Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Circ Res ; 124(2): 292-305, 2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30582455

RESUMEN

RATIONALE: The Hippo pathway plays an important role in determining organ size through regulation of cell proliferation and apoptosis. Hippo inactivation and consequent activation of YAP (Yes-associated protein), a transcription cofactor, have been proposed as a strategy to promote myocardial regeneration after myocardial infarction. However, the long-term effects of Hippo deficiency on cardiac function under stress remain unknown. OBJECTIVE: We investigated the long-term effect of Hippo deficiency on cardiac function in the presence of pressure overload (PO). METHODS AND RESULTS: We used mice with cardiac-specific homozygous knockout of WW45 (WW45cKO), in which activation of Mst1 (Mammalian sterile 20-like 1) and Lats2 (large tumor suppressor kinase 2), the upstream kinases of the Hippo pathway, is effectively suppressed because of the absence of the scaffolding protein. We used male mice at 3 to 4 month of age in all animal experiments. We subjected WW45cKO mice to transverse aortic constriction for up to 12 weeks. WW45cKO mice exhibited higher levels of nuclear YAP in cardiomyocytes during PO. Unexpectedly, the progression of cardiac dysfunction induced by PO was exacerbated in WW45cKO mice, despite decreased apoptosis and activated cardiomyocyte cell cycle reentry. WW45cKO mice exhibited cardiomyocyte sarcomere disarray and upregulation of TEAD1 (transcriptional enhancer factor) target genes involved in cardiomyocyte dedifferentiation during PO. Genetic and pharmacological inactivation of the YAP-TEAD1 pathway reduced the PO-induced cardiac dysfunction in WW45cKO mice and attenuated cardiomyocyte dedifferentiation. Furthermore, the YAP-TEAD1 pathway upregulated OSM (oncostatin M) and OSM receptors, which played an essential role in mediating cardiomyocyte dedifferentiation. OSM also upregulated YAP and TEAD1 and promoted cardiomyocyte dedifferentiation, indicating the existence of a positive feedback mechanism consisting of YAP, TEAD1, and OSM. CONCLUSIONS: Although activation of YAP promotes cardiomyocyte regeneration after cardiac injury, it induces cardiomyocyte dedifferentiation and heart failure in the long-term in the presence of PO through activation of the YAP-TEAD1-OSM positive feedback mechanism.


Asunto(s)
Proteínas de Ciclo Celular/deficiencia , Desdiferenciación Celular , Insuficiencia Cardíaca/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Disfunción Ventricular Izquierda/metabolismo , Función Ventricular Izquierda , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis , Ciclo Celular , Proteínas de Ciclo Celular/genética , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Vía de Señalización Hippo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/patología , Oncostatina M/metabolismo , Fosfoproteínas/metabolismo , Ratas Wistar , Transducción de Señal , Factores de Transcripción de Dominio TEA , Factores de Transcripción/metabolismo , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología , Proteínas Señalizadoras YAP
2.
Biochem Eng J ; 131: 39-46, 2018 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-29736144

RESUMEN

Three-dimensional aggregation of human mesenchymal stem cells (hMSCs) has been used to enhance their therapeutic properties but current fabrication protocols depend on laboratory methods and are not scalable. In this study, we developed thermal responsive poly(N-isopropylacrylamide) grafted microcarriers (PNIPAM-MCs), which supported expansion and thermal detachment of hMSCs at reduced temperature (23.0 °C). hMSCs were cultured on the PNIPAM-MCs in both spinner flask (SF) and PBS Vertical-Wheel (PBS-VW) bioreactors for expansion. At room temperature, hMSCs were detached as small cell sheets, which subsequently self-assembled into 3D hMSC aggregates in PBS-VW bioreactor and remain as single cells in SF bioreactor owing to different hydrodynamic conditions. hMSC aggregates generated from the bioreactor maintained comparable immunomodulation and cytokine secretion properties compared to the ones made from the AggreWell®. The results of the current study demonstrate the feasibility of scale-up production of hMSC aggregates in the suspension bioreactor using thermal responsive microcarriers for integrated cell expansion and 3D aggregation in a close bioreactor system and highlight the critical role of hydrodynamics in self-assembly of detached hMSC in suspension.

4.
Cancer Res ; 67(19): 9055-65, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17909009

RESUMEN

Identification of genes with cancer-specific overexpression offers the potential to efficiently discover cancer-specific activities in an unbiased manner. We apply this paradigm to study mesothelin (MSLN) overexpression, a nearly ubiquitous, diagnostically and therapeutically useful characteristic of pancreatic cancer. We identified an 18-bp upstream enhancer, termed CanScript, strongly activating transcription from an otherwise weak tissue-nonspecific promoter and operating selectively in cells having aberrantly elevated cancer-specific MSLN transcription. Introducing mutations into CanScript showed two functionally distinct sites: an Sp1-like site and an MCAT element. Gel retardation and chromatin immunoprecipitation assays showed the MCAT element to be bound by transcription enhancer factor (TEF)-1 (TEAD1) in vitro and in vivo. The presence of TEF-1 was required for MSLN protein overexpression as determined by TEF-1 knockdown experiments. The cancer specificity seemed to be provided by a putative limiting cofactor of TEF-1 that could be outcompeted by exogenous TEF-1 only in a MSLN-overexpressing cell line. A CanScript concatemer offered enhanced activity. These results identify a TEF family member as a major regulator of MSLN overexpression, a fundamental characteristic of pancreatic and other cancers, perhaps due to an upstream and highly frequent aberrant cellular activity. The CanScript sequence represents a modular element for cancer-specific targeting, potentially suitable for nearly a third of human malignancies.


Asunto(s)
Proteínas de Unión al ADN/genética , Elementos de Facilitación Genéticos , Glicoproteínas de Membrana/biosíntesis , Proteínas Nucleares/genética , Factores de Transcripción/genética , Secuencia de Bases , Línea Celular Tumoral , ADN Complementario/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/metabolismo , Proteínas Ligadas a GPI , Células HeLa , Humanos , Inmunohistoquímica , Glicoproteínas de Membrana/genética , Mesotelina , Datos de Secuencia Molecular , Proteínas Nucleares/deficiencia , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción de Dominio TEA , Factores de Transcripción/deficiencia , Factores de Transcripción/metabolismo , Transcripción Genética , Transfección
5.
Biochemistry ; 47(51): 13711-25, 2008 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-19046074

RESUMEN

Native membrane sarcoplasmic reticulum (SR) Ca(2+)-ATPase isolated from skeletal muscle (SERCA1) exhibits oligomeric kinetic behavior [Mahaney, J. E., Thomas, D. D., and Froehlich, J. P. (2004) Biochemistry 43, 4400-4416]. In the present study we used quenched-flow mixing, electron paramagnetic resonance (EPR), and chemical cross-linking to probe for intermolecular interactions at physiological (0.1 M) and high (0.4 M) KCl. Exposure of SR membranes to water- and lipid-soluble cross-linking reagents revealed a mixture of SERCA1 oligomeric species consisting mainly of dimers and trimers. Titration of iodoacetamide spin-labeled SERCA1 with AMPPCP in the presence of 10 microM Ca(2+) and 0.1 M KCl revealed high- (K(D) = 45 microM) and low-affinity (K(D) = 315 microM) nucleotide binding sites in a 2:1 ratio, respectively. Raising the [KCl] to 0.4 M increased the fraction of weak binding sites and lowered the K(D) of the high-affinity component (20 microM). Phosphorylation by 10 microM ATP at 21 degrees C and 0.1 M KCl produced an early burst of P(i) production without a corresponding decline in the steady-state phosphoenzyme (EP) level. The steady-state EP level was twice as large as the P(i) burst and received equal contributions from E1P and E2P. Chasing the phosphoenzyme at 0.4 M KCl and 2 degrees C with ADP revealed a biphasic time course of E1P formation with a slow phase that matched the kinetics of the transient EPR signal from the spin-labeled Ca(2+)-ATPase. The absence of a fast component in the EPR signal excludes E1P as its source. Instead, it arises from a slow, KCl-dependent transformation at the start of the cycle which controls the formation of downstream intermediates with an increased mole fraction of rotationally restricted probes. We modeled this behavior with a SERCA1 trimer in which the formation of E1P/E2/E2P from E1ATP/E2P/E1P results from concerted transformations in the subunits coupling phosphorylation (E1ATP --> E1P + ADP) to dephosphorylation (E2P --> E2 + P(i)) and the conversion of E1P to E2P.


Asunto(s)
ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/química , Animales , Sitios de Unión , Catálisis , Dimerización , Espectroscopía de Resonancia por Spin del Electrón , Cinética , Músculo Esquelético/metabolismo , Fosforilación , Cloruro de Potasio/química , Unión Proteica , Conformación Proteica , Conejos , Retículo Sarcoplasmático/metabolismo , Factores de Tiempo
6.
Biochem J ; 388(Pt 1): 217-25, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15628970

RESUMEN

Members of the highly related TEF-1 (transcriptional enhancer factor-1) family (also known as TEAD, for TEF-1, TEC1, ABAA domain) bind to MCAT (muscle C, A and T sites) and A/T-rich sites in promoters active in cardiac, skeletal and smooth muscle, placenta, and neural crest. TEF-1 activity is regulated by interactions with transcriptional co-factors [p160, TONDU (Vgl-1, Vestigial-like protein-1), Vgl-2 and YAP65 (Yes-associated protein 65 kDa)]. The strong transcriptional co-activator YAP65 interacts with all TEF-1 family members, and, since YAP65 is related to TAZ (transcriptional co-activator with PDZ-binding motif), we wanted to determine if TAZ also interacts with members of the TEF-1 family. In the present study, we show by GST (glutathione S-transferase) pull-down assays, by co-immunoprecipitation and by modified mammalian two-hybrid assays that TEF-1 interacts with TAZ in vitro and in vivo. Electrophoretic mobility-shift assays with purified TEF-1 and GST-TAZ fusion protein showed that TAZ interacts with TEF-1 bound to MCAT DNA. TAZ can interact with endogenous TEF-1 proteins, since exogenous TAZ activated MCAT-dependent reporter promoters. Like YAP65, TAZ interacted with all four TEF-1 family members. GST pull-down assays with increasing amounts of [35S]TEF-1 and [35S]RTEF-1 (related TEF-1) showed that TAZ interacts more efficiently with TEF-1 than with RTEF-1. This differential interaction also extended to the interaction of TEF-1 and RTEF-1 with TAZ in vivo, as assayed by a modified mammalian two-hybrid experiment. These data show that differential association of TEF-1 proteins with transcriptional co-activators may regulate the activity of TEF-1 family members.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Factores de Transcripción/metabolismo , Aciltransferasas , Línea Celular Tumoral , Regulación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Familia de Multigenes , Proteínas Musculares/metabolismo , Regiones Promotoras Genéticas , Factores de Transcripción de Dominio TEA , Transactivadores , Transcripción Genética , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Proteínas Señalizadoras YAP
7.
Am J Physiol Heart Circ Physiol ; 293(4): H2320-7, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17644577

RESUMEN

In endothelial cells, NF-kappaB is an important intracellular signaling molecule by which changes in wall shear stress are transduced into the nucleus to initiate downstream endothelial nitric oxide synthase (NOS3) gene expression. We investigated whether NF-kappa light-chain gene enhancer in B cells 1 (NFKB1) promoter polymorphism ((-94)NFKB1 I/D, where I is the insertion allele and D is the deletion allele) was associated with 1) NOS3 gene expression in endothelial cells under physiological levels of unidirectional laminar shear stress (LSS) and 2) endothelial function in prehypertensive and stage I hypertensive individuals before and after a 6-mo supervised endurance exercise intervention. Competitive EMSAs revealed that proteins present in the nuclei of endothelial cells preferentially bound to the I allele NFKB1 promoter compared with the D allele. Reporter gene assays showed that the I allele promoter had significantly higher activity than the D allele. In agreement with these observations, homozygous II genotype cells had higher p50 expression levels than homozygous DD genotype cells. Cells with the homozygous II genotype showed a greater increase in NOS3 protein expression than did homozygous DD genotype cells under LSS. Functional experiments on volunteers confirmed higher baseline reactive hyperemic forearm blood flow, and, furthermore, the subgroup analysis revealed that DD homozygotes were significantly less prevalent in the exercise responder group compared with II and ID genotypes. We conclude that the (-94)NFKB1 I/D promoter variation contributes to the modulation of vascular function and adaptability to exercise-induced flow shear stress, most likely due to differences in NFKB1 gene transactivity.


Asunto(s)
Endotelio Vascular/fisiopatología , Terapia por Ejercicio , Hipertensión/genética , Subunidad p50 de NF-kappa B/genética , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Polimorfismo Genético , Regiones Promotoras Genéticas , Anciano , Células Cultivadas , Endotelio Vascular/enzimología , Endotelio Vascular/metabolismo , Inducción Enzimática , Femenino , Antebrazo/irrigación sanguínea , Predisposición Genética a la Enfermedad , Homocigoto , Humanos , Hiperemia/genética , Hiperemia/metabolismo , Hiperemia/fisiopatología , Hipertensión/enzimología , Hipertensión/metabolismo , Hipertensión/fisiopatología , Hipertensión/terapia , Masculino , Persona de Mediana Edad , Subunidad p50 de NF-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Fenotipo , Factores de Riesgo , Índice de Severidad de la Enfermedad , Estrés Mecánico , Transcripción Genética , Transfección , Resultado del Tratamiento , Vasodilatación
8.
J Biol Chem ; 277(27): 24346-52, 2002 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-11986313

RESUMEN

alpha(1)-Adrenergic signaling in cardiac myocytes activates the skeletal muscle alpha-actin gene through an MCAT cis-element, the binding site of the transcriptional enhancer factor-1 (TEF-1) family of transcription factors. TEF-1 accounts for more than 85% of the MCAT binding activity in neonatal rat cardiac myocytes. Other TEF-1 family members account for the rest. Although TEF-1 itself has little effect on the alpha(1)-adrenergic activation of skeletal muscle alpha-actin, the related factor RTEF-1 augments the response and is a target of alpha(1)-adrenergic signaling. Here, we examined another TEF-1 family member expressed in cardiac muscle, DTEF-1, and observed that it also augmented the alpha(1)-adrenergic response of skeletal muscle alpha-actin. A DTEF-1 peptide-specific antibody revealed that endogenous DTEF-1 accounts for up to 5% of the MCAT binding activity in neonatal rat cardiac myocytes. A TEF-1/DTEF-1 chimera suggests that alpha(1)-adrenergic signaling modulates DTEF-1 function. Orthophosphate labeling and immunoprecipitation of an epitope-tagged DTEF-1 showed that DTEF-1 is phosphorylated in vivo. alpha(1)-Adrenergic stimulation increased while phosphatase treatment lowered the MCAT binding by DTEF-1 and the endogenous non-TEF-1 MCAT-binding factor. In contrast, alpha(1)-adrenergic stimulation did not alter, and phosphatase treatment increased, MCAT binding of TEF-1 and RTEF-1. Taken together, these results suggest that DTEF-1 is a target for alpha(1)-adrenergic activation of the skeletal muscle alpha-actin gene in neonatal rat cardiac myocytes.


Asunto(s)
Agonistas alfa-Adrenérgicos/farmacología , Proteínas de Unión al ADN/metabolismo , Corazón/fisiología , Miocardio/citología , Receptores Adrenérgicos alfa 1/fisiología , Factores de Transcripción/metabolismo , Actinas/genética , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Secuencia de Bases , Núcleo Celular/efectos de los fármacos , Núcleo Celular/fisiología , Células Cultivadas , Cartilla de ADN , Corazón/efectos de los fármacos , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas , Ratas , Receptores Adrenérgicos alfa 1/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Factores de Transcripción de Dominio TEA , Transactivadores/metabolismo , Transfección
9.
J Mol Cell Cardiol ; 35(5): 461-71, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12738228

RESUMEN

A culture model for cardiac hypertrophy, stimulation of neonatal rat cardiac myocytes by alpha(1)-adrenergic agonists, has been used extensively to identify transcription factors that activate genes during cardiac hypertrophy, such as skeletal alpha-actin, beta-myosin heavy chain (betaMHC), and B-natriuretic peptide. We used this culture model to further investigate transcription factors regulating the betaMHC promoter in cardiac myocytes under basal conditions and during hypertrophy. We found that the rat betaMHC promoter contains two other MCAT sites, in addition to the two MCATs reported previously. The four MCAT sites are conserved in some but not all of the mammalian betaMHC promoters examined, and all bind TEF-1 but with varying affinity. As assayed by transient transfection into cardiac myocytes, the four MCATs within 348 bp of the transcription start site are required for full activity of the rat betaMHC promoter in the absence and presence of the alpha(1)-adrenergic agonist phenylephrine (PE). We found that the betaMHC promoter also contains a binding site for the NFAT family of transcription factors, which are activated by calcineurin and are implicated in the hypertrophic process. Although this site bound NFAT3 in vitro and has been reported to be required for betaMHC promoter activity in slow skeletal muscle, mutation of the site had no effect on basal or on PE-induced activity of the promoter in cardiac myocytes. Our results show that full activity of minimal betaMHC promoters in the presence and absence of hypertrophic agents requires multiple MCAT sites but not NFAT-binding sites.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 1 , Miocitos Cardíacos/metabolismo , Proteínas Nucleares , Regiones Promotoras Genéticas , Miosinas Ventriculares/genética , Animales , Secuencia de Bases , Sitios de Unión , Proteínas de Unión al ADN/metabolismo , Datos de Secuencia Molecular , Mutación , Factores de Transcripción NFATC , Ratas , Factores de Transcripción de Dominio TEA , Factores de Transcripción/metabolismo
10.
Am J Physiol Cell Physiol ; 286(3): C556-64, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14592812

RESUMEN

We evaluated various constructs to obtain cell-specific expression of the sarco(endo)plasmic reticulum Ca2+ -ATPase (SERCA) gene in cardiac myocytes after cDNA transfer by means of transfections or infections with adenovirus vectors. Expression of exogenous enhanced green fluorescent protein (EGFP) and SERCA genes was studied in cultured chicken embryo and neonatal rat cardiac myocytes, skeletal and smooth muscle cells, fibroblasts, and hepatocytes. Whereas the cytomegalovirus (CMV) promoter yielded high levels of protein expression in all cells studied, cardiac troponin T (cTnT) promoter segments demonstrated high specificity for cardiac myocytes. Their efficiency for protein expression was lower than that of the CMV promoter, but higher than that of cardiac myosin light chain or beta-myosin heavy chain promoter segments. A double virus system for Cre-dependent expression under control of the CMV promoter and Cre expression under control of a cardiac-specific promoter yielded high protein levels in cardiac myocytes, but only partial cell specificity due to significant Cre expression in hepatocytes. Specific intracellular targeting of gene products was demonstrated in situ by specific immunostaining of exogenous SERCA1 and endogenous SERCA2 and comparative fluorescence microscopy. The -374 cTnT promoter segment was the most advantageous of the promoters studied, producing cell-specific SERCA expression and a definite increase over endogenous Ca2+ -ATPase activity as well as faster removal of cytosolic calcium after membrane excitation. We conclude that analysis of promoter efficiency and cell specificity is of definite advantage when cell-specific expression of exogenous SERCA is wanted in cardiac myocytes after cDNA delivery to mixed cell populations.


Asunto(s)
Adenoviridae/genética , ATPasas Transportadoras de Calcio/genética , Regulación Enzimológica de la Expresión Génica , Vectores Genéticos , Miocitos Cardíacos/enzimología , Animales , Calcio/metabolismo , ATPasas Transportadoras de Calcio/metabolismo , Células Cultivadas , Embrión de Pollo , ADN Complementario , Proteínas Fluorescentes Verdes , Indicadores y Reactivos/metabolismo , Proteínas Luminiscentes/genética , Biología Molecular/métodos , Miocitos Cardíacos/citología , Regiones Promotoras Genéticas , Ratas , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico
11.
Am J Physiol Heart Circ Physiol ; 282(3): H872-9, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11834481

RESUMEN

Although tumor necrosis factor (TNF)-alpha is implicated in numerous cardiac pathologies, the intracellular events leading to its production by heart cells are largely unknown. The goal of the present study was to identify the role of the transcription factor nuclear factor (NF)-kappaB in this process. Among the many inducers of TNF-alpha expression in myeloid cells, only lipopolysaccharide (LPS) led to its induction in cultured neonatal myocytes. LPS also activated the NF-kappaB pathway, as evidenced by the degradation of the inhibitory protein IkappaB and the appearance of NF-kappaB-binding complexes in nuclear extracts. Furthermore, inhibitors of NF-kappaB activation, such as lactacystin, MG132, and pyrrolidine dithiocarbamate, were found to completely block the production of TNF-alpha in response to LPS stimulation, indicating a requirement of NF-kappaB for TNF-alpha expression. However, interleukin-1beta and phorbol 12-myristate 13-acetate also activated NF-kappaB but did not evoke TNF-alpha expression, revealing that this factor is not sufficient for cytokine production. Detailed examination of the NF-kappaB cascade revealed that cardiac cells displayed a unique pattern of IkappaB degradation in response to LPS, with IkappaBbeta but not IkappaBalpha being degraded upon stimulation. Additionally, two specific p65-containing DNA-binding complexes were observed in the nuclear extracts of neonatal cardiomyocytes: an inducible complex that is necessary for TNF-alpha expression and a constitutive species. Taken together, these results reveal that NF-kappaB is not only involved in cytokine production but also may be linked to other pathways that subserve a constitutive, protective mechanism for the heart cell.


Asunto(s)
Acetilcisteína/análogos & derivados , Endotoxinas/toxicidad , Corazón/fisiología , Lipopolisacáridos/toxicidad , FN-kappa B/metabolismo , Pirrolidinas/farmacología , Tiocarbamatos/farmacología , Factor de Necrosis Tumoral alfa/genética , Acetilcisteína/farmacología , Animales , Animales Recién Nacidos , Antioxidantes/farmacología , Western Blotting , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Cisteína Endopeptidasas/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Corazón/efectos de los fármacos , Ratones , Complejos Multienzimáticos/metabolismo , Miocardio/citología , Miocardio/metabolismo , Complejo de la Endopetidasa Proteasomal , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estrés Fisiológico , Transcripción Genética , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA