Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 183
Filtrar
1.
Nat Commun ; 13(1): 149, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013221

RESUMEN

Cachexia is associated with poor prognosis in chronic heart failure patients, but the underlying mechanisms of cachexia triggered disease progression remain poorly understood. Here, we investigate whether the dysregulation of myokine expression from wasting skeletal muscle exaggerates heart failure. RNA sequencing from wasting skeletal muscles of mice with heart failure reveals a reduced expression of Ostn, which encodes the secreted myokine Musclin, previously implicated in the enhancement of natriuretic peptide signaling. By generating skeletal muscle specific Ostn knock-out and overexpressing mice, we demonstrate that reduced skeletal muscle Musclin levels exaggerate, while its overexpression in muscle attenuates cardiac dysfunction and myocardial fibrosis during pressure overload. Mechanistically, Musclin enhances the abundance of C-type natriuretic peptide (CNP), thereby promoting cardiomyocyte contractility through protein kinase A and inhibiting fibroblast activation through protein kinase G signaling. Because we also find reduced OSTN expression in skeletal muscle of heart failure patients, augmentation of Musclin might serve as therapeutic strategy.


Asunto(s)
Caquexia/genética , Fibrosis Endomiocárdica/genética , Insuficiencia Cardíaca/genética , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Atrofia Muscular/genética , Factores de Transcripción/genética , 2',3'-Nucleótido Cíclico 3'-Fosfodiesterasa/genética , 2',3'-Nucleótido Cíclico 3'-Fosfodiesterasa/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Caquexia/metabolismo , Caquexia/fisiopatología , Caquexia/prevención & control , Estudios de Casos y Controles , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Fibrosis Endomiocárdica/metabolismo , Fibrosis Endomiocárdica/fisiopatología , Fibrosis Endomiocárdica/prevención & control , Femenino , Regulación de la Expresión Génica , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/prevención & control , Pruebas de Función Cardíaca , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/agonistas , Proteínas Musculares/antagonistas & inhibidores , Proteínas Musculares/deficiencia , Atrofia Muscular/metabolismo , Atrofia Muscular/fisiopatología , Atrofia Muscular/prevención & control , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Factores de Transcripción/agonistas , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/deficiencia
2.
Int J Mol Sci ; 22(18)2021 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-34575879

RESUMEN

Different forms of sudden cardiac death have been described, including a recently identified form of genetic arrhythmogenic disorder, named "Triadin KnockOut Syndrome" (TKOS). TKOS is associated with recessive mutations in the TRDN gene, encoding for TRIADIN, but the pathogenic mechanism underlying the malignant phenotype has yet to be completely defined. Moreover, patients with TKOS are often refractory to conventional treatment, substantiating the need to identify new therapeutic strategies in order to prevent or treat cardiac events. The zebrafish (Danio rerio) heart is highly comparable to the human heart in terms of functions, signal pathways and ion channels, representing a good model to study cardiac disorders. In this work, we generated the first zebrafish model for trdn loss-of-function, by means of trdn morpholino injections, and characterized its phenotype. Although we did not observe any gross cardiac morphological defect between trdn loss-of-function embryos and controls, we found altered cardiac rhythm that was recovered by the administration of arrhythmic drugs. Our model will provide a suitable platform to study the effect of TRDN mutations and to perform drug screening to identify new pharmacological strategies for patients carrying TRDN mutations.


Asunto(s)
Muerte Súbita Cardíaca/etiología , Modelos Animales de Enfermedad , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Proteínas Musculares/deficiencia , Animales , Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/genética , Proteínas Portadoras , Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Mutación con Pérdida de Función , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Fenotipo , Síndrome , Pez Cebra
4.
Int J Mol Sci ; 22(14)2021 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-34298987

RESUMEN

Limb-girdle muscular dystrophy R1 calpain 3-related (LGMDR1) is an autosomal recessive muscular dystrophy produced by mutations in the CAPN3 gene. It is a rare disease and there is no cure or treatment for the disease while the pathophysiological mechanism by which the absence of calpain 3 provokes the dystrophy in muscles is not clear. However, key proteins implicated in Wnt and mTOR signaling pathways, which regulate muscle homeostasis, showed a considerable reduction in their expression and in their phosphorylation in LGMDR1 patients' muscles. Finally, the administration of tideglusib and VP0.7, ATP non-competitive inhibitors of glycogen synthase kinase 3ß (GSK-3ß), restore the expression and phosphorylation of these proteins in LGMDR1 cells, opening the possibility of their use as therapeutic options.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Distrofia Muscular de Cinturas/tratamiento farmacológico , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Sitio Alostérico/efectos de los fármacos , Antígeno CD56/análisis , Calpaína/deficiencia , Calpaína/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/química , Humanos , Hidrazinas/farmacología , Hidrazinas/uso terapéutico , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Distrofia Muscular de Cinturas/enzimología , Proteínas del Tejido Nervioso/química , Fosforilación , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/fisiología , Quinolonas/farmacología , Quinolonas/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/fisiología , Tiadiazoles/farmacología , Tiadiazoles/uso terapéutico , Vía de Señalización Wnt/efectos de los fármacos
5.
FEBS Open Bio ; 11(6): 1731-1738, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33932144

RESUMEN

Nonreceptor tyrosine kinase c-Abl participates in several cellular processes by phosphorylating transcription factors or cofactors. c-Abl binds and phosphorylates four-and-a-half-LIM-only protein 2 (FHL2), but the identity of the phosphorylation sites and their contribution to cell cycle regulation is unclear. In this study, we demonstrate that c-Abl highly phosphorylates FHL2 at Y97, Y176, Y217, and Y236 through mass spectrometry and tyrosine-to-phenylalanine (Y â†’ F) mutant analysis. Proliferation was inhibited in cells expressing wild-type (WT) FHL2 but not cells expressing the phosphorylation-defective mutant FHL2(4YF). Moreover, FHL2 contributed to cell cycle arrest at G2/M induced by ionizing radiation (IR). FHL2 WT but not FHL2(4YF) rescued FHL2 function in FHL2-depleted cells by causing IR-induced G2/M arrest. These results demonstrate that c-Abl regulates cell cycle progression by phosphorylating FHL2.


Asunto(s)
Proteínas con Homeodominio LIM/metabolismo , Proteínas Musculares/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Factores de Transcripción/metabolismo , Proliferación Celular , Células Cultivadas , Puntos de Control de la Fase G2 del Ciclo Celular , Humanos , Proteínas con Homeodominio LIM/deficiencia , Proteínas Musculares/deficiencia , Fosforilación , Radiación Ionizante , Factores de Transcripción/deficiencia
6.
Oxid Med Cell Longev ; 2021: 5564884, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33859778

RESUMEN

Vascular smooth muscle cell (VSMC) apoptosis is a major defining feature of abdominal aortic aneurysm (AAA) and mainly caused by inflammatory cell infiltration. Smooth muscle (SM) 22α prevents AAA formation through suppressing NF-κB activation. However, the role of SM22α in VSMC apoptosis is controversial. Here, we identified that SM22α loss contributed to apoptosis of VSMCs via activation of macrophages. Firstly, deficiency of SM22α enhanced the interaction of VSMCs with macrophages. Macrophages were retained and activated by Sm22α -/- VSMCs via upregulating VCAM-1 expression. The ratio of apoptosis was increased by 1.62-fold in VSMCs treated with the conditional media (CM) from activated RAW264.7 cells, compared to that of the control CM (P < 0.01), and apoptosis of Sm22α -/- VSMCs was higher than that of WT VSMCs (P < 0.001). Next, circRasGEF1B from activated macrophages was delivered into VSMCs promoting ZFP36 expression via stabilization of ZFP36 mRNA. Importantly, circRasGEF1B, as a scaffold, guided ZFP36 to preferentially bind to and decay Bcl-2 mRNA in a sequence-specific manner and triggered apoptosis of VSMCs, especially in Sm22α -/- VSMCs. These findings reveal a novel mechanism by which the circRasGEF1B-ZFP36 axis mediates macrophage-induced VSMC apoptosis via decay of Bcl-2 mRNA, whereas Sm22α -/- VSMCs have a higher sensitivity to apoptosis.


Asunto(s)
Macrófagos/citología , Macrófagos/metabolismo , Proteínas de Microfilamentos/deficiencia , Proteínas Musculares/deficiencia , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , ARN Circular/metabolismo , Animales , Apoptosis/fisiología , Comunicación Celular/fisiología , Técnicas de Reprogramación Celular , Humanos , Masculino , Ratones , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Células RAW 264.7 , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Tristetraprolina/biosíntesis , Tristetraprolina/genética , Tristetraprolina/metabolismo
7.
Toxicol Lett ; 345: 54-60, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-33872746

RESUMEN

Congenital clubfoot (CCF) is a common birth defect. Maternal smoking during pregnancy increases the risk of CCF. In previous research, we found muscle phenotypes similar to CCF in four and a half LIM domain protein 1 (FHLI) offspring mice (FHL1-/y). However, the role of P2RX7-mediated pyroptosis in the effect of cigarette smoke (CS) on the skeletal muscle of FHL1-/y mice during pregnancy is unclear. In the present study, pregnant mice at 11 days of gestation were exposed to CS and male offspring of wild-type (WT) and FHL1-/y mice were divided into four groups (Control-WT, Control-KO, CS-WT, CS-KO). The histomorphology of lower limb muscles was examined using hematoxylin and eosin (H&E) staining. P2RX7, indicators of pyroptosis (NLRP3, ASC, cleaved-caspase 1, IL-1ß), and cytoskeletal proteins (MYBPC2, LDB3) were also detected using immunoblotting. CS exposure during pregnancy aggravated the muscle phenotype similar to CCF in FHL1-/y offspring mice. FHL1 gene knockout (KO) or CS exposure during pregnancy each activated the expression of P2RX7, cell pyroptosis-related proteins (NLRP3, ASC, cleaved-caspase 1, IL-1ß), a muscle injury marker (MYOD1), and cytoskeletal proteins (MYBPC2, LDB3); these two factors had an additive effect. The results showed maternal smoking during pregnancy aggravated muscle phenotype similar to CCF in FHL1-/y offspring mice through P2RX7-mediated pyroptosis.


Asunto(s)
Pie Equinovaro/metabolismo , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Proteínas con Dominio LIM/deficiencia , Proteínas Musculares/deficiencia , Músculo Esquelético/metabolismo , Efectos Tardíos de la Exposición Prenatal , Receptores Purinérgicos P2X7/metabolismo , Fumar/efectos adversos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas Portadoras/metabolismo , Pie Equinovaro/genética , Pie Equinovaro/patología , Femenino , Edad Gestacional , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Masculino , Exposición Materna/efectos adversos , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/genética , Músculo Esquelético/patología , Proteína MioD/metabolismo , Fenotipo , Embarazo , Piroptosis , Transducción de Señal
8.
In Vitro Cell Dev Biol Anim ; 56(8): 604-613, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32914385

RESUMEN

Knockout of multifunction gene cysteine- and glycine-rich protein 3 (CSRP3) in cardiomyocytes (CMs) of mice leads to heart dilation, severely affecting its functions. In humans, CSRP3 mutations are associated with hypertrophic (HCM) and dilated cardiomyopathy (DCM). The absence of the CSRP3 expression produces unknown effects on in vitro neonatal CMs' metabolism. The metabolome changes in culture media conditioned by CSRP3 knockout (KO-CSRP3), and wild type (WT) neonatal cardiomyocytes were investigated under untreated or after metabolic challenging conditions produced by isoproterenol (ISO) stimulation, by in vitro high-resolution proton magnetic resonance spectroscopy (1H-MRS)-based metabolomics. Metabolic differences between neonatal KO-CSRP3 and WT rats' CMs were identified. After 72 h of culture, ISO administration was associated with increased CMs' energy requirements and increased levels of threonine, alanine, and 3-hydroxybutyrate in both neonatal KO-CSRP3 and WT CMs conditioned media. When compared with KO-CSRP3, culture media derived from WT cells presented higher lactate concentrations either under basal or ISO-stimulated conditions. The higher activity of ketogenic biochemical pathways met the elevated energy requirements of the contractile cells. Both cells are considered phenotypically indistinguishable in the neonatal period of animal lives, but the observed metabolic stress responses of KO-CSRP3 and WT CMs to ISO were different. KO-CSRP3 CMs produced less lactate than WT CMs in both basal and stimulated conditions. Mainly, ISO-stimulated conditions produced evidence for lactate overload within KO-CSRP3 CMs, while WT CMs succeeded to manage the metabolic stress. Thus, 1H-MRS-based metabolomics was suitable to identify early inefficient energetic metabolism in neonatal KO-CSRP3 CMs. These results may reflect an apparent lower lactate transport and consumption, in association with protein catabolism.


Asunto(s)
Medios de Cultivo/química , Proteínas con Dominio LIM/metabolismo , Proteínas Musculares/metabolismo , Miocitos Cardíacos/metabolismo , Espectroscopía de Protones por Resonancia Magnética , Animales , Animales Recién Nacidos , Forma de la Célula , Análisis Discriminante , Isoproterenol/farmacología , Proteínas con Dominio LIM/deficiencia , Análisis de los Mínimos Cuadrados , Proteínas Musculares/deficiencia , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Ratas , Estadística como Asunto
9.
Biochem Biophys Res Commun ; 529(4): 1086-1093, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32819569

RESUMEN

Non-small cell lung cancer (NSCLC) is the leading cause of tumor mortality worldwide. However, the mechanisms underlying NSCLC tumorigenesis are incompletely understood. TAGLN, also named SM22, as a member of the calponin family, is highly expressed in many types of tumors. Nevertheless, its effects on NSCLC progression remain unclear. In this study, we found that TAGLN was over-expressed in tumor tissues of NSCLC patients and cell lines. Additionally, NSCLC patients with high expression showed worse overall survival rate. Then, gene silencing results indicated that TAGLN knockdown markedly inhibited proliferation and induced apoptosis in NSCLC cells, while rescue study exhibited opposite results. Moreover, suppressing TAGLN significantly reduced migration and invasion of NSCLC cells, and its over-expression promoted the migratory and invasive activities of NSCLC cells. The in vivo studies confirmed the oncogenic roles of TAGLN in NSCLC, along with clearly elevated metastasis. Notably, these effects were abrogated in mice with TAGLN deletion. Finally, we found that TAGLN knockdown could improve the sensitivity of NSCLC cells to sorafenib (SFB) and 5-FU treatment, further suppressing the proliferation, migration and invasion of NSCLC cells. Consistently, TAGLN deletion attenuated tumor xenografts growth and metastasis of NSCLC in mouse models by enhancing the anti-cancer effects of SFB and 5-FU. Altogether, these findings demonstrated that TAGLN functioned as an oncogene as well as a chemotherapeutic regulator during NSCLC development, which suggested a potential therapeutic strategy for NSCLC treatment mainly through repressing TAGLN expression.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Proteínas de Microfilamentos/deficiencia , Proteínas Musculares/deficiencia , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Fluorouracilo/farmacología , Fluorouracilo/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/genética , Masculino , Ratones Endogámicos BALB C , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , Sorafenib/farmacología , Sorafenib/uso terapéutico , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
10.
Biochem Biophys Res Commun ; 529(4): 884-889, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32819594

RESUMEN

Smooth muscle 22α (SM22α, namely Transgelin), as an actin-binding protein, regulates the contractility of vascular smooth muscle cells (VSMCs) by modulation of the stress fiber formation. However, little is known about the roles of SM22α in the regulation of uterine contraction during parturition. Here, we showed that contraction in response to oxytocin (OT) was significantly decreased in the uterine muscle strips from SM22α knockout (Sm22α-KO) mice, especially at full-term pregnancy, which may be resulted from impaired formation of stress fibers. Furthermore, serious mitochondrial damage such as the mitochondrial swelling, cristae disruption and even disappearance were observed in the myometrium of Sm22α-KO mice at full-term pregnancy, eventually resulting in the collapse of mitochondrial membrane potential and impairment in ATP synthesis. Our data indicate that SM22α is necessary to maintain uterine contractility at delivery in mice, and acts as a novel target for preventive or therapeutic manipulation of uterine atony during parturition.


Asunto(s)
Proteínas de Microfilamentos/genética , Proteínas Musculares/genética , Músculo Liso Vascular/efectos de los fármacos , Miometrio/efectos de los fármacos , Oxitocina/farmacología , Contracción Uterina/efectos de los fármacos , Inercia Uterina/genética , Adenosina Trifosfato/deficiencia , Animales , Femenino , Regulación de la Expresión Génica , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/deficiencia , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/metabolismo , Dilatación Mitocondrial/genética , Proteínas Musculares/deficiencia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Miometrio/metabolismo , Miometrio/patología , Parto , Embarazo , Cultivo Primario de Células , Fibras de Estrés/efectos de los fármacos , Fibras de Estrés/metabolismo , Fibras de Estrés/patología , Técnicas de Cultivo de Tejidos , Inercia Uterina/metabolismo , Inercia Uterina/patología
11.
Arterioscler Thromb Vasc Biol ; 40(10): 2425-2439, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32787520

RESUMEN

OBJECTIVE: The calcineurin-NFAT (nuclear factor for activated T cells)-DSCR (Down syndrome critical region)-1 pathway plays a crucial role as the downstream effector of VEGF (vascular endothelial growth factor)-mediated tumor angiogenesis in endothelial cells. A role for DSCR-1 in different organ microenvironment such as the cornea and its role in ocular diseases is not well understood. Corneal changes can be indicators of various disease states and are easily detected through ocular examinations. Approach and Results: The presentation of a corneal arcus or a corneal opacity due to lipid deposition in the cornea often indicates hyperlipidemia and in most cases, hypercholesterolemia. Although the loss of Apo (apolipoprotein) E has been well characterized and is known to lead to elevated serum cholesterol levels, there are few corneal changes observed in ApoE-/- mice. In this study, we show that the combined loss of ApoE and DSCR-1 leads to a dramatic increase in serum cholesterol levels and severe corneal opacity with complete penetrance. The cornea is normally maintained in an avascular state; however, loss of Dscr-1 is sufficient to induce hyper-inflammatory and -oxidative condition, increased corneal neovascularization, and lymphangiogenesis. Furthermore, immunohistological analysis and genome-wide screening revealed that loss of Dscr-1 in mice triggers increased immune cell infiltration and upregulation of SDF (stromal derived factor)-1 and its receptor, CXCR4 (C-X-C motif chemokine ligand receptor-4), potentiating this signaling axis in the cornea, thereby contributing to pathological corneal angiogenesis and opacity. CONCLUSIONS: This study is the first demonstration of the critical role for the endogenous inhibitor of calcineurin, DSCR-1, and pathological corneal angiogenesis in hypercholesterolemia induced corneal opacity.


Asunto(s)
Proteínas de Unión al Calcio/deficiencia , Neovascularización de la Córnea/etiología , Opacidad de la Córnea/etiología , Células Endoteliales/metabolismo , Endotelio Corneal/metabolismo , Hipercolesterolemia/complicaciones , Proteínas Musculares/deficiencia , Animales , Proteínas de Unión al Calcio/genética , Quimiocina CXCL12/metabolismo , Quimiotaxis de Leucocito , Neovascularización de la Córnea/genética , Neovascularización de la Córnea/metabolismo , Neovascularización de la Córnea/patología , Opacidad de la Córnea/genética , Opacidad de la Córnea/metabolismo , Opacidad de la Córnea/patología , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Endoteliales/patología , Endotelio Corneal/patología , Infecciones Fúngicas del Ojo/metabolismo , Infecciones Fúngicas del Ojo/patología , Células HEK293 , Humanos , Hipercolesterolemia/genética , Hipercolesterolemia/metabolismo , Linfangiogénesis , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Estrés Oxidativo , Receptores CXCR4/metabolismo , Transducción de Señal , Síndrome de Stevens-Johnson/metabolismo , Síndrome de Stevens-Johnson/patología , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
Circ Genom Precis Med ; 13(2): e002731, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32167373

RESUMEN

BACKGROUND: Triadin knockout syndrome (TKOS) is a potentially lethal arrhythmia disorder caused by recessively inherited null variants in TRDN-encoded cardiac triadin. Despite its malignant phenotype, the prevalence of TKOS in sudden infant death syndrome and sudden unexplained death in the young is unknown. METHODS: Exome sequencing was performed on 599 sudden infant death syndrome and 258 sudden unexplained death in the young cases. Allele frequencies of all TRDN null variants identified in the cardiac-specific isoform of TRDN in the Genome Aggregation Database were used to determine the estimated prevalence and ethnic distribution of TKOS. RESULTS: No triadin null individuals were identified in 599 sudden infant death syndrome and 258 sudden unexplained death in the young exomes. Using the Genome Aggregation Database, we estimate the overall prevalence of TKOS to be ≈1:22.7 million individuals. However, TKOS prevalence is 5.5-fold higher in those of African descent (≈1:4.1 million). CONCLUSIONS: TKOS is an exceedingly rare clinical entity that does not contribute meaningfully to either sudden infant death syndrome or sudden unexplained death in the young. However, despite its rarity and absence in large sudden death cohorts, TKOS remains a malignant and potentially lethal disorder which requires further research to better care for these patients.


Asunto(s)
Arritmias Cardíacas/patología , Muerte Súbita Cardíaca/patología , Predisposición Genética a la Enfermedad , Proteínas Musculares/deficiencia , Muerte Súbita del Lactante/patología , Adolescente , Adulto , Arritmias Cardíacas/epidemiología , Arritmias Cardíacas/genética , Proteínas Portadoras/genética , Niño , Preescolar , Estudios de Cohortes , Muerte Súbita Cardíaca/epidemiología , Exoma , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Proteínas Musculares/genética , Fenotipo , Muerte Súbita del Lactante/epidemiología , Muerte Súbita del Lactante/genética , Síndrome , Estados Unidos/epidemiología , Adulto Joven
13.
Nat Commun ; 11(1): 484, 2020 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-31980627

RESUMEN

ß-Cell dysfunction and reduction in ß-cell mass are hallmark events of diabetes mellitus. Here we show that ß-cells express abundant Kindlin-2 and deleting its expression causes severe diabetes-like phenotypes without markedly causing peripheral insulin resistance. Kindlin-2, through its C-terminal region, binds to and stabilizes MafA, which activates insulin expression. Kindlin-2 loss impairs insulin secretion in primary human and mouse islets in vitro and in mice by reducing, at least in part, Ca2+ release in ß-cells. Kindlin-2 loss activates GSK-3ß and downregulates ß-catenin, leading to reduced ß-cell proliferation and mass. Kindlin-2 loss reduces the percentage of ß-cells and concomitantly increases that of α-cells during early pancreatic development. Genetic activation of ß-catenin in ß-cells restores the diabetes-like phenotypes induced by Kindlin-2 loss. Finally, the inducible deletion of ß-cell Kindlin-2 causes diabetic phenotypes in adult mice. Collectively, our results establish an important function of Kindlin-2 and provide a potential therapeutic target for diabetes.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Proteínas Musculares/metabolismo , beta Catenina/metabolismo , Animales , Proliferación Celular , Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/genética , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Insulina/genética , Resistencia a la Insulina , Islotes Pancreáticos/crecimiento & desarrollo , Islotes Pancreáticos/metabolismo , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fenotipo , Estabilidad Proteica , beta Catenina/genética
14.
Cells ; 9(1)2020 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-31963815

RESUMEN

Cholestasis occurs in different clinical circumstances and leads to severe hepatic disorders. The four-and-a-half LIM-domain protein 2 (FHL2) is a scaffolding protein that modulates multiple signal transduction pathways in a tissue- and cell context-specific manner. In this study, we aimed to gain insight into the function of FHL2 in cholestatic liver injury. FHL2 expression was significantly increased in the bile duct ligation (BDL) model in mice. In Fhl2-deficient (Fhl2-ko) mice, BDL caused a more severe portal and parenchymal inflammation, extended portal fibrosis, higher serum transaminase levels, and higher pro-inflammatory and pro-fibrogenic gene expression compared to wild type (wt) mice. FHL2 depletion in HepG2 cells with siRNA resulted in a higher expression of the bile acid transporter Na+-taurocholate cotransporting polypeptide (NTCP) gene. Furthermore, FHL2-depleted HepG2 cells showed higher expression of markers for oxidative stress, lower B-cell lymphoma 2 (Bcl2) expression, and higher Bcl2-associated X protein (BAX) expression after stimulation with deoxycholic acid (DCA). In hepatic stellate cells (HSCs), FHL2 depletion caused an increased expression of TGF-ß and several pro-fibrogenic matrix metalloproteinases. In summary, our study shows that deficiency in FHL2 aggravates cholestatic liver injury and suggests FHL2-mediated effects on bile acid metabolisms and HSCs as potential mechanisms for pronounced hepatocellular injury and fibrosis.


Asunto(s)
Colestasis/metabolismo , Colestasis/patología , Proteínas con Homeodominio LIM/deficiencia , Hígado/lesiones , Proteínas Musculares/deficiencia , Factores de Transcripción/deficiencia , Animales , Ácidos y Sales Biliares/metabolismo , Conductos Biliares/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Células Hep G2 , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Humanos , Inflamación/patología , Proteínas con Homeodominio LIM/metabolismo , Ligadura , Hígado/patología , Cirrosis Hepática/patología , Masculino , Ratones Noqueados , Proteínas Musculares/metabolismo , Factores de Transcripción/metabolismo
15.
Vascul Pharmacol ; 125-126: 106634, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31866461

RESUMEN

Despite the advent of new-generation drug-eluting stents, in-stent restenosis remains a significant problem in patients with coronary artery disease. In- stent restenosis is defined as the gradual re-narrowing of a stented coronary artery lesion due to arterial damage with subsequent local inflammation of the vessel wall and excessive growth of the vascular smooth muscle cells (vSMCs). Four-and-a-half LIM-domain protein 2 (FHL2) is a scaffold protein involved in regulating vSMC function and inflammation. Previously we have demonstrated that FHL2 prevents vSMC proliferation in a murine carotid artery ligation model. However, the effect of FHL2 on the inflammatory response of the vSMCs is not investigated. Therefore, we studied the inflammatory response in the vessel wall of FHL2-deficient (-KO) mice after carotid artery ligation. We found that circulating cytokines and local macrophage infiltration in the ligated carotid vessels were increased in FHL2-KO mice after carotid artery ligation. Moreover, FHL2-KO vSMCs showed increased secretion of cytokines such as SDF-1α and RANTES, and enhanced activation of the NFκB pathway. Finally, we found that blocking the NFκB signalling pathway abrogated this pro-inflammatory state in FHL2-KO vSMCs. Taken together, our results demonstrate that FHL2 decreases the inflammatory response of vSMCs through inhibition of the NFkB-signalling pathway.


Asunto(s)
Enfermedades de las Arterias Carótidas/metabolismo , Inflamación/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Proteínas Musculares/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , FN-kappa B/metabolismo , Factores de Transcripción/metabolismo , Animales , Antiinflamatorios/farmacología , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/prevención & control , Células Cultivadas , Citocinas/sangre , Modelos Animales de Enfermedad , Inflamación/genética , Inflamación/patología , Inflamación/prevención & control , Proteínas con Homeodominio LIM/deficiencia , Proteínas con Homeodominio LIM/genética , Macrófagos/metabolismo , Macrófagos/patología , Ratones Noqueados , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , Transducción de Señal , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
16.
Exp Mol Pathol ; 113: 104363, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31881201

RESUMEN

Non-alcoholic steatohepatitis (NASH) is characterized by immune cell infiltration. Loss of the scaffold protein alpha-syntrophin (SNTA) protected mice from hepatic inflammation in the methionine-choline-deficient (MCD) diet model. Here, we determined increased numbers of macrophages and CD8+ T-cells in MCD diet induced NASH liver of wild type mice. In the mutant animals these NASH associated changes in immune cell composition were less pronounced. Further, there were more γδ T-cells in the NASH liver of the null mice. Galectin-3 protein in the hepatic non-parenchymal cell fraction was strongly induced in MCD diet fed wild type but not mutant mice. Antioxidant enzymes declined in NASH liver with no differences between the genotypes. To identify the target cells responsive to SNTA loss in-vitro experiments were performed. In the human hepatic stellate cell line LX-2, SNTA did not regulate pro-fibrotic or antioxidant proteins like alpha-smooth muscle actin or catalase. Soluble galectin-3 was, however, reduced upon SNTA knock-down and increased upon SNTA overexpression. SNTA deficiency neither affected cell proliferation nor cell death of LX-2 cells. In the macrophage cell line RAW264.7 low SNTA indeed caused higher galectin-3 production whereas release of TNF and cell viability were normal. Moreover, SNTA had no effect on hepatocyte chemerin and CCL2 expression. Overall, SNTA loss improved NASH without causing major effects in macrophage, hepatocyte and hepatic stellate cell lines. SNTA null mice fed the MCD diet had less body weight loss and this seems to contribute to improved liver health of the mutant mice.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Proteínas de Unión al Calcio/deficiencia , Galectina 3/metabolismo , Hígado/patología , Macrófagos/patología , Proteínas de la Membrana/deficiencia , Proteínas Musculares/deficiencia , Enfermedad del Hígado Graso no Alcohólico/inmunología , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Actinas/metabolismo , Animales , Antioxidantes/metabolismo , Peso Corporal , Proteínas de Unión al Calcio/metabolismo , Catalasa/metabolismo , Línea Celular , Proliferación Celular , Supervivencia Celular , Quimiocina CCL2/metabolismo , Quimiocinas/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Ácido Graso Sintasas/metabolismo , Conducta Alimentaria , Hemo-Oxigenasa 1/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interleucina-6/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Macrófagos/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Proteínas Musculares/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Superóxido Dismutasa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
17.
Skelet Muscle ; 9(1): 26, 2019 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-31666122

RESUMEN

BACKGROUND: Skeletal muscle mass and strength are crucial determinants of health. Muscle mass loss is associated with weakness, fatigue, and insulin resistance. In fact, it is predicted that controlling muscle atrophy can reduce morbidity and mortality associated with diseases such as cancer cachexia and sarcopenia. METHODS: We analyzed gene expression data from muscle of mice or human patients with diverse muscle pathologies and identified LMCD1 as a gene strongly associated with skeletal muscle function. We transiently expressed or silenced LMCD1 in mouse gastrocnemius muscle or in mouse primary muscle cells and determined muscle/cell size, targeted gene expression, kinase activity with kinase arrays, protein immunoblotting, and protein synthesis levels. To evaluate force, calcium handling, and fatigue, we transduced the flexor digitorum brevis muscle with a LMCD1-expressing adenovirus and measured specific force and sarcoplasmic reticulum Ca2+ release in individual fibers. Finally, to explore the relationship between LMCD1 and calcineurin, we ectopically expressed Lmcd1 in the gastrocnemius muscle and treated those mice with cyclosporine A (calcineurin inhibitor). In addition, we used a luciferase reporter construct containing the myoregulin gene promoter to confirm the role of a LMCD1-calcineurin-myoregulin axis in skeletal muscle mass control and calcium handling. RESULTS: Here, we identify LIM and cysteine-rich domains 1 (LMCD1) as a positive regulator of muscle mass, that increases muscle protein synthesis and fiber size. LMCD1 expression in vivo was sufficient to increase specific force with lower requirement for calcium handling and to reduce muscle fatigue. Conversely, silencing LMCD1 expression impairs calcium handling and force, and induces muscle fatigue without overt atrophy. The actions of LMCD1 were dependent on calcineurin, as its inhibition using cyclosporine A reverted the observed hypertrophic phenotype. Finally, we determined that LMCD1 represses the expression of myoregulin, a known negative regulator of muscle performance. Interestingly, we observed that skeletal muscle LMCD1 expression is reduced in patients with skeletal muscle disease. CONCLUSIONS: Our gain- and loss-of-function studies show that LMCD1 controls protein synthesis, muscle fiber size, specific force, Ca2+ handling, and fatigue resistance. This work uncovers a novel role for LMCD1 in the regulation of skeletal muscle mass and function with potential therapeutic implications.


Asunto(s)
Proteínas Co-Represoras/genética , Proteínas Co-Represoras/fisiología , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/fisiología , Músculo Esquelético/fisiología , Animales , Calcineurina/fisiología , Inhibidores de la Calcineurina/farmacología , Calcio/metabolismo , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Hipertrofia/genética , Hipertrofia/patología , Hipertrofia/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Fibras Musculares Esqueléticas/patología , Fibras Musculares Esqueléticas/fisiología , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Proteínas Musculares/fisiología , Fuerza Muscular/genética , Fuerza Muscular/fisiología , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Enfermedades Musculares/fisiopatología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal
18.
Rinsho Shinkeigaku ; 59(11): 740-745, 2019 Nov 08.
Artículo en Japonés | MEDLINE | ID: mdl-31656265

RESUMEN

A 33-year-old man presented with slowly progressive weakness in the lower extremities over 8 years. At the age of 16 years, the elevation of serum creatine kinase level was detected. Physical examination revealed scapular winging, exaggerated lumbar lordosis and tendoachilles contracture. Gowers sign was positive and proximal dominant limb weakness was noted. Hypertrophy was observed in the upper limbs such as the biceps brachii and forearm flexor muscles. Muscle biopsy showed distinct differences in size of muscle fibers and regenerating and necrotic muscle fibers. A histological study revealed decreased calpain3 expression. Gene analysis of CAPN3 revealed two known gene mutations, leading to a diagnosis of calpainopathy (limb girdle muscular dystrophy 2A; LGMD2A). We here report our patient to discuss findings of upper limb hypertrophy, which are frequently missed compared to the lower limb, but important clinical findings.


Asunto(s)
Músculo Esquelético/patología , Distrofia Muscular de Cinturas/diagnóstico , Extremidad Superior , Adulto , Biopsia , Calpaína/deficiencia , Calpaína/genética , Diagnóstico Diferencial , Imagen de Difusión por Resonancia Magnética , Humanos , Hipertrofia , Masculino , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Músculo Esquelético/diagnóstico por imagen , Distrofia Muscular de Cinturas/clasificación , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/patología , Mutación , Tomografía Computarizada por Rayos X
19.
Cell Death Dis ; 10(8): 610, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31406109

RESUMEN

Muscle LIM protein (MLP, CSRP3) is a key regulator of striated muscle function, and its mutations can lead to both hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) in patients. However, due to lack of human models, mechanisms underlining the pathogenesis of MLP defects remain unclear. In this study, we generated a knockout MLP/CSRP3 human embryonic stem cell (hESC) H9 cell line using CRISPR/Cas9 mediated gene disruption. CSRP3 disruption had no impact on the cardiac differentiation of H9 cells and led to confirmed MLP deficiency in hESC-derived cardiomyocytes (ESC-CMs). MLP-deficient hESC-CMs were found to develop phenotypic features of HCM early after differentiation, such as enlarged cell size, multinucleation, and disorganized sarcomeric ultrastructure. Cellular phenotypes of MLP-deficient hESC-CMs subsequently progressed to mimic heart failure (HF) by 30 days post differentiation, including exhibiting mitochondrial damage, increased ROS generation, and impaired Ca2+ handling. Pharmaceutical treatment with beta agonist, such as isoproterenol, was found to accelerate the manifestation of HCM and HF, consistent with transgenic animal models of MLP deficiency. Furthermore, restoration of Ca2+ homeostasis by verapamil prevented the development of HCM and HF phenotypes, suggesting that elevated intracellular Ca2+ concentration is a central mechanism for pathogenesis of MLP deficiency. In summary, MLP-deficient hESC-CMs recapitulate the pathogenesis of HCM and its progression toward HF, providing an important human model for investigation of CSRP3/MLP-associated disease pathogenesis. More importantly, correction of the autonomous dysfunction of Ca2+ handling was found to be an effective method for treating the in vitro development of cardiomyopathy disease phenotype.


Asunto(s)
Señalización del Calcio , Cardiomiopatía Hipertrófica/complicaciones , Insuficiencia Cardíaca/complicaciones , Proteínas con Dominio LIM/deficiencia , Proteínas Musculares/deficiencia , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes/metabolismo , Señalización del Calcio/efectos de los fármacos , Cardiomiopatía Hipertrófica/genética , Diferenciación Celular/efectos de los fármacos , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Insuficiencia Cardíaca/genética , Homocigoto , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Embrionarias Humanas/metabolismo , Humanos , Isoproterenol/farmacología , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Fenotipo , Células Madre Pluripotentes/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Verapamilo/farmacología
20.
Skelet Muscle ; 9(1): 14, 2019 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-31133047

RESUMEN

BACKGROUND: Myopalladin (MYPN) is a component of the sarcomere that tethers nebulin in skeletal muscle and nebulette in cardiac muscle to alpha-actinin at the Z lines. Autosomal dominant MYPN mutations cause hypertrophic, dilated, or restrictive cardiomyopathy. Autosomal recessive MYPN mutations have been reported in only six families showing a mildly progressive nemaline or cap myopathy with cardiomyopathy in some patients. CASE PRESENTATION: A consanguineous family with congenital to adult-onset muscle weakness and hanging big toe was reported. Muscle biopsy showed minimal changes with internal nuclei, type 1 fiber predominance, and ultrastructural defects of Z line. Muscle CT imaging showed marked hypodensity of the sartorius bilaterally and MRI scattered abnormal high-intensity areas in the internal tongue muscle and in the posterior cervical muscles. Cardiac involvement was demonstrated by magnetic resonance imaging and late gadolinium enhancement. Whole exome sequencing analysis identified a homozygous loss of function single nucleotide deletion in the exon 11 of the MYPN gene in two siblings. Full-length MYPN protein was undetectable on immunoblotting, and on immunofluorescence, its localization at the Z line was missed. CONCLUSIONS: This report extends the phenotypic spectrum of recessive MYPN-related myopathies showing: (1) the two patients had hanging big toe and the oldest one developed spine and hand contractures, none of these signs observed in the previously reported patients, (2) specific ultrastructural changes consisting in Z line fragmentation, but (3) no nemaline or caps on muscle pathology.


Asunto(s)
Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Miotonía Congénita/genética , Adulto , Cardiomiopatías/diagnóstico por imagen , Cardiomiopatías/genética , Cardiomiopatías/fisiopatología , Consanguinidad , Exones , Femenino , Genes Recesivos , Homocigoto , Humanos , Mutación con Pérdida de Función , Masculino , Microscopía Electrónica de Transmisión , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestructura , Miotonía Congénita/diagnóstico por imagen , Miotonía Congénita/fisiopatología , Linaje , Eliminación de Secuencia , Dedos del Pie/diagnóstico por imagen
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA