Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Sci Transl Med ; 16(743): eadi0077, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38630848

RESUMEN

Peripartum cardiomyopathy (PPCM) is an idiopathic form of pregnancy-induced heart failure associated with preeclampsia. Circulating factors in late pregnancy are thought to contribute to both diseases, suggesting a common underlying pathophysiological process. However, what drives this process remains unclear. Using serum proteomics, we identified the senescence-associated secretory phenotype (SASP), a marker of cellular senescence associated with biological aging, as the most highly up-regulated pathway in young women with PPCM or preeclampsia. Placentas from women with preeclampsia displayed multiple markers of amplified senescence and tissue aging, as well as overall increased gene expression of 28 circulating proteins that contributed to SASP pathway enrichment in serum samples from patients with preeclampsia or PPCM. The most highly expressed placental SASP factor, activin A, was associated with cardiac dysfunction or heart failure severity in women with preeclampsia or PPCM. In a murine model of PPCM induced by cardiomyocyte-specific deletion of the gene encoding peroxisome proliferator-activated receptor γ coactivator-1α, inhibiting activin A signaling in the early postpartum period with a monoclonal antibody to the activin type II receptor improved heart function. In addition, attenuating placental senescence with the senolytic compound fisetin in late pregnancy improved cardiac function in these animals. These findings link senescence biology to cardiac dysfunction in pregnancy and help to elucidate the pathogenesis underlying cardiovascular diseases of pregnancy.


Asunto(s)
Cardiomiopatías , Cardiopatías , Insuficiencia Cardíaca , Preeclampsia , Humanos , Embarazo , Femenino , Ratones , Animales , Periodo Periparto , Placenta , Factores de Transcripción
2.
Diabetes ; 73(2): 260-279, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-37934943

RESUMEN

Metabolic dysfunction-associated steatotic liver disease (MASLD, formerly known as nonalcoholic fatty liver disease [NAFLD]) and metabolic dysfunction-associated steatohepatitis (MASH, formerly known as nonalcoholic steatohepatitis [NASH]) are leading chronic liver diseases, driving cirrhosis, hepatocellular carcinoma, and mortality. MASLD/MASH is associated with increased senescence proteins, including Activin A, and senolytics have been proposed as a therapeutic approach. To test the role of Activin A, we induced hepatic expression of Activin A in a murine MASLD/MASH model. Surprisingly, overexpression of hepatic Activin A dramatically mitigated MASLD, reducing liver steatosis and inflammation as well as systemic fat accumulation, while improving insulin sensitivity. Further studies identified a dramatic decrease in the lipid-associated macrophages marker glycoprotein NMB (Gpnmb) by Activin A, and Gpnmb knockdown in the same model produced similar benefits and transcriptional changes to Activin A expression. These studies reveal a surprising protective role for Activin A in MASLD and the potential for SASP proteins to have context-specific beneficial effects. Moreover, they implicate both Activin A and Gpnmb as potential therapeutic targets for this condition.


Asunto(s)
Activinas , Enfermedades Metabólicas , Enfermedad del Hígado Graso no Alcohólico , Animales , Ratones , Activinas/genética , Activinas/metabolismo , Proteínas del Ojo , Glicoproteínas de Membrana/genética , Factores de Transcripción
4.
Arterioscler Thromb Vasc Biol ; 43(2): 330-349, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36453275

RESUMEN

BACKGROUND: Atherosclerosis is an inflammatory vascular disease marked by hyperlipidemia and hematopoietic stem cell expansion. Activin A, a member of the Activin/GDF/TGFß/BMP (growth/differentiation factor/transforming growth factor beta/bone morphogenetic protein) family is broadly expressed and increases in human atherosclerosis, but its functional effects in vivo in this context remain unclear. METHODS: We studied LDLR-/- mice on a Western diet for 12 weeks and used adeno-associated viral vectors with a liver-specific TBG (thyroxine-binding globulin) promoter to express Activin A or GFP (control). Atherosclerotic lesions were analyzed by oil red staining. Blood lipid profiling was performed by high-performance liquid chromatography, and immune cells were evaluated by flow cytometry. Liver RNA-sequencing was performed to explore the underlying mechanisms. RESULTS: Activin A expression decreased in both livers and aortae from LDLR-/- mice fed a Western diet compared with standard laboratory diet. Adenoassociated virus-TBG-Activin A increased Activin A hepatic expression ≈10-fold at 12 weeks; P<0.001) and circulating Activin A levels ≈2000 pg/ml versus ≈50 pg/ml; P<0.001, compared with controls). Hepatic Activin A expression decreased plasma total and LDL (low-density lipoprotein) cholesterol ≈60% and ≈40%, respectively), reduced inflammatory cells in aortae and proliferating hematopoietic stem cells in bone marrow, and reduced atherosclerotic lesion and necrotic core area in aortae. Activin A also attenuated liver steatosis and expression of the lipogenesis genes, Srebp1 and Srebp2. RNA sequencing revealed Activin A not only blocked expression of genes involved in hepatic de novo lipogenesis but also fatty acid uptake and liver inflammation. In addition, Activin A expressed in the liver also reduced white fat tissue accumulation, decreased adipocyte size, and improved glucose tolerance. CONCLUSIONS: Our studies reveal hepatic Activin A expression reduces inflammation, hematopoietic stem cell expansion, liver steatosis, circulating cholesterol, and fat accumulation, which likely all contribute to the observed protection against atherosclerosis. The reduced Activin A observed in LDLR-/- mice on a Western diet seems maladaptive and deleterious for atherogenesis.


Asunto(s)
Aterosclerosis , Hígado Graso , Humanos , Animales , Ratones , Hígado/metabolismo , Inflamación/genética , Inflamación/prevención & control , Inflamación/metabolismo , Aterosclerosis/genética , Aterosclerosis/prevención & control , Aterosclerosis/metabolismo , Activinas/genética , Activinas/metabolismo , Hígado Graso/genética , Hígado Graso/prevención & control , Colesterol/metabolismo , Redes y Vías Metabólicas , Receptores de LDL/genética , Receptores de LDL/metabolismo , Ratones Noqueados , Ratones Endogámicos C57BL
5.
Circulation ; 145(16): 1218-1233, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35114812

RESUMEN

BACKGROUND: The heart grows in response to pathological and physiological stimuli. The former often precedes cardiomyocyte loss and heart failure; the latter paradoxically protects the heart and enhances cardiomyogenesis. The mechanisms underlying these differences remain incompletely understood. Although long noncoding RNAs (lncRNAs) are important in cardiac development and disease, less is known about their roles in physiological hypertrophy or cardiomyogenesis. METHODS: RNA sequencing was applied to hearts from mice after 8 weeks of voluntary exercise-induced physiological hypertrophy and cardiomyogenesis or transverse aortic constriction for 2 or 8 weeks to induce pathological hypertrophy or heart failure. The top lncRNA candidate was overexpressed in hearts with adeno-associated virus vectors and inhibited with antisense locked nucleic acid-GapmeRs to examine its function. Downstream effectors were identified through promoter analyses and binding assays. The functional roles of a novel downstream effector, dachsous cadherin-related 2 (DCHS2), were examined through transgenic overexpression in zebrafish and cardiac-specific deletion in Cas9-knockin mice. RESULTS: We identified exercise-regulated cardiac lncRNAs, called lncExACTs. lncExACT1 was evolutionarily conserved and decreased in exercised hearts but increased in human and experimental heart failure. Cardiac lncExACT1 overexpression caused pathological hypertrophy and heart failure; lncExACT1 inhibition induced physiological hypertrophy and cardiomyogenesis, protecting against cardiac fibrosis and dysfunction. lncExACT1 functioned by regulating microRNA-222, calcineurin signaling, and Hippo/Yap1 signaling through DCHS2. Cardiomyocyte DCHS2 overexpression in zebrafish induced pathological hypertrophy and impaired cardiac regeneration, promoting scarring after injury. In contrast, murine DCHS2 deletion induced physiological hypertrophy and promoted cardiomyogenesis. CONCLUSIONS: These studies identify lncExACT1-DCHS2 as a novel pathway regulating cardiac hypertrophy and cardiomyogenesis. lncExACT1-DCHS2 acts as a master switch toggling the heart between physiological and pathological growth to determine functional outcomes, providing a potentially tractable therapeutic target for harnessing the beneficial effects of exercise.


Asunto(s)
Proteínas Relacionadas con las Cadherinas/metabolismo , Insuficiencia Cardíaca , MicroARNs , ARN Largo no Codificante , Animales , Cardiomegalia/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Pez Cebra/genética
6.
R Soc Open Sci ; 3(8): 160169, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27853593

RESUMEN

Animals often retreat to refugia when alarmed and the time they spend hiding reflects an economic decision that trades off reducing predation risk with other beneficial activities. Typically, refugia such as burrows are static, but some refugia are dynamic. For species with defensive mutualisms, hiding might be contingent on their mutualist's behaviour. We disturbed and quantified hiding time in magnificent sea anemones, Heteractis magnifica, and their associated domino damselfish, Dascyllus trimaculatus. We found that sea anemone hiding behaviour was dependent on the number and behaviour of their commensal fish: anemones emerged sooner when they had more associated fish and faster returning fish. Together, these results demonstrate that hiding behaviour can be influenced by the behaviour of a commensal; such dynamic mutualisms may be found in other systems.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA