Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Arch Biochem Biophys ; 726: 109241, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35667908

RESUMEN

A stacking sodium dodecyl sulfate polyacrylamide gel electrophoresis system has been used to resolve and quantify all the major myofibrillar protein components (actin, myosin, tropomyosin, and troponin C, T, and I). Quantification was achieved by densitometry of the fast green-stained gels calibrated with the use of purified proteins. The approximate molar ratios of these proteins in rabbit muscle are: actin : myosin: tropomyosin: troponin T: troponin I: troponin C = 7:1:1:1:1:1. On the basis of these results and available structural information one obtains an estimate of 254 myosin molecules per thick filament.


Asunto(s)
Miofibrillas , Tropomiosina , Actinas/metabolismo , Animales , Electroforesis en Gel de Poliacrilamida , Músculo Esquelético/metabolismo , Miofibrillas/metabolismo , Miosinas/metabolismo , Conejos , Tropomiosina/metabolismo , Troponina C/metabolismo
2.
J Biol Chem ; 287(3): 2156-67, 2012 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-22086914

RESUMEN

The R21C substitution in cardiac troponin I (cTnI) is the only identified mutation within its unique N-terminal extension that is associated with hypertrophic cardiomyopathy (HCM) in man. Particularly, this mutation is located in the consensus sequence for ß-adrenergic-activated protein kinase A (PKA)-mediated phosphorylation. The mechanisms by which this mutation leads to heart disease are still unclear. Therefore, we generated cTnI knock-in mouse models carrying an R21C mutation to evaluate the resultant functional consequences. Measuring the in vivo levels of incorporated mutant and WT cTnI, and their basal phosphorylation levels by top-down mass spectrometry demonstrated: 1) a dominant-negative effect such that, the R21C+/- hearts incorporated 24.9% of the mutant cTnI within the myofilament; and 2) the R21C mutation abolished the in vivo phosphorylation of Ser(23)/Ser(24) in the mutant cTnI. Adult heterozygous (R21C+/-) and homozygous (R21C+/+) mutant mice activated the fetal gene program and developed a remarkable degree of cardiac hypertrophy and fibrosis. Investigation of cardiac skinned fibers isolated from WT and heterozygous mice revealed that the WT cTnI was completely phosphorylated at Ser(23)/Ser(24) unless the mice were pre-treated with propranolol. After propranolol treatment (-PKA), the pCa-tension relationships of all three mice (i.e. WT, R21C+/-, and R21C+/+) were essentially the same. However, after treatment with propranolol and PKA, the R21C cTnI mutation reduced (R21C+/-) or abolished (R21C+/+) the well known decrease in the Ca(2+) sensitivity of tension that accompanies Ser(23)/Ser(24) cTnI phosphorylation. Altogether, the combined effects of the R21C mutation appear to contribute toward the development of HCM and suggest that another physiological role for the phosphorylation of Ser(23)/Ser(24) in cTnI is to prevent cardiac hypertrophy.


Asunto(s)
Sustitución de Aminoácidos , Cardiomiopatía Hipertrófica Familiar/metabolismo , Mutación Missense , Miocardio/metabolismo , Miofibrillas/metabolismo , Troponina I/metabolismo , Animales , Antiarrítmicos/farmacología , Calcio/metabolismo , Cardiomiopatía Hipertrófica Familiar/genética , Cardiomiopatía Hipertrófica Familiar/patología , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fibrosis Endomiocárdica/genética , Fibrosis Endomiocárdica/metabolismo , Técnicas de Sustitución del Gen , Humanos , Ratones , Ratones Mutantes , Miocardio/patología , Miofibrillas/genética , Miofibrillas/patología , Fosforilación/genética , Propranolol/farmacología , Troponina I/genética
3.
J Biol Chem ; 287(38): 31845-55, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22815480

RESUMEN

Defined as clinically unexplained hypertrophy of the left ventricle, hypertrophic cardiomyopathy (HCM) is traditionally understood as a disease of the cardiac sarcomere. Mutations in TNNC1-encoded cardiac troponin C (cTnC) are a relatively rare cause of HCM. Here, we report clinical and functional characterization of a novel TNNC1 mutation, A31S, identified in a pediatric HCM proband with multiple episodes of ventricular fibrillation and aborted sudden cardiac death. Diagnosed at age 5, the proband is family history-negative for HCM or sudden cardiac death, suggesting a de novo mutation. TnC-extracted cardiac skinned fibers were reconstituted with the cTnC-A31S mutant, which increased Ca(2+) sensitivity with no effect on the maximal contractile force generation. Reconstituted actomyosin ATPase assays with 50% cTnC-A31S:50% cTnC-WT demonstrated Ca(2+) sensitivity that was intermediate between 100% cTnC-A31S and 100% cTnC-WT, whereas the mutant increased the activation of the actomyosin ATPase without affecting the inhibitory qualities of the ATPase. The secondary structure of the cTnC mutant was evaluated by circular dichroism, which did not indicate global changes in structure. Fluorescence studies demonstrated increased Ca(2+) affinity in isolated cTnC, the troponin complex, thin filament, and to a lesser degree, thin filament with myosin subfragment 1. These results suggest that this mutation has a direct effect on the Ca(2+) sensitivity of the myofilament, which may alter Ca(2+) handling and contribute to the arrhythmogenesis observed in the proband. In summary, we report a novel mutation in the TNNC1 gene that is associated with HCM pathogenesis and may predispose to the pathogenesis of a fatal arrhythmogenic subtype of HCM.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Predisposición Genética a la Enfermedad , Mutación , Miocardio/metabolismo , Troponina C/genética , Troponina C/metabolismo , Fibrilación Ventricular/genética , Alelos , Sitios de Unión , Calcio/química , Calcio/metabolismo , Cardiomiopatía Hipertrófica/fisiopatología , Dicroismo Circular , Clonación Molecular , Estudios de Cohortes , Humanos , Conformación Molecular , Miofibrillas/metabolismo , Miosinas/química , Fibrilación Ventricular/fisiopatología
4.
J Biol Chem ; 287(44): 37362-70, 2012 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-22977240

RESUMEN

Human slow skeletal troponin T (HSSTnT) shares a high degree of homology with cardiac TnT (CTnT). Although the presence of HSSTnT has not been confirmed in the heart at the protein level, detectable levels of HSSTnT mRNA have been found. Whether HSSTnT isoforms are expressed transiently remains unknown. Because transient re-expression of HSSTnT may be a potential mechanism of regulating function, we explored the effect of HSSTnT on the regulation of cardiac muscle. At least three HSSTnT isoforms have been found to exist in slow skeletal muscle: HSSTnT1 (+exons 5 and 12), HSSTnT2 (+exon 5, -exon 12), and HSSTnT3 (-exons 5 and 12). Another isoform, HSSTnT hypothetical (Hyp) (-exon 5, +exon 12), has only been found at the mRNA level. Compared with HCTnT3 (adult isoform), Tn complexes containing HSSTnT1, -2, and -3 did not alter the actomyosin ATPase activation and inhibition in the presence and absence of Ca(2+), respectively. HSSTnTHyp was not evaluated as it did not form a Tn complex under a variety of conditions. Porcine papillary skinned fibers displaced with HSSTnT1, -2, or -3 and reconstituted with human cardiac troponin I and troponin C (HCTnI·TnC) complex showed a decrease in the Ca(2+) sensitivity of force development and an increase in maximal recovered force (HSSTnT1 and -3) compared with HCTnT3. In contrast, HSSTnTHyp showed an increase in the Ca(2+) sensitivity of force development. This suggests that re- or overexpression of specific SSTnT isoforms might have therapeutic potential in the failing heart because they increase the maximal force of contraction. In addition, circular dichroism and proteolytic digestion experiments revealed structural differences between HSSTnT isoforms and HCTnT3 and that HSSTnT1 is more susceptible to calpain and trypsin proteolysis than the other HSSTnTs. Overall, HSSTnT isoforms despite being homologues of CTnT may display distinct functional properties in muscle regulation.


Asunto(s)
Contracción Miocárdica , Miocardio/citología , Miocitos Cardíacos/fisiología , Troponina T/fisiología , Animales , Calcio/fisiología , Calpaína/química , Dicroismo Circular , Humanos , Técnicas In Vitro , Miocardio/enzimología , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Miosinas/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Estructura Secundaria de Proteína , Proteolisis , Sus scrofa , Troponina T/química , Troponina T/metabolismo , Tripsina/química
5.
J Muscle Res Cell Motil ; 34(5-6): 441-6, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24264290

RESUMEN

Dr. John Gergely passed away on July 26, 2013 after a long and distinguished career. His publications spanned 67 years. He founded the Department of Muscle Research in the Retina Foundation (which later became the Boston Biomedical Research Institute) and served as director for 34 years. Dr. Gergely served on the editorial boards of ten scientific journals. He was elected as a Fellow of both the Biophysical Society and the American Association for the Advancement of Science. Dr. Gergely made major contributions concerning muscle protein structure and function. He was best known for his work on the troponin complex. The insights of John and his associates have provided the foundation for our understanding of calcium regulation in skeletal and cardiac muscle.


Asunto(s)
Bioquímica/historia , Fisiología/historia , Investigación Biomédica , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Proteínas Musculares/química
6.
J Biol Chem ; 286(2): 1005-13, 2011 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-21056975

RESUMEN

This spectroscopic study examined the steady-state and kinetic parameters governing the cross-bridge effect on the increased Ca(2+) affinity of hypertrophic cardiomyopathy-cardiac troponin C (HCM-cTnC) mutants. Previously, we found that incorporation of the A8V and D145E HCM-cTnC mutants, but not E134D into thin filaments (TFs), increased the apparent Ca(2+) affinity relative to TFs containing the WT protein. Here, we show that the addition of myosin subfragment 1 (S1) to TFs reconstituted with these mutants in the absence of MgATP(2-), the condition conducive to rigor cross-bridge formation, further increased the apparent Ca(2+) affinity. Stopped-flow fluorescence techniques were used to determine the kinetics of Ca(2+) dissociation (k(off)) from the cTnC mutants in the presence of TFs and S1. At a high level of complexity (i.e. TF + S1), an increase in the Ca(2+) affinity and decrease in k(off) was achieved for the A8V and D145E mutants when compared with WT. Therefore, it appears that the cTnC Ca(2+) off-rate is most likely to be affected rather than the Ca(2+) on rate. At all levels of TF complexity, the results obtained with the E134D mutant reproduced those seen with the WT protein. We conclude that strong cross-bridges potentiate the Ca(2+)-sensitizing effect of HCM-cTnC mutants on the myofilament. Finally, the slower k(off) from the A8V and D145E mutants can be directly correlated with the diastolic dysfunction seen in these patients.


Asunto(s)
Calcio/metabolismo , Cardiomiopatía Hipertrófica , Miocitos Cardíacos/fisiología , Troponina C , Citoesqueleto de Actina/fisiología , Animales , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/metabolismo , Cardiomiopatía Hipertrófica/fisiopatología , Reactivos de Enlaces Cruzados/química , Reactivos de Enlaces Cruzados/metabolismo , Humanos , Cinética , Mutagénesis , Contracción Miocárdica/fisiología , Miocitos Cardíacos/citología , Subfragmentos de Miosina/metabolismo , Conformación Proteica , Conejos , Porcinos , Troponina C/química , Troponina C/genética , Troponina C/metabolismo
7.
J Biol Chem ; 286(39): 34404-12, 2011 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-21832052

RESUMEN

TNNC1, which encodes cardiac troponin C (cTnC), remains elusive as a dilated cardiomyopathy (DCM) gene. Here, we report the clinical, genetic, and functional characterization of four TNNC1 rare variants (Y5H, M103I, D145E, and I148V), all previously reported by us in association with DCM (Hershberger, R. E., Norton, N., Morales, A., Li, D., Siegfried, J. D., and Gonzalez-Quintana, J. (2010) Circ. Cardiovasc. Genet. 3, 155-161); in the previous study, two variants (Y5H and D145E) were identified in subjects who also carried MYH7 and MYBPC3 rare variants, respectively. Functional studies using the recombinant human mutant cTnC proteins reconstituted into porcine papillary skinned fibers showed decreased Ca(2+) sensitivity of force development (Y5H and M103I). Furthermore, the cTnC mutants diminished (Y5H and I148V) or abolished (M103I) the effects of PKA phosphorylation on Ca(2+) sensitivity. Only M103I decreased the troponin activation properties of the actomyosin ATPase when Ca(2+) was present. CD spectroscopic studies of apo (absence of divalent cations)-, Mg(2+)-, and Ca(2+)/Mg(2+)-bound states indicated that all of the cTnC mutants (except I148V in the Ca(2+)/Mg(2+) condition) decreased the α-helical content. These results suggest that each mutation alters the function/ability of the myofilament to bind Ca(2+) as a result of modifications in cTnC structure. One variant (D145E) that was previously reported in association with hypertrophic cardiomyopathy and that produced results in vivo in this study consistent with prior hypertrophic cardiomyopathy functional studies was found associated with the MYBPC3 P910T rare variant, likely contributing to the observed DCM phenotype. We conclude that these rare variants alter the regulation of contraction in some way, and the combined clinical, molecular, genetic, and functional data reinforce the importance of TNNC1 rare variants in the pathogenesis of DCM.


Asunto(s)
Cardiomiopatía Dilatada/metabolismo , Mutación Missense , Miofibrillas/metabolismo , Troponina C/metabolismo , Animales , Cardiomiopatía Dilatada/genética , Femenino , Humanos , Masculino , Miofibrillas/genética , Estructura Secundaria de Proteína , Troponina C/química , Troponina C/genética
8.
J Biol Chem ; 286(23): 20901-12, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21502316

RESUMEN

A novel double deletion in cardiac troponin T (cTnT) of two highly conserved amino acids (Asn-100 and Glu-101) was found in a restrictive cardiomyopathic (RCM) pediatric patient. Clinical evaluation revealed the presence of left atrial enlargement and marked left ventricle diastolic dysfunction. The explanted heart examined by electron microscopy revealed myofibrillar disarray and mild fibrosis. Pedigree analysis established that this mutation arose de novo. The patient tested negative for six other sarcomeric genes. The single and double recombinant cTnT mutants were generated, and their functional consequences were analyzed in porcine skinned cardiac muscle. In the adult Tn environment (cTnT3 + cardiac troponin I), the single cTnT3-ΔN100 and cTnT3-ΔE101 mutations had opposing effects on the Ca(2+) sensitivity of force development compared with WT, whereas the double deletion cTnT3-ΔN100/ΔE101 increased the Ca(2+) sensitivity + 0.19 pCa units. In addition, cTnT3-ΔN100/ΔE101 decreased the cooperativity of force development, suggesting alterations in intrafilament protein-protein interactions. In the fetal Tn environment, (cTnT1 + slow skeletal troponin I), the single (cTnT1-ΔN110) and double (cTnT1-ΔN110/ΔE111) deletions did not change the Ca(2+) sensitivity compared with control. To recreate the patient's heterozygous genotype, we performed a reconstituted ATPase activity assay. Thin filaments containing 50:50 cTnT3-ΔN100/ΔE101:cTnT3-WT also increased the myofilament Ca(2+) sensitivity compared with WT. Co-sedimentation of thin filament proteins indicated that no significant changes occurred in the binding of Tn containing the RCM cTnT mutation to actin-Tm. This report reveals the protective role of Tn fetal isoforms as they rescue the increased Ca(2+) sensitivity produced by a cTnT-RCM mutation and may account for the lack of lethality during gestation.


Asunto(s)
Cardiomiopatía Restrictiva , Mutación INDEL , Contracción Miocárdica , Miocardio , Troponina T , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/patología , Adulto , Animales , Calcio/metabolismo , Cardiomiopatía Restrictiva/genética , Cardiomiopatía Restrictiva/metabolismo , Cardiomiopatía Restrictiva/patología , Niño , Preescolar , Femenino , Genotipo , Humanos , Masculino , Miocardio/metabolismo , Miocardio/patología , Linaje , Proteínas Recombinantes , Porcinos , Troponina T/genética , Troponina T/metabolismo
9.
J Biol Chem ; 285(36): 27785-97, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20566645

RESUMEN

Cardiac diseases associated with mutations in troponin subunits include hypertrophic cardiomyopathy (HCM), dilated cardiomyopathy (DCM), and restrictive cardiomyopathy (RCM). Altered calcium handling in these diseases is evidenced by changes in the Ca(2+) sensitivity of contraction. Mutations in the Ca(2+) sensor, troponin C (TnC), were generated to increase/decrease the Ca(2+) sensitivity of cardiac skinned fibers to create the characteristic effects of DCM, HCM, and RCM. We also used a reconstituted assay to determine the mutation effects on ATPase activation and inhibition. One mutant (A23Q) was found with HCM-like properties (increased Ca(2+) sensitivity of force and normal levels of ATPase inhibition). Three mutants (S37G, V44Q, and L48Q) were identified with RCM-like properties (a large increase in Ca(2+) sensitivity, partial loss of ATPase inhibition, and increased basal force). Two mutations were identified (E40A and I61Q) with DCM properties (decreased Ca(2+) sensitivity, maximal force recovery, and activation of the ATPase at high [Ca(2+)]). Steady-state fluorescence was utilized to assess Ca(2+) affinity in isolated cardiac (c)TnCs containing F27W and did not necessarily mirror the fiber Ca(2+) sensitivity. Circular dichroism of mutant cTnCs revealed a trend where increased alpha-helical content correlated with increased Ca(2+) sensitivity in skinned fibers and vice versa. The main findings from this study were as follows: 1) cTnC mutants demonstrated distinct functional phenotypes reminiscent of bona fide HCM, RCM, and DCM mutations; 2) a region in cTnC associated with increased Ca(2+) sensitivity in skinned fibers was identified; and 3) the F27W reporter mutation affected Ca(2+) sensitivity, maximal force, and ATPase activation of some mutants.


Asunto(s)
Calcio/metabolismo , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Fenotipo , Troponina C/metabolismo , Actomiosina/metabolismo , Adenosina Trifosfatasas/metabolismo , Animales , Calcio/farmacología , Activación Enzimática/efectos de los fármacos , Humanos , Ratones , Modelos Moleculares , Fibras Musculares Esqueléticas/metabolismo , Mutación , Contracción Miocárdica/efectos de los fármacos , Ingeniería de Proteínas , Estructura Secundaria de Proteína , Conejos , Troponina C/química , Troponina C/genética
10.
J Biol Chem ; 285(23): 17371-9, 2010 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-20371872

RESUMEN

In this study we explore the mechanisms by which a double mutation (E59D/D75Y) in cardiac troponin C (CTnC) associated with dilated cardiomyopathy reduces the Ca(2+)-activated maximal tension of cardiac muscle. Studying the single mutants (i.e. E59D or D75Y) indicates that D75Y, but not E59D, causes a reduction in the calcium affinity of CTnC in troponin complex, regulated thin filaments (RTF), and the Ca(2+) sensitivity of contraction and ATPase in cardiac muscle preparations. However, both D75Y and E59D are required to reduce the actomyosin ATPase activity and maximal force in muscle fibers, indicating that E59D enhances the effects of D75Y. Part of the reduction in force/ATPase may be due to a defect in the interactions between CTnC and cardiac troponin T, which are known to be necessary for ATPase activation. An additional mechanism for the reduction in force/ATPase comes from measurements of the binding stoichiometry of myosin subfragment-1 (S-1) to the RTF. Using wild type RTFs, 4.8 mol S-1 was bound per mol filament (seven actins), whereas with E59D/D75Y RTFs, the number of binding sites was reduced by approximately 23% to 3.7. Altogether, these results suggest that the reduction in force and ATPase activation is possibly due to a thin filament conformation that promotes fewer accessible S-1-binding sites. In the absence of any family segregation data, the functional results presented here support the concept that this is likely a dilated cardiomyopathy-causing mutation.


Asunto(s)
Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/metabolismo , Mutación , Troponina C/genética , Actinas/química , Adenosina Trifosfatasas/química , Animales , Sitios de Unión , Calcio/química , Activación Enzimática , Humanos , Contracción Muscular , Miosinas/química , Unión Proteica , Conformación Proteica , Porcinos
11.
J Clin Invest ; 118(12): 3893-903, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19033660

RESUMEN

In human cardiomyopathy, anatomical abnormalities such as hypertrophy and fibrosis contribute to the risk of ventricular arrhythmias and sudden death. Here we have shown that increased myofilament Ca2+ sensitivity, also a common feature in both inherited and acquired human cardiomyopathies, created arrhythmia susceptibility in mice, even in the absence of anatomical abnormalities. In mice expressing troponin T mutants that cause hypertrophic cardiomyopathy in humans, the risk of developing ventricular tachycardia was directly proportional to the degree of Ca2+ sensitization caused by the troponin T mutation. Arrhythmia susceptibility was reproduced with the Ca2+-sensitizing agent EMD 57033 and prevented by myofilament Ca2+ desensitization with blebbistatin. Ca2+ sensitization markedly changed the shape of ventricular action potentials, resulting in shorter effective refractory periods, greater beat-to-beat variability of action potential durations, and increased dispersion of ventricular conduction velocities at fast heart rates. Together these effects created an arrhythmogenic substrate. Thus, myofilament Ca2+ sensitization represents a heretofore unrecognized arrhythmia mechanism. The protective effect of blebbistatin provides what we believe to be the first direct evidence that reduction of Ca2+ sensitivity in myofilaments is antiarrhythmic and might be beneficial to individuals with hypertrophic cardiomyopathy.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Calcio/metabolismo , Cardiomiopatía Hipertrófica/metabolismo , Cardiotónicos/efectos adversos , Quinolinas/efectos adversos , Taquicardia Ventricular/metabolismo , Tiadiazinas/efectos adversos , Citoesqueleto de Actina/patología , Potenciales de Acción/efectos de los fármacos , Animales , Cardiomiopatía Hipertrófica/inducido químicamente , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/patología , Cardiomiopatía Hipertrófica/fisiopatología , Cardiotónicos/farmacología , Gatos , Muerte Súbita Cardíaca , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Fibrosis/inducido químicamente , Fibrosis/genética , Fibrosis/metabolismo , Fibrosis/patología , Fibrosis/fisiopatología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Masculino , Ratones , Ratones Mutantes , Quinolinas/farmacología , Factores de Riesgo , Taquicardia Ventricular/inducido químicamente , Taquicardia Ventricular/genética , Taquicardia Ventricular/patología , Taquicardia Ventricular/fisiopatología , Tiadiazinas/farmacología , Troponina T/genética , Troponina T/metabolismo
12.
J Mol Cell Cardiol ; 48(5): 882-92, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19914256

RESUMEN

Troponin (Tn) is a critical regulator of muscle contraction in cardiac muscle. Mutations in Tn subunits are associated with hypertrophic, dilated and restrictive cardiomyopathies. Improved diagnosis of cardiomyopathies as well as intensive investigation of new mouse cardiomyopathy models has significantly enhanced this field of research. Recent investigations have showed that the physiological effects of Tn mutations associated with hypertrophic, dilated and restrictive cardiomyopathies are different. Impaired relaxation is a universal finding of most transgenic models of HCM, predicted directly from the significant changes in Ca(2+) sensitivity of force production. Mutations associated with HCM and RCM show increased Ca(2+) sensitivity of force production while mutations associated with DCM demonstrate decreased Ca(2+) sensitivity of force production. This review spotlights recent advances in our understanding on the role of Tn mutations on ATPase activity, maximal force development and heart function as well as the correlation between the locations of these Tn mutations within the thin filament and myofilament function.


Asunto(s)
Cardiomiopatía Dilatada/genética , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Restrictiva/genética , Troponina/genética , Animales , Calcio/metabolismo , Cardiomiopatía Dilatada/etiología , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Hipertrófica/etiología , Cardiomiopatía Hipertrófica/metabolismo , Cardiomiopatía Restrictiva/etiología , Cardiomiopatía Restrictiva/metabolismo , Humanos , Mutación
13.
J Mol Cell Cardiol ; 49(3): 402-11, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20580639

RESUMEN

Several cardiac troponin I (cTnI) mutations are associated with restrictive cardiomyopathy (RCM) in humans. We have created transgenic mice (cTnI(193His) mice) that express the corresponding human RCM R192H mutation. Phenotype of this RCM animal model includes restrictive ventricles, biatrial enlargement and sudden cardiac death, which are similar to those observed in RCM patients carrying the same cTnI mutation. In the present study, we modified the overall cTnI in cardiac muscle by crossing cTnI(193His) mice with transgenic mice expressing an N-terminal truncated cTnI (cTnI-ND) that enhances relaxation. Protein analyses determined that wild type cTnI was replaced by cTnI-ND in the heart of double transgenic mice (Double TG), which express only cTnI-ND and cTnI R193H in cardiac myocytes. The presence of cTnI-ND effectively rescued the lethal phenotype of RCM mice by reducing the mortality rate. Cardiac function was significantly improved in Double TG mice when measured by echocardiography. The hypersensitivity to Ca(2+) and the prolonged relaxation of RCM cTnI(193His) cardiac myocytes were completely reversed by the presence of cTnI-ND in RCM hearts. The results demonstrate that myofibril hypersensitivity to Ca(2+) is a key mechanism that causes impaired relaxation in RCM cTnI mutant hearts and Ca(2+) desensitization by cTnI-ND can correct diastolic dysfunction and rescue the RCM phenotypes, suggesting that Ca(2+) desensitization in myofibrils is a therapeutic option for treatment of diastolic dysfunction without interventions directed at the systemic beta-adrenergic-PKA pathways.


Asunto(s)
Calcio/metabolismo , Cardiomiopatía Restrictiva/fisiopatología , Mutación/genética , Miocitos Cardíacos/patología , Troponina I/fisiología , Animales , Western Blotting , Diástole , Ecocardiografía , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , Fenotipo , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
J Biomed Biotechnol ; 2010: 350706, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20617149

RESUMEN

Mutations in sarcomeric proteins have recently been established as heritable causes of Restrictive Cardiomyopathy (RCM). RCM is clinically characterized as a defect in cardiac diastolic function, such as, impaired ventricular relaxation, reduced diastolic volume and increased end-diastolic pressure. To date, mutations have been identified in the cardiac genes for desmin, alpha-actin, troponin I and troponin T. Functional studies in skinned muscle fibers reconstituted with troponin mutants have established phenotypes consistent with the clinical findings which include an increase in myofilament Ca(2+) sensitivity and basal force. Moreover, when RCM mutants are incorporated into reconstituted myofilaments, the ability to inhibit the ATPase activity is reduced. A majority of the mutations cluster in specific regions of cardiac troponin and appear to be mutational "hot spots". This paper highlights the functional and clinical characteristics of RCM linked mutations within the troponin complex.


Asunto(s)
Cardiomiopatía Restrictiva/genética , Mutación , Troponina C/genética , Animales , Cardiomiopatía Restrictiva/fisiopatología , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Isoformas de Proteínas , Troponina C/fisiología
15.
J Biol Chem ; 2008 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-18245079

RESUMEN

This article was withdrawn by the authors before final publication on April 22, 2008.

16.
J Mol Cell Cardiol ; 45(2): 281-8, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18572189

RESUMEN

Hypertrophic Cardiomyopathy (HCM) is a common primary cardiac disorder defined by a hypertrophied left ventricle, is one of the main causes of sudden death in young athletes, and has been associated with mutations in most sarcomeric proteins (tropomyosin, troponin T and I, and actin, etc.). Many of these mutations appear to affect the functional properties of cardiac troponin C (cTnC), i.e., by increasing the Ca(2+)-sensitivity of contraction, a hallmark of HCM, yet surprisingly, prior to this report, cTnC had not been classified as a HCM-susceptibility gene. In this study, we show that mutations occurring in the human cTnC (HcTnC) gene (TNNC1) have the same prevalence (~0.4%) as well established HCM-susceptibility genes that encode other sarcomeric proteins. Comprehensive open reading frame/splice site mutation analysis of TNNC1 performed on 1025 unrelated HCM patients enrolled over the last 10 years revealed novel missense mutations in TNNC1: A8V, C84Y, E134D, and D145E. Functional studies with these recombinant HcTnC HCM mutations showed increased Ca(2+) sensitivity of force development (A8V, C84Y and D145E) and force recovery (A8V and D145E). These results are consistent with the HCM functional phenotypes seen with other sarcomeric-HCM mutations (E134D showed no changes in these parameters). This is the largest cohort analysis of TNNC1 in HCM that details the discovery of at least three novel HCM-associated mutations and more strongly links TNNC1 to HCM along with functional evidence that supports a central role for its involvement in the disease. This study may help to further define TNNC1 as an HCM-susceptibility gene, a classification that has already been established for the other members of the troponin complex.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/metabolismo , Predisposición Genética a la Enfermedad , Mutación Missense/genética , Troponina C/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Animales , Cardiomiopatía Hipertrófica/patología , Células Cultivadas , Estudios de Cohortes , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Datos de Secuencia Molecular , Ratas , Porcinos , Troponina C/clasificación , Troponina C/fisiología
17.
Biochem Biophys Res Commun ; 369(1): 74-81, 2008 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-18157941

RESUMEN

The troponin complex was discovered over thirty years ago and since then much insight has been gained into how this complex senses fluctuating levels of Ca(2+) and transmits this signal to the myofilament. Advances in genetics methods have allowed identification of mutations that lead to the phenotypically distinct cardiomyopathies: hypertrophic cardiomyopathy (HCM), restrictive cardiomyopathy (RCM) and dilated cardiomyopathy (DCM). This review serves to highlight key in vivo studies of mutation effects that have followed many years of functional studies and discusses how these mutations alter energetics and promote the characteristic remodeling associated with cardiomyopathic diseases. Studies have been performed that examine alterations in signaling and genomic methods have been employed to isolate upregulated proteins, however these processes are complex as there are multiple roads to hypertrophy or dilation associated with genetic cardiomyopathies. This review suggests future directions to explore in the troponin field that would heighten our understanding of the complex regulation of cardiac muscle contraction.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Cardiomiopatías/fisiopatología , Corazón/fisiopatología , Modelos Cardiovasculares , Contracción Miocárdica , Troponina/metabolismo , Animales , Humanos
18.
FASEB J ; 21(9): 2205-14, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17356007

RESUMEN

In skeletal muscle, the myosin molecule contains two sets of noncovalently attached low molecular weight proteins, the regulatory (RLC) and essential (ELC) light chains. To assess the functional and developmental significance of the fast skeletal isoform of the RLC (RLC-f), the murine fast skeletal RLC gene (Mylpf) was disrupted by homologous recombination. Heterozygotes containing an intronic neo cassette (RLC-/+) had approximately one-half of the amount of the RLC-f mRNA compared to wild-type (WT) mice but their muscles were histologically normal in both adults and neonates. In contrast, homozygous mice (RLC-/-) had no RLC-f mRNA or protein and completely lacked both fast and slow skeletal muscle. This was likely due to interference with mRNA processing in the presence of the neo cassette. These RLC-f null mice died immediately after birth, presumably due to respiratory failure since their diaphragms lacked skeletal muscle. The body weight of newborn RLC-f null mice was decreased 30% compared to heterozygous or WT newborn mice. The lack of skeletal muscle formation in the null mice did not affect the development of other organs including the heart. In addition, we found that WT mice did not express the ventricular/slow skeletal RLC isoform (RLC-v/s) until after birth, while it was expressed normally in the embryonic heart. The lack of skeletal muscle formation observed in RLC-f null mice indicates the total dependence of skeletal muscle development on the presence of RLC-f during embryogenesis. This observation, along with the normal function of the RLC-v/s in the heart, implicates a coupled, diverse pathway for RLC-v/s and RLC-f during embryogenesis, where RLC-v/s is responsible for heart development and RLC-f is necessary for skeletal muscle formation. In conclusion, in this study we demonstrate that the Mylpf gene is critically important for fast and slow skeletal muscle development.


Asunto(s)
Fibras Musculares de Contracción Rápida/patología , Fibras Musculares de Contracción Lenta/patología , Músculo Esquelético/anomalías , Cadenas Ligeras de Miosina/fisiología , Animales , Animales Recién Nacidos , Cruzamientos Genéticos , Femenino , Corazón Fetal/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Genes Letales , Genotipo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/embriología , Músculo Esquelético/patología , Miocardio/patología , Cadenas Ligeras de Miosina/deficiencia , Cadenas Ligeras de Miosina/genética , Reacción en Cadena de la Polimerasa
19.
Eur J Heart Fail ; 8(2): 115-21, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16214409

RESUMEN

BACKGROUND: The cardiac troponin T I79N mutation, linked to familial hypertrophic cardiomyopathy, carries a high risk of sudden cardiac death even in the absence of significant cardiac hypertrophy. The pathology underlying this mechanism has not yet been identified. AIMS: To study the underlying mechanism of this phenomenon we characterized the left ventricular (LV) performance of transgenic mice carrying the human troponin T mutation I79N under basal and isoproterenol-induced stress conditions. METHODS AND RESULTS: LV function was analyzed by recording pressure-volume loops using a microconductance catheter. Despite a hypercontractile systolic function under basal conditions TnT-I79N mice showed a diastolic dysfunction indicated by an increase in end-diastolic pressure-volume relationship (EDPVR), a load-independent factor of LV stiffness (0.06+/-0.01 vs. 0.02+/-0.01; P<0.05), when compared to mice expressing human wild-type troponin T (TnT-WT). TnT-I79N mutants developed severe diastolic heart failure and cardiac sudden death under isoproterenol stress. This was prevented after pretreatment with the L-type Ca2+ channel inhibitor diltiazem. CONCLUSIONS: Diastolic dysfunction due to increased LV stiffness in TnT-I79N mice leads to severe primary diastolic heart failure and finally to cardiac sudden death, which can be prevented by diltiazem.


Asunto(s)
Bloqueadores de los Canales de Calcio/uso terapéutico , Cardiomiopatía Hipertrófica Familiar/tratamiento farmacológico , Muerte Súbita Cardíaca/prevención & control , Diltiazem/uso terapéutico , Insuficiencia Cardíaca Diastólica/prevención & control , Análisis de Varianza , Animales , Presión Sanguínea/efectos de los fármacos , Bloqueadores de los Canales de Calcio/administración & dosificación , Bloqueadores de los Canales de Calcio/farmacología , Cardiomiopatía Hipertrófica Familiar/complicaciones , Cardiomiopatía Hipertrófica Familiar/genética , Colágeno/análisis , Colágeno/efectos de los fármacos , Diltiazem/administración & dosificación , Diltiazem/farmacología , Modelos Animales de Enfermedad , Insuficiencia Cardíaca Diastólica/etiología , Frecuencia Cardíaca/efectos de los fármacos , Ratones , Ratones Transgénicos , Mutación , Miocardio/química , Distribución Aleatoria , Resultado del Tratamiento , Troponina T/genética , Función Ventricular Izquierda/efectos de los fármacos
20.
Circ Res ; 92(4): 428-36, 2003 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-12600890

RESUMEN

The cardiac troponin T (TnT) I79N mutation has been linked to familial hypertrophic cardiomyopathy and high incidence of sudden death, despite causing little or no cardiac hypertrophy in patients. Transgenic mice expressing mutant human TnT (I79N-Tg) have increased cardiac contractility, but no ventricular hypertrophy or fibrosis. Enhanced cardiac function has been associated with myofilament Ca2+ sensitization, suggesting altered cellular Ca2+ handling. In the present study, we compare cellular Ca2+ transients and electrophysiological parameters of 64 I79N-Tg and 106 control mice in isolated myocytes, isolated perfused hearts, and whole animals. Ventricular action potentials (APs) measured in isolated I79N-Tg hearts and myocytes were significantly shortened only at 70% repolarization. No significant differences were found either in L-type Ca2+ or transient outward K+ currents, but inward rectifier K+ current (IK1) was significantly decreased. More critically, Ca2+ transients of field-stimulated ventricular I79N-Tg myocytes were reduced and had slow decay kinetics, consistent with increased Ca2+ sensitivity of I79N mutant fibers. AP differences were abolished when myocytes were dialyzed with Ca2+ buffers or after the Na+-Ca2+ exchanger was blocked by Li+. At higher pacing rates or in presence of isoproterenol, diastolic Ca2+ became significantly elevated in I79N-Tg compared with control myocytes. Ventricular ectopy could be induced by isoproterenol-challenge in isolated I79N-Tg hearts and anesthetized I79N-Tg mice. Freely moving I79N-Tg mice had a higher incidence of nonsustained ventricular tachycardia (VT) during mental stress (warm air jets). We conclude that the TnT-I79N mutation causes stress-induced VT even in absence of hypertrophy and/or fibrosis, arising possibly from the combination of AP remodeling related to altered Ca2+ transients and suppression of IK1.


Asunto(s)
Potenciales de Acción/fisiología , Calcio/metabolismo , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Taquicardia Ventricular/fisiopatología , Troponina T/genética , Potenciales de Acción/efectos de los fármacos , Anestesia , Animales , Presión Sanguínea/efectos de los fármacos , Cardiomiopatía Hipertrófica Familiar/genética , Cardiotónicos/farmacología , Electrocardiografía , Genotipo , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/fisiopatología , Humanos , Técnicas In Vitro , Isoproterenol/farmacología , Ratones , Ratones Transgénicos , Mutación , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Estrés Psicológico/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA