observed at the organ level with underlying ion channel expression. Alternans-susceptible and alternans-resistant regions were identified by an incremental pacing protocol. Biopsies from these sites (n = 13) demonstrated greater RNA expression in Calsequestrin (CSQN) and Ryanodine (RyR) and ion channels underlying IK1 and Ito at alternans-susceptible sites. Electrical restitution properties (n = 7) showed no difference between alternans-susceptible and resistant sites, whereas spatial gradients of repolarization were greater in alternans-susceptible than in alternans-resistant sites (P = 0.001). The degree of histological fibrosis between alternans-susceptible and resistant sites was equivalent. Mathematical modelling of these changes indicated that both CSQN and RyR up-regulation are key determinants of APD alternans. CONCLUSION: Combined intact heart and cellular electrophysiology show that regions of myocardium in the in vivo human heart exhibiting APD alternans are associated with greater expression of CSQN and RyR and show no difference in restitution properties compared to non-alternans regions. In silico modelling identifies up-regulation and interaction of CSQN with RyR as a major mechanism underlying APD alternans.


Assuntos
Arritmias Cardíacas/fisiopatologia , Técnicas Eletrofisiológicas Cardíacas , Sistema de Condução Cardíaco/fisiopatologia , Potenciais de Ação , Biópsia , Calsequestrina/metabolismo , Feminino , Humanos , Canais Iônicos/metabolismo , Masculino , Pessoa de Meia-Idade , Rianodina/metabolismo
8.
J Gene Med ; 20(12): e3060, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30393908

RESUMO

BACKGROUND: Cardiac gene therapy using the adeno-associated virus serotype 9 vector is widely used because of its efficient transduction. However, the promoters used to drive expression often cause off-target localization. To overcome this, studies have applied cardiac-specific promoters, although expression is debilitated compared to that of ubiquitous promoters. To address these issues in the context of atrial-specific gene expression, an enhancer calsequestrin cis-regulatory module 4 (CRM4) and the highly atrial-specific promoter sarcolipin were combined to enhance expression and minimize off tissue expression. METHODS: To observe expression and bio-distribution, constructs were generated using two different reporter genes: luciferase and enhanced green fluorescent protein (EGFP). The ubiquitous cytomegalovirus (CMV), sarcolipin (SLN) and CRM4 combined with sarcolipin (CRM4.SLN) were compared and analyzed using the luciferase assay, western blotting, a quantitative polymerase chain reaction and fluorescence imaging. RESULTS: The CMV promoter containing vectors showed the strongest expression in vitro and in vivo. However, the module SLN combination showed enhanced atrial expression and a minimized off-target effect even when compared with the individual SLN promoter. CONCLUSIONS: For gene therapy involving atrial gene transfer, the CRM4.SLN combination is a promising alternative to the use of the CMV promoter. CRM4.SLN had significant atrial expression and minimized extra-atrial expression.


Assuntos
Calsequestrina/genética , Regulação da Expressão Gênica , Átrios do Coração/metabolismo , Proteínas Musculares/genética , Regiões Promotoras Genéticas/genética , Proteolipídeos/genética , Animais , Calsequestrina/metabolismo , Citomegalovirus/genética , Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/terapia , Humanos , Luciferases/genética , Luciferases/metabolismo , Camundongos , Proteínas Musculares/metabolismo , Proteolipídeos/metabolismo , Transfecção
9.
Biochem Biophys Res Commun ; 506(1): 41-47, 2018 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-30336983

RESUMO

OBJECTIVE: Glucocorticoids (GCs)-induced osteoblast apoptosis has been identified as an important cause of GCs related osteonecrosis of the femoral head (ONFH). Glycogen synthase kinase 3ß (GSK3ß) has been proved to mediate dexamethasone (Dex)-induced osteoblast apoptosis. This study aimed to investigate the underlying mechanism of GSK3ß in Dex-induced osteoblast apoptosis. METHODS: Osteoblast cells were transfected with lentivirus expressing GSK3ß-shRNA, and a DNA microarray was performed to analyze gene expression after Dex treatment with or without GSK3ß-shRNA. Some differentially expressed genes were further validated by quantitative real-time-PCR (qRT-PCR). RESULTS: 460 genes were up-regulated (at least 2-fold) with Dex treatment but down-regulated (at least 2-fold) with GSK3ß-shRNA treatment. In addition, 315 genes were down-regulated (at least 2-fold) with Dex treatment but up-regulated (at least 2-fold) with GSK3ß-shRNA treatment. Among these genes, the apoptosis-related genes Hoxb8, Kif18a, Dock8, Dlk1, Tnfsf14, Casq2, Bcl2l14 and mechanosensation-related gene Piezo2 were selected for further qRT-PCR analysis. 7 of 8 genes (Piezo2, Hoxb8, Kif18a, Dlk1, Tnfsf14, Casq2, Bcl2l14) showed the same tendency between gene chip results and qRT-PCR results. The microarray data also showed that apoptotic pathway, MAPK pathway, TGFß pathway and Wnt pathway might be related to the mechanism of GSK3ß in Dex-induced osteoblast apoptosis. CONCLUSION: Our findings indicate that GSK3ß-shRNA treatment can alter various genes expression levels and change diverse signaling pathways involved in Dex-induced osteoblast apoptosis. Furthermore, Piezo2, Hoxb8, Kif18a, Dlk1, Tnfsf14, Casq2 and Bcl2l14 genes may play an important role in the GSK3ß-mediated osteoblast apoptosis process.


Assuntos
Apoptose/efeitos dos fármacos , Dexametasona/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta/genética , Osteoblastos/efeitos dos fármacos , Animais , Apoptose/genética , Proteínas de Ligação ao Cálcio , Calsequestrina/genética , Calsequestrina/metabolismo , Linhagem Celular , Perfilação da Expressão Gênica , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Canais Iônicos/genética , Canais Iônicos/metabolismo , Cinesinas/genética , Cinesinas/metabolismo , Mecanotransdução Celular , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Osteoblastos/citologia , Osteoblastos/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo
10.
J Appl Physiol (1985) ; 124(3): 684-695, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29122966

RESUMO

While cancer-induced skeletal muscle wasting has been widely investigated, the drivers of cancer-induced muscle functional decrements are only beginning to be understood. Decreased muscle function impacts cancer patient quality of life and health status, and several potential therapeutics have failed in clinical trials due to a lack of functional improvement. Furthermore, systemic inflammation and intrinsic inflammatory signaling's role in the cachectic disruption of muscle function requires further investigation. We examined skeletal muscle functional properties during cancer cachexia and determined their relationship to systemic and intrinsic cachexia indices. Male ApcMin/+ (MIN) mice were stratified by percent body weight loss into weight stable (WS; <5% loss) or cachectic (CX; >5% loss). Age-matched C57BL/6 littermates served as controls. Tibialis anterior (TA) twitch properties, tetanic force, and fatigability were examined in situ. TA protein and mRNA expression were examined in the nonstimulated leg. CX decreased muscle mass, tetanic force (Po), and specific tetanic force (sPo). Whole body and muscle fatigability were increased in WS and CX. CX had slower contraction rates, +dP/d t and -dP/d t, which were inversely associated with muscle signal transducer and activator of transcription 3 ( STAT3) and p65 activation. STAT3 and p65 activation were also inversely associated with Po. However, STAT3 was not related to sPo or fatigue. Muscle suppressor of cytokine signaling 3 mRNA expression was negatively associated with TA weight, Po, and sPo but not fatigue. Our study demonstrates that multiple functional deficits that occur with cancer cachexia are associated with increased muscle inflammatory signaling. Notably, muscle fatigability is increased in the MIN mouse before cachexia development. NEW & NOTEWORTHY Recent studies have identified decrements in skeletal muscle function during cachexia. We have extended these studies by directly relating decrements in muscle function to established cachexia indices. Our results demonstrate that a slow-fatigable contractile phenotype is developed during the progression of cachexia that coincides with increased muscle inflammatory signaling. Furthermore, regression analysis identified predictors of cancer-induced muscle dysfunction. Last, we report the novel finding that whole body and muscle fatigability were increased before cachexia development.


Assuntos
Caquexia/fisiopatologia , Músculo Esquelético/fisiopatologia , Animais , Caquexia/etiologia , Caquexia/metabolismo , Calsequestrina/metabolismo , Progressão da Doença , Masculino , Camundongos , Contração Muscular , Fadiga Muscular , Força Muscular , Músculo Esquelético/metabolismo , Neoplasias/complicações , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo
11.
Cell Physiol Biochem ; 44(3): 1199-1212, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29179218

RESUMO

BACKGROUND/AIMS: The embryonic stem cell-derived cardiomyocytes (ES-CMs) serve as potential sources for cardiac regenerative therapy. However, the immature sarcoplasmic reticulum (SR) function of ES-CMs prevents its application. In this report, we examined the effect of puerarin, an isoflavone compound, on SR function of murine ES-CMs. METHODS: Murine ES-CMs were harvested by embryoid body-based differentiation method. Confocal calcium imaging and whole-cell patch clamps were performed to assess the function of SR. The mRNA expression levels of SR-related genes were examined by quantitative PCR. The protein expression of sarcoplasmic reticulum calcium-ATPase 2a (SERCA2a) was evaluated by immunofluorescent and western blot. RESULTS: Long-term application of puerarin promotes basic properties of spontaneous calcium transient with increased amplitude, decay velocity, and decreased duration. Puerarin fails to alter ICa,L but increases the Ca2+ content of SR. Puerarin-treated ES-CMs have intact SR Ca2+ cycling with more SR Ca2+ reuptake. Long-term application of puerarin asynchronously upregulates the mRNA and protein expression of SERCA2a, as well as the transcripts of calsequestrin and triadin in developing ES-CMs. Application of puerarin during the stage of post-cardiac differentiation upregulates dose-dependently the transcripts of SERCA2a, phospholamban and tridin which can be reversed by the inhibitors of the PI3K/Akt and MAPK/ERK signaling pathways, but shows no effect on the protein expression of SERCA2a. CONCLUSION: This study demonstrates that long-term puerarin treatment enhances Ca2+ reuptake and Ca2+ content via upregulation of SERCA2a.


Assuntos
Cálcio/metabolismo , Isoflavonas/farmacologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Regulação para Cima/efeitos dos fármacos , Vasodilatadores/farmacologia , Potenciais de Ação/efeitos dos fármacos , Androstadienos/farmacologia , Animais , Benzamidas/farmacologia , Proteínas de Ligação ao Cálcio/metabolismo , Calsequestrina/genética , Calsequestrina/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Diferenciação Celular/efeitos dos fármacos , Difenilamina/análogos & derivados , Difenilamina/farmacologia , Camundongos , Microscopia Confocal , Células-Tronco Embrionárias Murinas/citologia , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/metabolismo , Retículo Sarcoplasmático/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/antagonistas & inibidores , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Transdução de Sinais/efeitos dos fármacos , Tapsigargina/farmacologia , Wortmanina
12.
Oxid Med Cell Longev ; 2017: 6936897, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29062464

RESUMO

Oxidative stress has been proposed to play a key role in malignant hyperthermia (MH), a syndrome caused by excessive Ca2+ release in skeletal muscle. Incidence of mortality in male calsequestrin-1 knockout (CASQ1-null) mice during exposure to halothane and heat (a syndrome closely resembling human MH) is far greater than that in females. To investigate the possible role of sex hormones in this still unexplained gender difference, we treated male and female CASQ1-null mice for 1 month, respectively, with Premarin (conjugated estrogens) and leuprolide (GnRH analog) and discovered that during exposure to halothane and heat Premarin reduced the mortality rate in males (79-27% and 86-20%), while leuprolide increased the incidence of mortality in females (18-73% and 24-82%). We then evaluated the (a) responsiveness of isolated muscles to temperature and caffeine, (b) sarcoplasmic reticulum (SR) Ca2+ release in single fibers, and (c) oxidative stress and the expression levels of main enzymes involved in the regulation of the redox balance in muscle. Premarin treatment reduced the temperature and caffeine sensitivity of EDL muscles, normalized SR Ca2+ release, and reduced oxidative stress in males, suggesting that female sex hormones may protect mice from lethal hyperthermic episodes by reducing both the SR Ca2+ leak and oxidative stress.


Assuntos
Calsequestrina/genética , Estrogênios/uso terapêutico , Hipertermia Maligna/tratamento farmacológico , Músculo Esquelético/metabolismo , Animais , Calsequestrina/metabolismo , Estrogênios/farmacologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Estresse Oxidativo
13.
Artigo em Inglês | MEDLINE | ID: mdl-28630169

RESUMO

BACKGROUND: Catecholaminergic polymorphic ventricular tachycardia type 2 (CPVT2) results from autosomal recessive CASQ2 mutations, causing abnormal Ca2+-handling and malignant ventricular arrhythmias. We aimed to establish a patient-specific human induced pluripotent stem cell (hiPSC) model of CPVT2 and to use the generated hiPSC-derived cardiomyocytes to gain insights into patient-specific disease mechanism and pharmacotherapy. METHODS AND RESULTS: hiPSC cardiomyocytes were derived from a CPVT2 patient (D307H-CASQ2 mutation) and from healthy controls. Laser-confocal Ca2+ and voltage imaging showed significant Ca2+-transient irregularities, marked arrhythmogenicity manifested by early afterdepolarizations and triggered arrhythmias, and reduced threshold for store overload-induced Ca2+-release events in the CPVT2-hiPSC cardiomyocytes when compared with healthy control cells. Pharmacological studies revealed the prevention of adrenergic-induced arrhythmias by ß-blockers (propranolol and carvedilol), flecainide, and the neuronal sodium-channel blocker riluzole; a direct antiarrhythmic action of carvedilol (independent of its α/ß-adrenergic blocking activity), flecainide, and riluzole; and suppression of abnormal Ca2+ cycling by the ryanodine stabilizer JTV-519 and carvedilol. Mechanistic insights were gained on the different antiarrhythmic actions of the aforementioned drugs, with carvedilol and JTV-519 (but not flecainide or riluzole) acting primarily through sarcoplasmic reticulum stabilization. Finally, comparable outcomes were found between flecainide and labetalol antiarrhythmic effects in vitro and the clinical results in the same patient. CONCLUSIONS: These results demonstrate the ability of hiPSCs cardiomyocytes to recapitulate CPVT2 disease phenotype and drug response in the culture dish, to provide novel insights into disease and drug therapy mechanisms, and potentially to tailor patient-specific drug therapy.


Assuntos
Antiarrítmicos/farmacologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Medicina de Precisão , Taquicardia Ventricular/tratamento farmacológico , Potenciais de Ação , Agonistas Adrenérgicos/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Calsequestrina/genética , Calsequestrina/metabolismo , Estudos de Casos e Controles , Linhagem Celular , Relação Dose-Resposta a Droga , Predisposição Genética para Doença , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Mutação , Miócitos Cardíacos/metabolismo , Seleção de Pacientes , Fenótipo , Retículo Sarcoplasmático/efeitos dos fármacos , Retículo Sarcoplasmático/metabolismo , Taquicardia Ventricular/genética , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/fisiopatologia , Fatores de Tempo , Adulto Jovem
14.
J Mol Cell Cardiol ; 102: 45-52, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27908661

RESUMO

Dystrophin deficiency results in Duchenne cardiomyopathy, a primary cause of death in Duchenne muscular dystrophy (DMD). Gene therapy has shown great promise in ameliorating the cardiac phenotype in mouse models of DMD. However, it is not completely clear how much dystrophin is required to treat dystrophic heart disease. We and others have shown that mosaic dystrophin expression at the wild-type level, depending on the percentage of dystrophin positive cardiomyocytes, can either delay the onset of or fully prevent cardiomyopathy in dystrophin-null mdx mice. Many gene therapy strategies will unlikely restore dystrophin to the wild-type level in a cardiomyocyte. To determine whether low-level dystrophin expression can reduce the cardiac manifestations in DMD, we examined heart histology, ECG and hemodynamics in 21-m-old normal BL6 and two strains of BL6-background dystrophin-deficient mice. Mdx3cv mice show uniform low-level expression of a near full-length dystrophin protein in every myofiber while mdx4cv mice have no dystrophin expression. Immunostaining and western blot confirmed marginal level dystrophin expression in the heart of mdx3cv mice. Although low-level expression did not reduce myocardial histopathology, it significantly ameliorated QRS prolongation and normalized diastolic hemodynamic deficiencies. Our study demonstrates for the first time that low-level dystrophin can partially preserve heart function.


Assuntos
Cardiomiopatias/etiologia , Cardiomiopatias/fisiopatologia , Distrofina/genética , Expressão Gênica , Distrofia Muscular de Duchenne/complicações , Miocárdio/metabolismo , Fatores Etários , Animais , Biomarcadores , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Calsequestrina/genética , Calsequestrina/metabolismo , Cardiomiopatias/diagnóstico , Diástole , Modelos Animais de Doenças , Distrofina/metabolismo , Eletrocardiografia , Camundongos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Miocárdio/patologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Função Ventricular
15.
PLoS One ; 11(10): e0164795, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27764147

RESUMO

INTRODUCTION: Human induced pluripotent stem cells (hiPSCs) offer a unique opportunity for disease modeling. However, it is not invariably successful to recapitulate the disease phenotype because of the immaturity of hiPSC-derived cardiomyocytes (hiPSC-CMs). The purpose of this study was to establish and analyze iPSC-based model of catecholaminergic polymorphic ventricular tachycardia (CPVT), which is characterized by adrenergically mediated lethal arrhythmias, more precisely using electrical pacing that could promote the development of new pharmacotherapies. METHOD AND RESULTS: We generated hiPSCs from a 37-year-old CPVT patient and differentiated them into cardiomyocytes. Under spontaneous beating conditions, no significant difference was found in the timing irregularity of spontaneous Ca2+ transients between control- and CPVT-hiPSC-CMs. Using Ca2+ imaging at 1 Hz electrical field stimulation, isoproterenol induced an abnormal diastolic Ca2+ increase more frequently in CPVT- than in control-hiPSC-CMs (control 12% vs. CPVT 43%, p<0.05). Action potential recordings of spontaneous beating hiPSC-CMs revealed no significant difference in the frequency of delayed afterdepolarizations (DADs) between control and CPVT cells. After isoproterenol application with pacing at 1 Hz, 87.5% of CPVT-hiPSC-CMs developed DADs, compared to 30% of control-hiPSC-CMs (p<0.05). Pre-incubation with 10 µM S107, which stabilizes the closed state of the ryanodine receptor 2, significantly decreased the percentage of CPVT-hiPSC-CMs presenting DADs to 25% (p<0.05). CONCLUSIONS: We recapitulated the electrophysiological features of CPVT-derived hiPSC-CMs using electrical pacing. The development of DADs in the presence of isoproterenol was significantly suppressed by S107. Our model provides a promising platform to study disease mechanisms and screen drugs.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Estimulação Elétrica , Modelos Biológicos , Taquicardia Ventricular/patologia , Taquicardia Ventricular/terapia , Tiazepinas/farmacologia , Adulto , Animais , Antiasmáticos/química , Antiasmáticos/farmacologia , Antiasmáticos/uso terapêutico , Cálcio/metabolismo , Calreticulina/genética , Calreticulina/metabolismo , Calsequestrina/genética , Calsequestrina/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Isoproterenol/farmacologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/transplante , Rianodina/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/química , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Taquicardia Ventricular/tratamento farmacológico , Tiazepinas/química , Tiazepinas/uso terapêutico , Transplante Heterólogo
16.
Sci Rep ; 5: 11349, 2015 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-26087026

RESUMO

Interaction between the endoplasmic reticulum (ER)-located stromal interaction molecue1 (STIM1) and the plasma membrane-located Ca(2+) channel subunit, Orai1, underlies store-operated Ca(2+) entry (SOCE). Calsequestrin1 (CSQ1), a sarcoplasmic reticulum Ca(2+) buffering protein, inhibits SOCE, but the mechanism of action is unknown. We identified an interaction between CSQ1 and STIM1 in HEK293 cells. An increase in monomeric CSQ1 induced by depleted Ca(2+) stores, or trifluoperazine (TFP), a blocker of CSQ folding and aggregation, enhanced the CSQ1-STIM1 interaction. In cells with Ca(2+) stores depleted, TFP further increased CSQ1 monomerization and CSQ1-STIM1 interaction, but reduced the association of STIM1 with Orai1 and SOCE. Over-expression of CSQ1 or a C-terminal (amino acid 388-396) deletion mutant significantly promoted the association of CSQ1 with STIM1, but suppressed both STIM1-Orai1 interaction and SOCE, while over-expression of the C-terminal (amino acid 362-396) deletion mutant had no effect. The physical interaction between low polymeric forms of CSQ1 and STIM1 likely acts by interfering with STIM1 oligimerization and inhibits STIM1-Orai1 interaction, providing a brake to SOCE under physiological conditions. This novel regulatory mechanism for SOCE may also contribute to the pathological Ca(2+) overload in calsequestrin deficient diseases, such as malignant hyperthermia and ventricular tachycardia.


Assuntos
Canais de Cálcio/metabolismo , Cálcio/metabolismo , Calsequestrina/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Canais de Cálcio/química , Canais de Cálcio/genética , Retículo Endoplasmático/metabolismo , Expressão Gênica , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Proteína ORAI1 , Ligação Proteica/efeitos dos fármacos , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Molécula 1 de Interação Estromal
17.
J Gen Physiol ; 145(6): 459-74, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26009541

RESUMO

A novel category of diseases of striated muscle is proposed, the couplonopathies, as those that affect components of the couplon and thereby alter its operation. Couplons are the functional units of intracellular calcium release in excitation-contraction coupling. They comprise dihydropyridine receptors, ryanodine receptors (Ca2+ release channels), and a growing list of ancillary proteins whose alteration may lead to disease. Within a generally similar plan, the couplons of skeletal and cardiac muscle show, in a few places, marked structural divergence associated with critical differences in the mechanisms whereby they fulfill their signaling role. Most important among these are the presence of a mechanical or allosteric communication between voltage sensors and Ca2+ release channels, exclusive to the skeletal couplon, and the smaller capacity of the Ca stores in cardiac muscle, which results in greater swings of store concentration during physiological function. Consideration of these structural and functional differences affords insights into the pathogenesis of several couplonopathies. The exclusive mechanical connection of the skeletal couplon explains differences in pathogenesis between malignant hyperthermia (MH) and catecholaminergic polymorphic ventricular tachycardia (CPVT), conditions most commonly caused by mutations in homologous regions of the skeletal and cardiac Ca(2+) release channels. Based on mechanistic considerations applicable to both couplons, we identify the plasmalemma as a site of secondary modifications, typically an increase in store-operated calcium entry, that are relevant in MH pathogenesis. Similar considerations help explain the different consequences that mutations in triadin and calsequestrin have in these two tissues. As more information is gathered on the composition of cardiac and skeletal couplons, this comparative and mechanistic approach to couplonopathies should be useful to understand pathogenesis, clarify diagnosis, and propose tissue-specific drug development.


Assuntos
Cardiomiopatias/metabolismo , Acoplamento Excitação-Contração , Músculo Esquelético/metabolismo , Doenças Musculares/metabolismo , Contração Miocárdica , Miocárdio/metabolismo , Animais , Canais de Cálcio Tipo L/metabolismo , Calsequestrina/genética , Calsequestrina/metabolismo , Cardiomiopatias/genética , Cardiomiopatias/fisiopatologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Predisposição Genética para Doença , Humanos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/fisiopatologia , Doenças Musculares/genética , Doenças Musculares/fisiopatologia , Mutação , Fenótipo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
18.
J Appl Physiol (1985) ; 118(8): 1050-8, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25701006

RESUMO

Sarcolipin (SLN) is a regulator of sarcoendoplasmic reticulum calcium ATPase in skeletal muscle. Recent studies using SLN-null mice have identified SLN as a key player in muscle thermogenesis and metabolism. In this study, we exploited a SLN overexpression (Sln(OE)) mouse model to determine whether increased SLN level affected muscle contractile properties, exercise capacity/fatigue, and metabolic rate in whole animals and isolated muscle. We found that Sln(OE) mice are more resistant to fatigue and can run significantly longer distances than wild-type (WT). Studies with isolated extensor digitorum longus (EDL) muscles showed that Sln(OE) EDL produced higher twitch force than WT. The force-frequency curves were not different between WT and Sln(OE) EDLs, but at lower frequencies the pyruvate-induced potentiation of force was significantly higher in Sln(OE) EDL. SLN overexpression did not alter the twitch and force-frequency curve in isolated soleus muscle. However, during a 10-min fatigue protocol, both EDL and soleus from Sln(OE) mice fatigued significantly less than WT muscles. Interestingly, Sln(OE) muscles showed higher carnitine palmitoyl transferase-1 protein expression, which could enhance fatty acid metabolism. In addition, lactate dehydrogenase expression was higher in Sln(OE) EDL, suggesting increased glycolytic capacity. We also found an increase in store-operated calcium entry (SOCE) in isolated flexor digitorum brevis fibers of Sln(OE) compared with WT mice. These data allow us to conclude that increased SLN expression improves skeletal muscle performance during prolonged muscle activity by increasing SOCE and muscle energetics.


Assuntos
Tolerância ao Exercício , Proteínas Musculares/fisiologia , Músculo Esquelético/fisiologia , Proteolipídeos/fisiologia , Animais , Cálcio/metabolismo , Calsequestrina/metabolismo , Carnitina O-Palmitoiltransferase/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Fadiga Muscular , Miosinas/metabolismo , Condicionamento Físico Animal , Ácido Pirúvico/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo
19.
Circulation ; 129(25): 2673-81, 2014 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-24888331

RESUMO

BACKGROUND: Catecholaminergic polymorphic ventricular tachycardia is an inherited arrhythmogenic disorder characterized by sudden cardiac death in children. Drug therapy is still insufficient to provide full protection against cardiac arrest, and the use of implantable defibrillators in the pediatric population is limited by side effects. There is therefore a need to explore the curative potential of gene therapy for this disease. We investigated the efficacy and durability of viral gene transfer of the calsequestrin 2 (CASQ2) wild-type gene in a catecholaminergic polymorphic ventricular tachycardia knock-in mouse model carrying the CASQ2(R33Q/R33Q) (R33Q) mutation. METHODS AND RESULTS: We engineered an adeno-associated viral vector serotype 9 (AAV9) containing cDNA of CASQ2 wild-type (AAV9-CASQ2) plus the green fluorescent protein (GFP) gene to infect newborn R33Q mice studied by in vivo and in vitro protocols at 6, 9, and 12 months to investigate the ability of the infection to prevent the disease and adult R33Q mice studied after 2 months to assess whether the AAV9-CASQ2 delivery could revert the catecholaminergic polymorphic ventricular tachycardia phenotype. In both protocols, we observed the restoration of physiological expression and interaction of CASQ2, junctin, and triadin; the rescue of electrophysiological and ultrastructural abnormalities in calcium release units present in R33Q mice; and the lack of life-threatening arrhythmias. CONCLUSIONS: Our data demonstrate that viral gene transfer of wild-type CASQ2 into the heart of R33Q mice prevents and reverts severe manifestations of catecholaminergic polymorphic ventricular tachycardia and that this curative effect lasts for 1 year after a single injection of the vector, thus posing the rationale for the design of a clinical trial.


Assuntos
Envelhecimento , Calsequestrina/genética , Dependovirus/genética , Taquicardia Ventricular/terapia , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Calsequestrina/metabolismo , Proteínas de Transporte/metabolismo , Modelos Animais de Doenças , Feminino , Terapia Genética , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Oxigenases de Função Mista/metabolismo , Proteínas Musculares/metabolismo , Mutação/genética , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/patologia , Resultado do Tratamento
20.
RNA ; 20(3): 373-81, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24412912

RESUMO

Polycistronic transcripts are considered rare in the human genome. Initiation of translation of internal ORFs of eukaryotic genes has been shown to use either leaky scanning or highly structured IRES regions to access initiation codons. Studies on mammalian viruses identified a mechanism of coupled translation termination-reinitiation that allows translation of an additional ORF. Here, the ribosome terminating translation of ORF-1 translocates upstream to reinitiate translation of ORF-2. We have devised an algorithm to identify mRNAs in the human transcriptome in which the major ORF-1 overlaps a second ORF capable of encoding a product of at least 50 aa in length. This identified 4368 transcripts representing 2214 genes. We investigated 24 transcripts, 22 of which were shown to express a protein from ORF-2 highlighting that 3' UTRs contain protein-coding potential more frequently than previously suspected. Five transcripts accessed ORF-2 using a process of coupled translation termination-reinitiation. Analysis of one transcript, encoding the CASQ2 protein, showed that the mechanism by which the coupling process of the cellular mRNAs was achieved was novel. This process was not directed by the mRNA sequence but required an aspartate-rich repeat region at the carboxyl terminus of the terminating ORF-1 protein. Introduction of wobble mutations for the aspartate codon had no effect, whereas replacing aspartate for glutamate repeats eliminated translational coupling. This is the first description of a coordinated expression of two proteins from cellular mRNAs using a coupled translation termination-reinitiation process and is the first example of such a process being determined at the amino acid level.


Assuntos
Ácido Aspártico/genética , Calsequestrina/genética , Fases de Leitura Aberta/genética , Iniciação Traducional da Cadeia Peptídica , Terminação Traducional da Cadeia Peptídica , RNA Mensageiro/genética , Algoritmos , Sequência de Bases , Western Blotting , Calsequestrina/metabolismo , Proliferação de Células , Ensaio de Imunoadsorção Enzimática , Humanos , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/metabolismo , Dados de Sequência Molecular , Homologia de Sequência do Ácido Nucleico , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA