Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Circulation ; 144(21): 1694-1713, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34648376

RESUMEN

BACKGROUND: Barth syndrome (BTHS) is caused by mutations of the gene encoding tafazzin, which catalyzes maturation of mitochondrial cardiolipin and often manifests with systolic dysfunction during early infancy. Beyond the first months of life, BTHS cardiomyopathy typically transitions to a phenotype of diastolic dysfunction with preserved ejection fraction, blunted contractile reserve during exercise, and arrhythmic vulnerability. Previous studies traced BTHS cardiomyopathy to mitochondrial formation of reactive oxygen species (ROS). Because mitochondrial function and ROS formation are regulated by excitation-contraction coupling, integrated analysis of mechano-energetic coupling is required to delineate the pathomechanisms of BTHS cardiomyopathy. METHODS: We analyzed cardiac function and structure in a mouse model with global knockdown of tafazzin (Taz-KD) compared with wild-type littermates. Respiratory chain assembly and function, ROS emission, and Ca2+ uptake were determined in isolated mitochondria. Excitation-contraction coupling was integrated with mitochondrial redox state, ROS, and Ca2+ uptake in isolated, unloaded or preloaded cardiac myocytes, and cardiac hemodynamics analyzed in vivo. RESULTS: Taz-KD mice develop heart failure with preserved ejection fraction (>50%) and age-dependent progression of diastolic dysfunction in the absence of fibrosis. Increased myofilament Ca2+ affinity and slowed cross-bridge cycling caused diastolic dysfunction, in part, compensated by accelerated diastolic Ca2+ decay through preactivated sarcoplasmic reticulum Ca2+-ATPase. Taz deficiency provoked heart-specific loss of mitochondrial Ca2+ uniporter protein that prevented Ca2+-induced activation of the Krebs cycle during ß-adrenergic stimulation, oxidizing pyridine nucleotides and triggering arrhythmias in cardiac myocytes. In vivo, Taz-KD mice displayed prolonged QRS duration as a substrate for arrhythmias, and a lack of inotropic response to ß-adrenergic stimulation. Cellular arrhythmias and QRS prolongation, but not the defective inotropic reserve, were restored by inhibiting Ca2+ export through the mitochondrial Na+/Ca2+ exchanger. All alterations occurred in the absence of excess mitochondrial ROS in vitro or in vivo. CONCLUSIONS: Downregulation of mitochondrial Ca2+ uniporter, increased myofilament Ca2+ affinity, and preactivated sarcoplasmic reticulum Ca2+-ATPase provoke mechano-energetic uncoupling that explains diastolic dysfunction and the lack of inotropic reserve in BTHS cardiomyopathy. Furthermore, defective mitochondrial Ca2+ uptake provides a trigger and a substrate for ventricular arrhythmias. These insights can guide the ongoing search for a cure of this orphaned disease.


Asunto(s)
Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/etiología , Síndrome de Barth/complicaciones , Síndrome de Barth/genética , Canales de Calcio/deficiencia , Contracción Miocárdica/genética , Adenosina Trifosfato/biosíntesis , Animales , Síndrome de Barth/metabolismo , Biomarcadores , Encéfalo/metabolismo , Calcio/metabolismo , Diástole , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Acoplamiento Excitación-Contracción/genética , Pruebas de Función Cardíaca , Humanos , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/genética , Mitocondrias Cardíacas/metabolismo , Músculo Esquelético/metabolismo , Miocitos Cardíacos/metabolismo , NADP/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Volumen Sistólico , Sístole
2.
Int J Mol Sci ; 22(11)2021 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-34072258

RESUMEN

Mutations in striated preferentially expressed protein kinase (SPEG), a member of the myosin light chain kinase protein family, are associated with centronuclear myopathy (CNM), cardiomyopathy, or a combination of both. Burgeoning evidence suggests that SPEG plays critical roles in the development, maintenance, and function of skeletal and cardiac muscles. Here we review the genotype-phenotype relationships and the molecular mechanisms of SPEG-related diseases. This review will focus on the progress made toward characterizing SPEG and its interacting partners, and its multifaceted functions in muscle regeneration, triad development and maintenance, and excitation-contraction coupling. We will also discuss future directions that are yet to be investigated including understanding of its tissue-specific roles, finding additional interacting proteins and their relationships. Understanding the basic mechanisms by which SPEG regulates muscle development and function will provide critical insights into these essential processes and help identify therapeutic targets in SPEG-related disorders.


Asunto(s)
Susceptibilidad a Enfermedades , Expresión Génica , Desarrollo de Músculos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Proteínas Portadoras/metabolismo , Acoplamiento Excitación-Contracción/genética , Regulación de la Expresión Génica , Humanos , Desarrollo de Músculos/genética , Proteínas Musculares/química , Músculo Esquelético/metabolismo , Mutación , Miocardio/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Isoformas de Proteínas , Proteínas Serina-Treonina Quinasas/química , Regeneración/genética , Transducción de Señal , Relación Estructura-Actividad
3.
JCI Insight ; 6(3)2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33411695

RESUMEN

Loss-of-function (LOF) variants in SCN1B, encoding voltage-gated sodium channel ß1 subunits, are linked to human diseases with high risk of sudden death, including developmental and epileptic encephalopathy and cardiac arrhythmia. ß1 Subunits modulate the cell-surface localization, gating, and kinetics of sodium channel pore-forming α subunits. They also participate in cell-cell and cell-matrix adhesion, resulting in intracellular signal transduction, promotion of cell migration, calcium handling, and regulation of cell morphology. Here, we investigated regulated intramembrane proteolysis (RIP) of ß1 by BACE1 and γ-secretase and show that ß1 subunits are substrates for sequential RIP by BACE1 and γ-secretase, resulting in the generation of a soluble intracellular domain (ICD) that is translocated to the nucleus. Using RNA sequencing, we identified a subset of genes that are downregulated by ß1-ICD overexpression in heterologous cells but upregulated in Scn1b-null cardiac tissue, which lacks ß1-ICD signaling, suggesting that the ß1-ICD may normally function as a molecular brake on gene transcription in vivo. We propose that human disease variants resulting in SCN1B LOF cause transcriptional dysregulation that contributes to altered excitability. Moreover, these results provide important insights into the mechanism of SCN1B-linked channelopathies, adding RIP-excitation coupling to the multifunctionality of sodium channel ß1 subunits.


Asunto(s)
Subunidad beta-1 de Canal de Sodio Activado por Voltaje/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Cricetulus , Acoplamiento Excitación-Contracción/genética , Acoplamiento Excitación-Contracción/fisiología , Expresión Génica , Células HEK293 , Humanos , Mutación con Pérdida de Función , Ratones , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Proteolisis , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo , Transducción de Señal , Subunidad beta-1 de Canal de Sodio Activado por Voltaje/deficiencia , Subunidad beta-1 de Canal de Sodio Activado por Voltaje/genética
4.
PLoS Genet ; 16(6): e1008829, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32502151

RESUMEN

Ion channels are present at specific levels within subcellular compartments of excitable cells. The regulation of ion channel trafficking and targeting is an effective way to control cell excitability. The BK channel is a calcium-activated potassium channel that serves as a negative feedback mechanism at presynaptic axon terminals and sites of muscle excitation. The C. elegans BK channel ortholog, SLO-1, requires an endoplasmic reticulum (ER) membrane protein for efficient anterograde transport to these locations. Here, we found that, in the absence of this ER membrane protein, SLO-1 channels that are seemingly normally folded and expressed at physiological levels undergo SEL-11/HRD1-mediated ER-associated degradation (ERAD). This SLO-1 degradation is also indirectly regulated by a SKN-1A/NRF1-mediated transcriptional mechanism that controls proteasome levels. Therefore, our data indicate that SLO-1 channel density is regulated by the competitive balance between the efficiency of ER trafficking machinery and the capacity of ERAD.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiología , Proteínas de Unión al ADN/metabolismo , Degradación Asociada con el Retículo Endoplásmico/genética , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Terminales Presinápticos/metabolismo , Factores de Transcripción/metabolismo , Aldicarb/farmacología , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Acoplamiento Excitación-Contracción/efectos de los fármacos , Acoplamiento Excitación-Contracción/genética , Retroalimentación Fisiológica/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Músculos/inervación , Terminales Presinápticos/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal , Isoformas de Proteínas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
5.
Gene ; 752: 144765, 2020 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-32413480

RESUMEN

The natural flight response in shrimp is powered by rapid contractions of the abdominal muscle fibres to propel themselves backwards away from perceived danger. This muscle contraction is dependent on repetitive depolarization of muscle plasma membrane, triggering tightly spaced cytoplasmic [Ca2+] transients and rapidly rising tetanic force responses. To achieve such high amplitude and high frequency of Ca2+ transients requires a high abundance of sarcoplasmic/endoplasmic reticulum Ca2+ ATPase (SERCA) to rapidly clear cytoplasmic Ca2+ between each transient and an efficient Ca2+ release system consisting of the Ryanodine Receptor (RyR), and voltage gated Ca2+ channels (CaVs). With the aim to expand our knowledge of muscle gene function and identify orthologous genes regulating muscle excitation-contraction (EC) coupling, this study assembled nine Penaeid shrimp muscle transcriptomes. On average, the nine transcriptomes contained 27,000 contigs, with an annotation rate of 36% and a BUSCO completeness of 70%. Despite maintaining their function, the crustacean RyR and CaV proteins showed evidence of significant diversification from mammalian orthologs, while SERCA remained more conserved. Several key components of protein interaction were conserved, while others showed distinct crustacean specific evolutionary adaptations. Lastly, this study revealed approximately 1,000 orthologous genes involved in muscle specific processes present across all nine species.


Asunto(s)
Acoplamiento Excitación-Contracción/genética , Penaeidae/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Animales , Evolución Biológica , Calcio/metabolismo , Canales de Calcio Tipo L/metabolismo , Señalización del Calcio/fisiología , Citosol/metabolismo , Evolución Molecular , Contracción Muscular/fisiología , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Especificidad de la Especie , Transcriptoma/genética
7.
Adv Exp Med Biol ; 1131: 281-320, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31646515

RESUMEN

In mammalian cardiomyocytes, Ca2+ influx through L-type voltage-gated Ca2+ channels (VGCCs) is amplified by release of Ca2+ via type 2 ryanodine receptors (RyR2) in the sarcoplasmic reticulum (SR): a process termed Ca2+-induced Ca2+-release (CICR). In mammalian skeletal muscles, VGCCs play a distinct role as voltage-sensors, physically interacting with RyR1 channels to initiate Ca2+ release in a mechanism termed depolarisation-induced Ca2+-release (DICR). In the current study, we surveyed the genomes of animals and their close relatives, to explore the evolutionary history of genes encoding three proteins pivotal for ECC: L-type VGCCs; RyRs; and a protein family that anchors intracellular organelles to plasma membranes, namely junctophilins (JPHs). In agreement with earlier studies, we find that non-vertebrate eukaryotes either lack VGCCs, RyRs and JPHs; or contain a single homologue of each protein. Furthermore, the molecular features of these proteins thought to be essential for DICR are only detectable within vertebrates and not in any other taxonomic group. Consistent with earlier physiological and ultrastructural observations, this suggests that CICR is the most basal form of ECC and that DICR is a vertebrate innovation. This development was accompanied by the appearance of multiple homologues of RyRs, VGCCs and junctophilins in vertebrates, thought to have arisen by 'whole genome replication' mechanisms. Subsequent gene duplications and losses have resulted in distinct assemblies of ECC components in different vertebrate clades, with striking examples being the apparent absence of RyR2 from amphibians, and additional duplication events for all three ECC proteins in teleost fish. This is consistent with teleosts possessing the most derived mode of DICR, with their Cav1.1 VGCCs completely lacking in Ca2+ channel activity.


Asunto(s)
Canales de Calcio Tipo L , Evolución Molecular , Acoplamiento Excitación-Contracción , Canal Liberador de Calcio Receptor de Rianodina , Animales , Canales de Calcio Tipo L/metabolismo , Acoplamiento Excitación-Contracción/genética , Peces/genética , Peces/metabolismo , Genoma/genética , Músculo Esquelético/fisiología , Miocitos Cardíacos/fisiología , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/fisiología
8.
J Mol Cell Cardiol ; 133: 57-66, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31158360

RESUMEN

AIMS: Cyclic AMP phosphodiesterases (PDEs) are important modulators of the cardiac response to ß-adrenergic receptor (ß-AR) stimulation. PDE3 is classically considered as the major cardiac PDE in large mammals and human, while PDE4 is preponderant in rodents. However, it remains unclear whether PDE4 also plays a functional role in large mammals. Our purpose was to understand the role of PDE4 in cAMP hydrolysis and excitation-contraction coupling (ECC) in the pig heart, a relevant pre-clinical model. METHODS AND RESULTS: Real-time cAMP variations were measured in isolated adult pig right ventricular myocytes (APVMs) using a Förster resonance energy transfer (FRET) biosensor. ECC was investigated in APVMs loaded with Fura-2 and paced at 1 Hz allowing simultaneous measurement of intracellular Ca2+ and sarcomere shortening. The expression of the different PDE4 subfamilies was assessed by Western blot in pig right ventricles and APVMs. Similarly to PDE3 inhibition with cilostamide (Cil), PDE4 inhibition with Ro 20-1724 (Ro) increased cAMP levels and inotropy under basal conditions. PDE4 inhibition enhanced the effects of the non-selective ß-AR agonist isoprenaline (Iso) and the effects of Cil, and increased spontaneous diastolic Ca2+ waves (SCWs) in these conditions. PDE3A, PDE4A, PDE4B and PDE4D subfamilies are expressed in pig ventricles. In APVMs isolated from a porcine model of repaired tetralogy of Fallot which leads to right ventricular failure, PDE4 inhibition also exerts inotropic and pro-arrhythmic effects. CONCLUSIONS: Our results show that PDE4 controls ECC in APVMs and suggest that PDE4 inhibitors exert inotropic and pro-arrhythmic effects upon PDE3 inhibition or ß-AR stimulation in our pre-clinical model. Thus, PDE4 inhibitors should be used with caution in clinics as they may lead to arrhythmogenic events upon stress.


Asunto(s)
AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Acoplamiento Excitación-Contracción/genética , Miocitos Cardíacos/fisiología , Potenciales de Acción/efectos de los fármacos , Agonistas Adrenérgicos beta/farmacología , Animales , Señalización del Calcio/efectos de los fármacos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Familia de Multigenes , Miocitos Cardíacos/efectos de los fármacos , Inhibidores de Fosfodiesterasa 3/farmacología , Inhibidores de Fosfodiesterasa 4/farmacología , Receptores Adrenérgicos beta/metabolismo , Porcinos
9.
J Mol Cell Cardiol ; 128: 145-157, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30731085

RESUMEN

Cardiac sodium (Na+) potassium ATPase (NaK) pumps, neuronal sodium channels (INa), and sodium calcium (Ca2+) exchangers (NCX1) may co-localize to form a Na+ microdomain. It remains controversial as to whether neuronal INa contributes to local Na+ accumulation, resulting in reversal of nearby NCX1 and influx of Ca2+ into the cell. Therefore, there has been great interest in the possible roles of a Na+ microdomain in cardiac Ca2+-induced Ca2+ release (CICR). In addition, the important role of co-localization of NaK and NCX1 in regulating localized Na+ and Ca2+ levels and CICR in ankyrin-B deficient (ankyrin-B+/-) cardiomyocytes has been examined in many recent studies. Altered Na+ dynamics may contribute to the appearance of arrhythmias, but the mechanisms underlying this relationship remain unclear. In order to investigate this, we present a mechanistic canine cardiomyocyte model which reproduces independent local dyadic junctional SR (JSR) Ca2+ release events underlying cell-wide excitation-contraction coupling, as well as a three-dimensional super-resolution model of the Ca2+ spark that describes local Na+ dynamics as governed by NaK pumps, neuronal INa, and NCX1. The model predicts the existence of Na+ sparks, which are generated by NCX1 and exhibit significantly slower dynamics as compared to Ca2+ sparks. Moreover, whole-cell simulations indicate that neuronal INa in the cardiac dyad plays a key role during the systolic phase. Rapid inward neuronal INa can elevate dyadic [Na+] to 35-40 mM, which drives reverse-mode NCX1 transport, and therefore promotes Ca2+ entry into the dyad, enhancing the trigger for JSR Ca2+ release. The specific role of decreased co-localization of NaK and NCX1 in ankyrin-B+/- cardiomyocytes was examined. Model results demonstrate that a reduction in the local NCX1- and NaK-mediated regulation of dyadic [Ca2+] and [Na+] results in an increase in Ca2+ spark activity during isoproterenol stimulation, which in turn stochastically activates NCX1 in the dyad. This alteration in NCX1/NaK co-localization interrupts the balance between NCX1 and NaK currents in a way that leads to enhanced depolarizing inward current during the action potential plateau, which ultimately leads to a higher probability of L-type Ca2+ channel reopening and arrhythmogenic early-afterdepolarizations.


Asunto(s)
Ancirinas/genética , Miocitos Cardíacos/metabolismo , Intercambiador de Sodio-Calcio/genética , ATPasa Intercambiadora de Sodio-Potasio/genética , Potenciales de Acción/genética , Animales , Calcio/metabolismo , Canales de Calcio Tipo L/genética , Señalización del Calcio/genética , Perros , Acoplamiento Excitación-Contracción/genética , Humanos , Miocitos Cardíacos/patología , Potasio/metabolismo , Sodio/metabolismo , Canales de Sodio/genética
10.
Science ; 362(6421)2018 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-30409805

RESUMEN

Junctophilin-2 (JP2) is a structural protein required for normal excitation-contraction (E-C) coupling. After cardiac stress, JP2 is cleaved by the calcium ion-dependent protease calpain, which disrupts the E-C coupling ultrastructural machinery and drives heart failure progression. We found that stress-induced proteolysis of JP2 liberates an N-terminal fragment (JP2NT) that translocates to the nucleus, binds to genomic DNA, and controls expression of a spectrum of genes in cardiomyocytes. Transgenic overexpression of JP2NT in mice modifies the transcriptional profile, resulting in attenuated pathological remodeling in response to cardiac stress. Conversely, loss of nuclear JP2NT function accelerates stress-induced development of hypertrophy and heart failure in mutant mice. These data reveal a self-protective mechanism in failing cardiomyocytes that transduce mechanical information (E-C uncoupling) into salutary transcriptional reprogramming in the stressed heart.


Asunto(s)
Cardiomegalia/genética , Núcleo Celular/metabolismo , Acoplamiento Excitación-Contracción/genética , Regulación de la Expresión Génica , Proteínas de la Membrana/metabolismo , Proteínas Musculares/metabolismo , Contracción Miocárdica/genética , Miocitos Cardíacos/patología , Remodelación Ventricular/genética , Transporte Activo de Núcleo Celular , Adaptación Fisiológica/genética , Animales , Calpaína/metabolismo , Cardiomegalia/fisiopatología , Humanos , Factores de Transcripción MEF2/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Musculares/química , Proteínas Musculares/genética , Miocitos Cardíacos/metabolismo , Proteolisis , Transcripción Genética
11.
Nat Commun ; 9(1): 4176, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30301979

RESUMEN

Long non-coding RNAs (lncRNAs) have emerged as a new class of gene expression regulators playing key roles in many biological and pathophysiological processes. Here, we identify cardiac conduction regulatory RNA (CCRR) as an antiarrhythmic lncRNA. CCRR is downregulated in a mouse model of heart failure (HF) and in patients with HF, and this downregulation slows cardiac conduction and enhances arrhythmogenicity. Moreover, CCRR silencing induces arrhythmias in healthy mice. CCRR overexpression eliminates these detrimental alterations. HF or CCRR knockdown causes destruction of intercalated discs and gap junctions to slow longitudinal cardiac conduction. CCRR overexpression improves cardiac conduction by blocking endocytic trafficking of connexin43 (Cx43) to prevent its degradation via binding to Cx43-interacting protein CIP85, whereas CCRR silence does the opposite. We identified the functional domain of CCRR, which can reproduce the functional roles and pertinent molecular events of full-length CCRR. Our study suggests CCRR replacement a potential therapeutic approach for pathological arrhythmias.


Asunto(s)
Acoplamiento Excitación-Contracción/genética , Espacio Extracelular/metabolismo , Sistema de Conducción Cardíaco/metabolismo , ARN Largo no Codificante/metabolismo , Animales , Arritmias Cardíacas/genética , Conexina 43/metabolismo , Uniones Comunicantes/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Insuficiencia Cardíaca/genética , Humanos , Masculino , Ratones Endogámicos C57BL , Modelos Biológicos , Miocardio/metabolismo , Miocardio/patología , Miocardio/ultraestructura , ARN Largo no Codificante/genética , Transducción de Señal , Fracciones Subcelulares/metabolismo , Proteínas de Unión al GTP rab/metabolismo
12.
PLoS One ; 13(10): e0205719, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30332462

RESUMEN

AIMS: Dilated cardiomyopathy (DCM), a myocardial disorder that can result in progressive heart failure and arrhythmias, is defined by ventricular chamber enlargement and dilatation, and systolic dysfunction. Despite extensive research, the pathological mechanisms of DCM are unclear mainly due to numerous mutations in different gene families resulting in the same outcome-decreased ventricular function. Titin (TTN)-a giant protein, expressed in cardiac and skeletal muscles, is an important part of the sarcomere, and thus TTN mutations are the most common cause of adult DCM. To decipher the basis for the cardiac pathology in titin-mutated patients, we investigated the hypothesis that induced Pluripotent Stem Cell (iPSC)-derived cardiomyocytes (iPSC-CM) generated from patients, recapitulate the disease phenotype. The hypothesis was tested by 3 Aims: (1) Investigate key features of the excitation-contraction-coupling machinery; (2) Investigate the responsiveness to positive inotropic interventions; (3) Investigate the proteome profile of the AuP cardiomyocytes using mass-spectrometry (MS). METHODS AND RESULTS: iPSC were generated from the patients' skin fibroblasts. The major findings were: (1) Sarcomeric organization analysis in mutated iPSC-CM showed defects in assembly and maintenance of sarcomeric structure. (2) Mutated iPSC-CM exhibited diminished inotropic and lusitropic responses to ß-adrenergic stimulation with isoproterenol, increased [Ca2+]out and angiotensin-II. Additionally, mutated iPSC-CM displayed prolonged recovery in response to caffeine. These findings may result from defective or lack of interactions of the sarcomeric components with titin through its kinase domain which is absent in the mutated cells. CONCLUSIONS: These findings show that the mutated cardiomyocytes from DCM patients recapitulate abnormalities of the inherited cardiomyopathies, expressed as blunted inotropic response.


Asunto(s)
Cardiomiopatía Dilatada/genética , Diferenciación Celular/genética , Conectina/genética , Contracción Miocárdica/genética , Miocitos Cardíacos/patología , Adulto , Anciano , Cardiomiopatía Dilatada/patología , Acoplamiento Excitación-Contracción/genética , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Isoproterenol/farmacología , Masculino , Mutación , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/fisiología , Proteoma
13.
Annu Rev Genet ; 52: 373-396, 2018 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-30208288

RESUMEN

Ion channels are membrane proteins responsible for the passage of ions down their electrochemical gradients and across biological membranes. In this, they generate and shape action potentials and provide secondary messengers for various signaling pathways. They are often part of larger complexes containing auxiliary subunits and regulatory proteins. Channelopathies arise from mutations in the genes encoding ion channels or their associated proteins. Recent advances in cryo-electron microscopy have resulted in an explosion of ion channel structures in multiple states, generating a wealth of new information on channelopathies. Disease-associated mutations fall into different categories, interfering with ion permeation, protein folding, voltage sensing, ligand and protein binding, and allosteric modulation of channel gating. Prime examples of these are Ca2+-selective channels expressed in myocytes, for which multiple structures in distinct conformational states have recently been uncovered. We discuss the latest insights into these calcium channelopathies from a structural viewpoint.


Asunto(s)
Canales de Calcio/genética , Calcio/metabolismo , Canalopatías/genética , Contracción Muscular/genética , Animales , Canales de Calcio/metabolismo , Membrana Celular/metabolismo , Membrana Celular/patología , Membrana Celular/ultraestructura , Canalopatías/metabolismo , Canalopatías/patología , Microscopía por Crioelectrón , Acoplamiento Excitación-Contracción/genética , Humanos , Transducción de Señal/genética
14.
JCI Insight ; 3(15)2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-30089727

RESUMEN

Cantu syndrome (CS) is characterized by multiple vascular and cardiac abnormalities including vascular dilation and tortuosity, systemic hypotension, and cardiomegaly. The disorder is caused by gain-of-function (GOF) mutations in genes encoding pore-forming (Kir6.1, KCNJ8) and accessory (SUR2, ABCC9) ATP-sensitive potassium (KATP) channel subunits. However, there is little understanding of the link between molecular dysfunction and the complex pathophysiology observed, and there is no known treatment, in large part due to the lack of appropriate preclinical disease models in which to test therapies. Notably, expression of Kir6.1 and SUR2 does not fully overlap, and the relative contribution of KATP GOF in various cardiovascular tissues remains to be elucidated. To investigate pathophysiologic mechanisms in CS we have used CRISPR/Cas9 engineering to introduce CS-associated SUR2[A478V] and Kir6.1[V65M] mutations to the equivalent endogenous loci in mice. Mirroring human CS, both of these animals exhibit low systemic blood pressure and dilated, compliant blood vessels, as well dramatic cardiac enlargement, the effects being more severe in V65M animals than in A478V animals. In both animals, whole-cell patch-clamp recordings reveal enhanced basal KATP conductance in vascular smooth muscle, explaining vasodilation and lower blood pressure, and demonstrating a cardinal role for smooth muscle KATP dysfunction in CS etiology. Echocardiography confirms in situ cardiac enlargement and increased cardiac output in both animals. Patch-clamp recordings reveal reduced ATP sensitivity of ventricular myocyte KATP channels in A478V, but normal ATP sensitivity in V65M, suggesting that cardiac remodeling occurs secondary to KATP overactivity outside of the heart. These SUR2[A478V] and Kir6.1[V65M] animals thus reiterate the key cardiovascular features seen in human CS. They establish the molecular basis of the pathophysiological consequences of reduced smooth muscle excitability resulting from SUR2/Kir6.1-dependent KATP GOF, and provide a validated animal model in which to examine potential therapeutic approaches to treating CS.


Asunto(s)
Cardiomegalia/fisiopatología , Ventrículos Cardíacos/fisiopatología , Hipertricosis/fisiopatología , Canales KATP/metabolismo , Osteocondrodisplasias/fisiopatología , Receptores de Sulfonilureas/metabolismo , Animales , Cardiomegalia/diagnóstico , Cardiomegalia/genética , Modelos Animales de Enfermedad , Ecocardiografía , Acoplamiento Excitación-Contracción/genética , Femenino , Mutación con Ganancia de Función , Técnicas de Sustitución del Gen , Ventrículos Cardíacos/diagnóstico por imagen , Humanos , Hipertricosis/diagnóstico , Hipertricosis/genética , Canales KATP/genética , Masculino , Ratones , Ratones Transgénicos , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiopatología , Miocitos Cardíacos , Osteocondrodisplasias/diagnóstico , Osteocondrodisplasias/genética , Técnicas de Placa-Clamp , Receptores de Sulfonilureas/genética , Vasodilatación/genética , Remodelación Ventricular/genética
15.
J Cell Physiol ; 233(12): 9045-9051, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30071129

RESUMEN

Excitation-contraction (EC) coupling in skeletal muscles operates through a physical interaction between the dihydropyridine receptor (DHPR), acting as a voltage sensor, and the ryanodine receptor (RyR1), acting as a calcium release channel. Recently, the adaptor protein SH3 and cysteine-rich containing protein 3 (STAC3) has been identified as a myopathy disease gene and as an additional essential EC coupling component. STAC3 interacts with DHPR sequences including the critical EC coupling domain and has been proposed to function in linking the DHPR and RyR1. However, we and others demonstrated that incorporation of recombinant STAC3 into skeletal muscle triads critically depends only on the DHPR but not the RyR1. On the contrary, here, we provide evidence that endogenous STAC3 incorporates into triads in the absence of the DHPR in myotubes and muscle fibers of dysgenic mice. This finding demonstrates that STAC3 interacts with additional triad proteins and is consistent with its proposed role in directly or indirectly linking the DHPR with the RyR1.


Asunto(s)
Canales de Calcio Tipo L/genética , Enfermedades Musculares/genética , Proteínas del Tejido Nervioso/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Calcio/metabolismo , Señalización del Calcio/genética , Acoplamiento Excitación-Contracción/genética , Acoplamiento Excitación-Contracción/fisiología , Humanos , Ratones , Fibras Musculares Esqueléticas , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiología , Enfermedades Musculares/fisiopatología
16.
Skelet Muscle ; 8(1): 8, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29510724

RESUMEN

BACKGROUND: Skeletal muscles express a highly specialized proteome that allows the metabolism of energy sources to mediate myofiber contraction. This muscle-specific proteome is partially derived through the muscle-specific transcription of a subset of genes. Surprisingly, RNA sequencing technologies have also revealed a significant role for muscle-specific alternative splicing in generating protein isoforms that give specialized function to the muscle proteome. MAIN BODY: In this review, we discuss the current knowledge with respect to the mechanisms that allow pre-mRNA transcripts to undergo muscle-specific alternative splicing while identifying some of the key trans-acting splicing factors essential to the process. The importance of specific splicing events to specialized muscle function is presented along with examples in which dysregulated splicing contributes to myopathies. Though there is now an appreciation that alternative splicing is a major contributor to proteome diversification, the emergence of improved "targeted" proteomic methodologies for detection of specific protein isoforms will soon allow us to better appreciate the extent to which alternative splicing modifies the activity of proteins (and their ability to interact with other proteins) in the skeletal muscle. In addition, we highlight a continued need to better explore the signaling pathways that contribute to the temporal control of trans-acting splicing factor activity to ensure specific protein isoforms are expressed in the proper cellular context. CONCLUSIONS: An understanding of the signal-dependent and signal-independent events driving muscle-specific alternative splicing has the potential to provide us with novel therapeutic strategies to treat different myopathies.


Asunto(s)
Empalme Alternativo/fisiología , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Proteoma/genética , Proteínas Contráctiles/genética , Proteínas Contráctiles/fisiología , Acoplamiento Excitación-Contracción/genética , Acoplamiento Excitación-Contracción/fisiología , Humanos , Factores de Transcripción MEF2/genética , Mitocondrias Musculares/genética , Mitocondrias Musculares/fisiología , Proteínas Musculares/metabolismo , Enfermedades Musculares/genética , Enfermedades Musculares/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Proteoma/metabolismo , Factores de Transcripción/genética
17.
Eur J Pharmacol ; 807: 117-126, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28428008

RESUMEN

Gravin (AKAP12) is an A-kinase-anchoring-protein that scaffolds protein kinase A (PKA), ß2-adrenergic receptor (ß2-AR), protein phosphatase 2B and protein kinase C. Gravin facilitates ß2-AR-dependent signal transduction through PKA to modulate cardiac excitation-contraction coupling and its removal positively affects cardiac contraction. Trabeculae from the right ventricles of gravin mutant (gravin-t/t) mice were employed for force determination. Simultaneously, corresponding intracellular Ca2+ transient ([Ca2+]i) were measured. Twitch force (Tf)-interval relationship, [Ca2+]i-interval relationship, and the rate of decay of post-extrasysolic potentiation (Rf) were also obtained. Western blot analysis were performed to correlate sarcomeric protein expression with alterations in calcium cycling between the WT and gravin-t/t hearts. Gravin-t/t muscles had similar developed force compared to WT muscles despite having lower [Ca2+]i at any given external Ca2+ concentration ([Ca2+]o). The time to peak force and peak [Ca2+]i were slower and the time to 75% relaxation was significantly prolonged in gravin-t/t muscles. Both Tf-interval and [Ca2+]i-interval relations were depressed in gravin-t/t muscles. Rf, however, did not change. Furthermore, Western blot analysis revealed decreased ryanodine receptor (RyR2) phosphorylation in gravin-t/t hearts. Gravin-t/t cardiac muscle exhibits increased force development in responsiveness to Ca2+. The Ca2+ cycling across the SR appears to be unaltered in gravin-t/t muscle. Our study suggests that gravin is an important component of cardiac contraction regulation via increasing myofilament sensitivity to calcium. Further elucidation of the mechanism can provide insights to role of gravin if any in the pathophysiology of impaired contractility.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/genética , Calcio/metabolismo , Proteínas de Ciclo Celular/genética , Espacio Intracelular/metabolismo , Fenómenos Mecánicos , Mutación , Miocardio/citología , Animales , Fenómenos Biomecánicos , Citosol/metabolismo , Acoplamiento Excitación-Contracción/genética , Regulación de la Expresión Génica/genética , Ratones , Miocardio/metabolismo , Retículo Sarcoplasmático/metabolismo
18.
J Mol Cell Cardiol ; 103: 40-47, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28065668

RESUMEN

BACKGROUND: Phosphorylation of the cardiac ryanodine receptor (RyR2) phospho-site S2808 has been touted by the Marks group as a hallmark of heart failure (HF) and a critical mediator of the physiological fight-or-flight response of the heart. In support of this hypothesis, mice unable to undergo phosphorylation at RyR2-S2808 (S2808A) were significantly protected against HF and displayed a blunted response to adrenergic stimulation. However, the issue remains highly controversial because several groups have been unable to reproduce these findings. An important variable not considered before is the genetic background of the mice used to obtain these divergent results. METHODS AND RESULTS: We backcrossed a RyR2-S2808A mouse into a congenic C57Bl/6 strain, the same strain used by the Marks group to conduct their experiments. We then performed several key experiments to confirm or discard the genetic background of the mouse as a relevant variable, including induction of HF by myocardial infarction and tests of integrity of adrenergic response. Congenic C57Bl/6 harboring the S2808A mutation showed similar echocardiographic parameters that indicated identical progression towards HF compared to wild type controls, and had a normal response to adrenergic stimulation in whole animal and cellular experiments. CONCLUSIONS: The genetic background of the different mouse models is unlikely to be the source of the divergent results obtained by the Marks group in comparison to several other groups. Cardiac adrenergic response and progression towards HF proceed unaltered in mice harboring the RyR2-S2808A mutation. Preventing RyR2-S2808 phosphorylation does not preclude a normal sympathetic response nor mitigates the pathophysiological consequences of MI.


Asunto(s)
Adrenérgicos/farmacología , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Calcio/metabolismo , Modelos Animales de Enfermedad , Acoplamiento Excitación-Contracción/efectos de los fármacos , Acoplamiento Excitación-Contracción/genética , Insuficiencia Cardíaca/mortalidad , Insuficiencia Cardíaca/patología , Pruebas de Función Cardíaca , Ratones , Ratones Transgénicos , Infarto del Miocardio/complicaciones , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Fosforilación/efectos de los fármacos , Canal Liberador de Calcio Receptor de Rianodina/deficiencia , Análisis de Secuencia de ADN
19.
Handb Exp Pharmacol ; 243: 395-421, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27796512

RESUMEN

Heart failure is a significant burden to the global healthcare system and represents an underserved market for new pharmacologic strategies, especially therapies which can address root cause myocyte dysfunction. Modern drugs, surgeries, and state-of-the-art interventions are costly and do not improve survival outcome measures. Gene therapy is an attractive strategy, whereby selected gene targets and their associated regulatory mechanisms can be permanently managed therapeutically in a single treatment. This in theory could be sustainable for the patient's life. Despite the promise, however, gene therapy has numerous challenges that must be addressed together as a treatment plan comprising these key elements: myocyte physiologic target validation, gene target manipulation strategy, vector selection for the correct level of manipulation, and carefully utilizing an efficient delivery route that can be implemented in the clinic to efficiently transfer the therapy within safety limits. This chapter summarizes the key developments in cardiac gene therapy from the perspective of understanding each of these components of the treatment plan. The latest pharmacologic gene targets, gene therapy vectors, delivery routes, and strategies are reviewed.


Asunto(s)
Terapia Genética/métodos , Insuficiencia Cardíaca/terapia , Adenilil Ciclasas/genética , Animales , Apoptosis/genética , Acoplamiento Excitación-Contracción/genética , Fibrosis/genética , Técnicas de Sustitución del Gen , Técnicas de Silenciamiento del Gen , Vectores Genéticos , Humanos , Miocardio , Miofibrillas/genética , Receptores Adrenérgicos beta , Regeneración/genética , Transducción de Señal/genética
20.
Development ; 143(9): 1547-59, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26965373

RESUMEN

Skeletal muscle excitation-contraction (EC) coupling is independent of calcium influx. In fact, alternative splicing of the voltage-gated calcium channel CaV1.1 actively suppresses calcium currents in mature muscle. Whether this is necessary for normal development and function of muscle is not known. However, splicing defects that cause aberrant expression of the calcium-conducting developmental CaV1.1e splice variant correlate with muscle weakness in myotonic dystrophy. Here, we deleted CaV1.1 (Cacna1s) exon 29 in mice. These mice displayed normal overall motor performance, although grip force and voluntary running were reduced. Continued expression of the developmental CaV1.1e splice variant in adult mice caused increased calcium influx during EC coupling, altered calcium homeostasis, and spontaneous calcium sparklets in isolated muscle fibers. Contractile force was reduced and endurance enhanced. Key regulators of fiber type specification were dysregulated and the fiber type composition was shifted toward slower fibers. However, oxidative enzyme activity and mitochondrial content declined. These findings indicate that limiting calcium influx during skeletal muscle EC coupling is important for the secondary function of the calcium signal in the activity-dependent regulation of fiber type composition and to prevent muscle disease.


Asunto(s)
Potenciales de Acción/fisiología , Canales de Calcio Tipo L/genética , Acoplamiento Excitación-Contracción/genética , Fibras Musculares de Contracción Rápida/citología , Fibras Musculares de Contracción Lenta/citología , Debilidad Muscular/genética , Músculo Esquelético/embriología , Empalme Alternativo/genética , Animales , Calcio/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibras Musculares de Contracción Rápida/fisiología , Fibras Musculares de Contracción Lenta/fisiología , Debilidad Muscular/metabolismo , Isoformas de Proteínas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...