Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
1.
FASEB J ; 37(5): e22917, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37039813

RESUMEN

Autonomic imbalance and metabolic inflammation are important pathological processes in diabetic cardiomyopathy. Gut microbiota dysbiosis and increased levels of bacterial component lipopolysaccharide (LPS) are associated with diabetic myocardial injury, but the mechanism by which gut microbes affect metabolic inflammation and cardiac injury remains unclear. We determined whether pyridostigmine (PYR), which inhibits cholinesterase to improve vagal activity, could regulate the disordered gut microbiota and attenuate gut barrier dysfunction, metabolic endotoxemia, and inflammation in diabetes. Db/db mice exhibited high blood glucose levels, insulin resistance, low vagal activity, and diabetic myocardial injury. Db/db mice also exhibited gut microbiota perturbations and subsequent disruption of gut barrier function, resulting in an influx of LPS, metabolic endotoxemia, and inflammation. PYR ameliorated the dysregulated glucose and lipid metabolism, modulated the overall structure of the gut microbiota, selectively enhanced the abundance of anti-inflammatory bacteria, and reduced the abundance of proinflammatory and potentially pathogenic bacteria in db/db mice. Importantly, PYR enhanced vagal activity, restored gut microbiota homeostasis, and alleviated gut barrier dysfunction. Therefore, the LPS-induced extracellular signal-regulated kinase (ERK)/early growth response-1 (Egr-1) pathway and consequent metabolic inflammation were inhibited, and eventually, cardiac hypertrophy, fibrosis, oxidative stress, and dysfunction were ameliorated in db/db mice. In vitro cardiomyocyte injury was induced by exposing primary neonatal rat ventricular cardiomyocytes to high glucose (HG) and LPS. In vitro analyses showed that HG + LPS induced ERK1/2 phosphorylation, Egr-1 expression, inflammation, and cell apoptosis, which were inhibited by acetylcholine (ACh). Alpha 7 nicotinic ACh receptor but not muscarinic 2 ACh receptor plays an important role in ACh-mediated anti-inflammatory effects and inhibiting the ERK/Egr-1 pathway in HG + LPS-administered neonatal rat ventricular cardiomyocytes. PYR and ACh ameliorated diabetic myocardial injury by inhibiting the LPS-induced ERK/Egr-1 pathway and metabolic inflammation. The vagus-gut-heart axis has provided new insights into the complex mechanisms of diabetes and offers novel therapeutic targets.


Asunto(s)
Diabetes Mellitus , Cardiomiopatías Diabéticas , Endotoxemia , Microbioma Gastrointestinal , Ratones , Ratas , Animales , Lipopolisacáridos/farmacología , Quinasas MAP Reguladas por Señal Extracelular , Endotoxemia/tratamiento farmacológico , Inflamación/metabolismo , Antiinflamatorios/farmacología , Acetilcolina/farmacología , Colinérgicos , Bacterias , Glucosa/farmacología , Receptores Colinérgicos , Diabetes Mellitus/tratamiento farmacológico
3.
Nat Commun ; 13(1): 3947, 2022 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-35803927

RESUMEN

Succinate dehydrogenase, which is known as mitochondrial complex II, has proven to be a fascinating machinery, attracting renewed and increased interest in its involvement in human diseases. Herein, we find that succinate dehydrogenase assembly factor 4 (SDHAF4) is downregulated in cardiac muscle in response to pathological stresses and in diseased hearts from human patients. Cardiac loss of Sdhaf4 suppresses complex II assembly and results in subunit degradation and complex II deficiency in fetal mice. These defects are exacerbated in young adults with globally impaired metabolic capacity and activation of dynamin-related protein 1, which induces excess mitochondrial fission and mitophagy, thereby causing progressive dilated cardiomyopathy and lethal heart failure in animals. Targeting mitochondria via supplementation with fumarate or inhibiting mitochondrial fission improves mitochondrial dynamics, partially restores cardiac function and prolongs the lifespan of mutant mice. Moreover, the addition of fumarate is found to dramatically improve cardiac function in myocardial infarction mice. These findings reveal a vital role for complex II assembly in the development of dilated cardiomyopathy and provide additional insights into therapeutic interventions for heart diseases.


Asunto(s)
Cardiomiopatía Dilatada , Animales , Cardiomiopatía Dilatada/metabolismo , Fumaratos/metabolismo , Ratones , Dinámicas Mitocondriales/fisiología , Mitofagia , Miocitos Cardíacos/metabolismo , Succinato Deshidrogenasa/genética , Succinato Deshidrogenasa/metabolismo
5.
Free Radic Biol Med ; 178: 369-379, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34906725

RESUMEN

Mitochondria participate in essential cellular functions, including energy production, metabolism, redox homeostasis regulation, intracellular Ca2+ handling, apoptosis, and cell fate determination. Disruption of mitochondrial homeostasis under pathological conditions results in mitochondrial reactive oxygen species (ROS) generation and energy insufficiency, which further disturb mitochondrial and cellular homeostasis in a deleterious loop. Mitochondrial redox status has therefore become a potential target for therapy against cardiovascular diseases. In this review, we highlight recent progress in determining the roles of mitochondrial processes in regulating mitochondrial redox status, including mitochondrial dynamics (fusion-fission pathways), mitochondrial cristae remodeling, mitophagy, biogenesis, and mitochondrion-organelle interactions (endoplasmic reticulum-mitochondrion interactions, nucleus-mitochondrion communication, and lipid droplet-mitochondrion interactions). The strategies that activate vagal system include direct vagal activation (electrical vagal stimulation and administration of vagal neurotransmitter acetylcholine) and pharmacological modulation (choline and cholinesterase inhibitors). The vagal system plays an important role in maintaining mitochondrial homeostasis and suppressing mitochondrial oxidative stress by promoting mitochondrial biogenesis and mitophagy, moderating mitochondrial fusion and fission, strengthening mitochondrial cristae stabilization, regulating mitochondrion-organelle interactions, and inhibiting mitochondrial Ca2+ overload. Therefore, enhancement of vagal activity can maintain mitochondrial homeostasis and represents a promising therapeutic strategy for cardiovascular diseases.


Asunto(s)
Enfermedades Cardiovasculares , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/metabolismo , Homeostasis , Humanos , Mitocondrias/metabolismo , Dinámicas Mitocondriales , Oxidación-Reducción
6.
Free Radic Biol Med ; 177: 337-346, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34715295

RESUMEN

As a key organelle in eukaryotic cells, mitochondria play a central role in maintaining normal cellular functions. Mitochondrial dysfunction is reported to be closely related with aging and various diseases. Epigenetic modifications in nuclear genome provide a substantial layer for the modulation of nuclear-encoded gene expression. However, whether mitochondria could also be subjected to such similar epigenetic alterations and the involved mechanisms remain largely obscure and controversial. Recently, accumulating evidence has suggested that mitochondrial epigenetics, also known as mitoepigenetics may serve as an intriguing regulatory layer in mitochondrial DNA (mtDNA)-encoded gene expression. Given the potential regulatory role of mitoepigenetics, mitochondrial dysfunction derived from mitoepigenetics-induced abnormal gene expression could also be closely associated with aging and disease development. In this review, we summarized the recent advances in mitoepigenetics, with a special focus on mtDNA methylation in aging and metabolic-related diseases as well as the new methods and technologies for the study of mitoepigenetics. Uncovering the regulatory role of mitoepigenetics will help to understand the underlying mechanisms of mitochondrial dysfunction and provide novel strategies for delaying aging and preventing metabolic-related diseases.


Asunto(s)
Metilación de ADN , ADN Mitocondrial , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Epigénesis Genética , Mitocondrias/genética , Mitocondrias/metabolismo
7.
Cell Cycle ; 20(18): 1890-1906, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34424820

RESUMEN

Lipid droplets (LDs), which are neutral lipid storage organelles, are important for lipid metabolism and energy homeostasis. LD lipolysis and interactions with mitochondria are tightly coupled to cellular metabolism and may be potential targets to buffer the effects of excessive toxic lipid species levels. Acetylcholine (ACh), the major neurotransmitter of the vagus nerve, exhibits cardioprotective effects. However, limited research has focused on its effects on LD lipolysis and the LD-mitochondria association in fatty acid (FA) overload models. Here, we reveal that palmitate (PA) induces an increase in expression of the FA transport protein cluster of differentiation 36 (CD36) and LD formation; remarkably reduces the expression of lipases involved in triacylglycerol (TAG) lipolysis, such as adipose triglyceride lipase (ATGL), hormone-sensitive lipase (HSL) and monoacylglycerol lipase (MGL); impairs LD-mitochondria interaction; and decreases perilipin 5 (PLIN5) expression, resulting in LD accumulation and mitochondrial dysfunction, which ultimately lead to cardiomyocyte apoptosis. ACh significantly upregulates PLIN5 expression and improved LD lipolysis and the LD-mitochondria association. Moreover, ACh reduces CD36 expression, LD deposition and mitochondrial dysfunction, ultimately suppressing apoptosis in PA-treated neonatal rat ventricular cardiomyocytes (NRVCs). Knockdown of PLIN5, which plays a role in LD-mitochondria contact site formation, abolishes the protective effects of ACh in PA-treated NRVCs. Thus, ACh protects cardiomyocytes from PA-induced apoptosis, at least partly, by promoting LD lipolysis and activating LD-mitochondria interactions via PLIN5. These findings may aid in developing novel therapeutic approaches that target LD lipolysis and PLIN5-mediated LD-mitochondria interactions to prevent or alleviate lipotoxic cardiomyopathy.


Asunto(s)
Acetilcolina/farmacología , Apoptosis/efectos de los fármacos , Gotas Lipídicas/metabolismo , Lipólisis/efectos de los fármacos , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo , Palmitatos/efectos adversos , Perilipina-5/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Animales Recién Nacidos , Antígenos CD36/metabolismo , Células Cultivadas , Gotas Lipídicas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Triglicéridos/metabolismo
8.
Adv Sci (Weinh) ; 8(11): 2004507, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34141522

RESUMEN

Mitochondrial epigenetics is rising as intriguing notion for its potential involvement in aging and diseases, while the details remain largely unexplored. Here it is shown that among the 13 mitochondrial DNA (mtDNA) encoded genes, NADH-dehydrogenase 6 (ND6) transcript is primarily decreased in obese and type 2 diabetes populations, which negatively correlates with its distinctive hypermethylation. Hepatic mtDNA sequencing in mice unveils that ND6 presents the highest methylation level, which dramatically increases under diabetic condition due to enhanced mitochondrial translocation of DNA methyltransferase 1 (DNMT1) promoted by free fatty acid through adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) activation. Hepatic knockdown of ND6 or overexpression of Dnmt1 similarly impairs mitochondrial function and induces systemic insulin resistance both in vivo and in vitro. Genetic or chemical targeting hepatic DNMT1 shows significant benefits against insulin resistance associated metabolic disorders. These findings highlight the pivotal role of ND6 epigenetic network in regulating mitochondrial function and onset of insulin resistance, shedding light on potential preventive and therapeutic strategies of insulin resistance and related metabolic disorders from a perspective of mitochondrial epigenetics.


Asunto(s)
Metilación de ADN/genética , ADN Mitocondrial/genética , Diabetes Mellitus Tipo 2/genética , Resistencia a la Insulina/genética , NADH Deshidrogenasa/genética , Anciano , Animales , Diabetes Mellitus Tipo 2/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad
9.
Front Pharmacol ; 12: 647481, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34084135

RESUMEN

The disruption of gut microbes is associated with diabetic cardiomyopathy, but the mechanism by which gut microbes affect cardiac damage remains unclear. We explored gut microbes and branched-chain amino acid (BCAA) metabolite catabolism in diabetic cardiomyopathy mice and investigated the cardioprotective effect of pyridostigmine. The experiments were conducted using a model of diabetic cardiomyopathy induced by a high-fat diet + streptozotocin in C57BL/6 mice. The results of high-throughput sequencing showed that diabetic cardiomyopathy mice exhibited decreased gut microbial diversity, altered abundance of the diabetes-related microbes, and increased abundance of the BCAA-producing microbes Clostridiales and Lachnospiraceae. In addition, diabetes downregulated tight junction proteins (ZO-1, occludin, and claudin-1) and increased intestinal permeability to impair the intestinal barrier. These impairments were accompanied by reduction in vagal activity that manifested as increased acetylcholinesterase levels, decreased acetylcholine levels, and heart rate variability, which eventually led to cardiac damage. Pyridostigmine enhanced vagal activity, restored gut microbiota homeostasis, decreased BCAA-producing microbe abundance, and improved the intestinal barrier to reduce circulating BCAA levels. Pyridostigmine also upregulated BCAT2 and PP2Cm and downregulated p-BCKDHA/BCKDHA and BCKDK to improve cardiac BCAA catabolism. Moreover, pyridostigmine alleviated abnormal mitochondrial structure; increased ATP production; decreased reactive oxygen species and mitochondria-related apoptosis; and attenuated cardiac dysfunction, hypertrophy, and fibrosis in diabetic cardiomyopathy mice. In conclusion, the gut microbiota, BCAA catabolism, and vagal activity were impaired in diabetic cardiomyopathy mice but were improved by pyridostigmine. These results provide novel insights for the development of a therapeutic strategy for diabetes-induced cardiac damage that targets gut microbes and BCAA catabolism.

10.
Lab Invest ; 101(7): 878-896, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33649466

RESUMEN

The key pathophysiological process leading to heart failure is cardiac remodeling, a term referring to cardiac hypertrophy, fibrosis, and apoptosis. We explored circadian rhythm disruption and calcium dyshomeostasis in cardiac remodeling and investigated the cardioprotective effect of choline. The experiments were conducted using a model of cardiac remodeling by abdominal aorta coarctation (AAC) in Sprague-Dawley rats. In vitro cardiomyocyte remodeling was induced by exposing neonatal rat cardiomyocytes to angiotensin II. The circadian rhythms of the transcript levels of the seven major components of the mammalian clock (Bmal1, Clock, Rev-erbα, Per1/2, and Cry1/2) were altered in AAC rat hearts during a normal 24 h light/dark cycle. AAC also upregulated the levels of proteins that mediate store-operated Ca2+ entry/receptor-operated Ca2+ entry (stromal interaction molecule 1 [STIM1], Orai1, and transient receptor potential canonical 6 [TRPC6]) in rat hearts. Moreover, choline ameliorated circadian rhythm disruption, reduced the upregulated protein levels of STIM1, Orai1, and TRPC6, and alleviated cardiac dysfunction and remodeling (evidenced by attenuated cardiac hypertrophy, fibrosis, and apoptosis) in AAC rats. In vitro analyses showed that choline ameliorated calcium overload, downregulated STIM1, Orai1, and TRPC6, and inhibited thapsigargin-induced store-operated Ca2+ entry and 1-oleoyl-2-acetyl-sn-glycerol-induced receptor-operated Ca2+ entry in angiotensin II-treated cardiomyocytes. In conclusion, choline attenuated AAC-induced cardiac remodeling and cardiac dysfunction, which was related to amelioration of circadian rhythm disruption and attenuation of calcium-handling protein defects. Modulation of vagal activity by choline targeting the circadian rhythm and calcium homeostasis may have therapeutic potential for cardiac remodeling and heart failure.


Asunto(s)
Calcio/metabolismo , Colina/farmacología , Ritmo Circadiano/efectos de los fármacos , Insuficiencia Cardíaca , Remodelación Ventricular/efectos de los fármacos , Animales , Aorta Abdominal/fisiopatología , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Corazón/efectos de los fármacos , Corazón/fisiopatología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Masculino , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Ratas , Ratas Sprague-Dawley
11.
Arterioscler Thromb Vasc Biol ; 40(11): 2649-2664, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32938216

RESUMEN

OBJECTIVE: Phenotypic switching of vascular smooth muscle cells (VSMCs) plays a critical role in atherosclerosis, vascular restenosis, and hypertension. Choline exerts cardioprotective effects; however, little is known about its effects on VSMC phenotypic switching and vascular remodeling. Here, we investigated whether choline modulates VSMC phenotypic changes and explored the underlying mechanisms. Approach and Results: In cultured VSMCs, choline promoted Nrf2 (nuclear factor erythroid 2-related factor 2) nuclear translocation, inducing the expression of HO-1 (heme oxygenase-1) and NQO-1 (NAD[P]H quinone oxidoreductase-1). Consequently, choline ameliorated Ang II (angiotensin II)-induced increases in NOX (NAD[P]H oxidase) expression and the mitochondrial reactive oxygen species level, thereby attenuating Ang II-induced VSMC phenotypic switching, proliferation, and migration, presumably via M3AChRs (type 3 muscarinic acetylcholine receptors). Downregulation of M3AChR or Nrf2 diminished choline-mediated upregulation of Nrf2, HO-1, and NQO-1 expression, as well as inhibition of VSMC phenotypic transformation, suggesting that M3AChR and Nrf2 activation are responsible for the protective effects of choline. Moreover, activation of the Nrf2 pathway by sulforaphane suppressed Ang II-induced VSMC phenotypic switching and proliferation, indicating that Nrf2 is a key regulator of VSMC phenotypic switching and vascular homeostasis. In a rat model of abdominal aortic constriction in vivo, choline attenuated VSMC phenotypic transformation and vascular remodeling in a manner related to activation of the Nrf2 pathway. CONCLUSIONS: These results reveal that choline impedes VSMC phenotypic switching, proliferation, migration, and vascular remodeling by activating M3AChR and Nrf2-antioxidant signaling and suggest a novel role for Nrf2 in VSMC phenotypic modulation.


Asunto(s)
Plasticidad de la Célula/efectos de los fármacos , Colina/farmacología , Agonistas Muscarínicos/farmacología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Receptor Muscarínico M3/agonistas , Remodelación Vascular/efectos de los fármacos , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Factor 2 Relacionado con NF-E2/genética , Fenotipo , Ratas Sprague-Dawley , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Transducción de Señal
12.
J Hypertens ; 38(9): 1745-1754, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32516287

RESUMEN

OBJECTIVE: Cyclooxygenase (COX)-2, an inducible isoform of the major rate-limiting enzymes that regulate the production of prostaglandins is associated with injury, inflammation and proliferation. We sought to examine whether plasma COX-2 levels and its genetic variants is associated with salt sensitivity, BP changes and/or hypertension in humans. METHODS: Eighty participants (aged 18-65 years) were maintained sequentially either on a usual diet for 3 days, a low-salt diet (3.0 g) for 7 days, and a high-salt diet (18.0 g) for an additional 7 days. In addition, we studied participants of the original Baoji Salt-Sensitive Study, recruited from 124 families from seven Chinese villages in 2004 who received the same salt intake intervention, and evaluated them for the development of hypertension. RESULTS: Plasma COX-2 levels were significantly decreased with reduction of salt intake from the usual to a low-salt diet and decreased further when converting from the low-salt to the high-salt diet. SNPs rs12042763 in the COX-2 gene was significantly associated with SBP responses to both low-salt and high-salt diet. SNPs rs689466 and rs12042763 were significantly associated with longitudinal changes in BPs. In addition, several COX-2 SNPs were significantly associated with incident hypertension over an 8-year follow-up. Gene-based analyses also supported the overall association of COX-2 with longitudinal changes in SBP and hypertension incidence. CONCLUSION: This study shows that dietary salt intake affects plasma COX-2 levels and that COX-2 may play a role in salt sensitivity, BP progression and development of hypertension in the Chinese populations studied.


Asunto(s)
Presión Sanguínea , Ciclooxigenasa 2 , Hipertensión/epidemiología , Cloruro de Sodio Dietético/análisis , Adolescente , Adulto , Anciano , Pueblo Asiatico , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/genética , Ciclooxigenasa 2/sangre , Ciclooxigenasa 2/genética , Humanos , Incidencia , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/genética , Adulto Joven
14.
Nat Commun ; 11(1): 2303, 2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-32385276

RESUMEN

White adipose tissue (WAT) expansion in obesity occurs through enlargement of preexisting adipocytes (hypertrophy) and through formation of new adipocytes (adipogenesis). Adipogenesis results in WAT hyperplasia, smaller adipocytes and a metabolically more favourable form of obesity. How obesogenic WAT hyperplasia is induced remains, however, poorly understood. Here, we show that the mechanosensitive cationic channel Piezo1 mediates diet-induced adipogenesis. Mice lacking Piezo1 in mature adipocytes demonstrated defective differentiation of preadipocyte into mature adipocytes when fed a high fat diet (HFD) resulting in larger adipocytes, increased WAT inflammation and reduced insulin sensitivity. Opening of Piezo1 in mature adipocytes causes the release of the adipogenic fibroblast growth factor 1 (FGF1), which induces adipocyte precursor differentiation through activation of the FGF-receptor-1. These data identify a central feed-back mechanism by which mature adipocytes control adipogenesis during the development of obesity and suggest Piezo1-mediated adipocyte mechano-signalling as a mechanism to modulate obesity and its metabolic consequences.


Asunto(s)
Adipocitos/metabolismo , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Canales Iónicos/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Adipogénesis/fisiología , Tejido Adiposo Blanco/metabolismo , Animales , Calorimetría , Células Cultivadas , Femenino , Factor 1 de Crecimiento de Fibroblastos/genética , Citometría de Flujo , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Insulina/sangre , Interleucina-6/sangre , Canales Iónicos/genética , Masculino , Ratones , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
15.
Nutrients ; 12(1)2020 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-31936037

RESUMEN

Hyperlipidemia is associated with metabolic disorders, but the detailed mechanisms and related interventions remain largely unclear. As a functional food in Asian diets, Herba houttuyniae has been reported to have beneficial effects on health. The present research was to investigate the protective effects of Herba houttuyniae aqueous extract (HAE) on hyperlipidemia-induced liver and heart impairments and its potential mechanisms. Male C57BL/6J mice were administered with 200 or 400 mg/kg/day HAE for 9 days, followed by intraperitoneal injection with 0.5 g/kg poloxamer 407 to induce acute hyperlipidemia. HAE treatment significantly attenuated excessive serum lipids and tissue damage markers, prevented hepatic lipid deposition, improved cardiac remodeling, and ameliorated hepatic and cardiac oxidative stress induced by hyperlipidemia. More importantly, NF-E2 related factor (Nrf2)-mediated antioxidant and peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α)-mediated mitochondrial biogenesis pathways as well as mitochondrial complex activities were downregulated in the hyperlipidemic mouse livers and hearts, which may be attributable to the loss of adenosine monophosphate (AMP)-activated protein kinase (AMPK) activity: all of these changes were reversed by HAE supplementation. Our findings link the AMPK/PGC-1α/Nrf2 cascade to hyperlipidemia-induced liver and heart impairments and demonstrate the protective effect of HAE as an AMPK activator in the prevention of hyperlipidemia-related diseases.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Medicamentos Herbarios Chinos/farmacología , Hiperlipidemias/tratamiento farmacológico , Factor 2 Relacionado con NF-E2/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Extractos Vegetales/farmacología , Proteínas Quinasas Activadas por AMP/genética , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Hiperlipidemias/inducido químicamente , Metabolismo de los Lípidos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Factor 2 Relacionado con NF-E2/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Extractos Vegetales/química
16.
Genes Genet Syst ; 94(4): 181, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31666448

RESUMEN

The article appeared in Genes & Genetic Systems Vol. 94, No. 1 (p. 23-34) was listed under the incorrect category. We apologize for this error. The correction is as follows:   Incorrect: Mini review   Identification of atrial fibrillation-associated microRNAs in left and right atria of rheumatic mitral valve disease patients Yang Yan, Rui Shi, Xiaojiang Yu, Chaofeng Sun, Weijin Zang and Hongyan Tian   Correct: Full paper   Identification of atrial fibrillation-associated microRNAs in left and right atria of rheumatic mitral valve disease patients Yang Yan, Rui Shi, Xiaojiang Yu, Chaofeng Sun, Weijin Zang and Hongyan Tian.

17.
Free Radic Biol Med ; 145: 103-117, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31553938

RESUMEN

Mitochondrial dysfunction is associated with obesity-induced cardiac remodelling. Recent research suggests that the cristae are the true bioenergetic components of cells. Acetylcholine (ACh), the major neurotransmitter of the vagus nerve, exerts cardio-protective effects against ischaemia. This study investigated the role of cristae remodelling in palmitate (PA)-induced neonatal rat cardiomyocyte hypertrophy and explored the beneficial effects of ACh. We found loose, fragmented and even lysed cristae in PA-treated neonatal cardiomyocytes along with declines in mitochondrial network and complex expression and overproduction of mitochondrial reactive oxygen species (ROS); these changes ultimately resulted in increased myocardial size. Overexpression of mitofilin by adenoviral infection partly improved cristae shape, mitochondrial network, and ATP content and attenuated cell hypertrophy. Interestingly, siRNA-mediated AMP-activated protein kinase (AMPK) silencing increased the number of cristae with a balloon-like morphology without disturbing mitofilin expression. Furthermore, AMPK knockdown abolished the effects of mitofilin overexpression on cristae remodelling and inhibited the interaction of mitofilin with sorting and assembly machinery 50 (Sam50) and coiled-coil helix coiled-coil helix domain-containing protein 3 (CHCHD3), two core components of the mitochondrial contact site and cristae organizing system (MICOS) complex. Intriguingly, ACh upregulated mitofilin expression and AMPK phosphorylation via the muscarinic ACh receptor (MAChR). Moreover, ACh enhanced protein-protein interactions between mitofilin and other components of the MICOS complex, thereby preventing PA-induced mitochondrial dysfunction and cardiomyocyte hypertrophy; however, these effects were abolished by AMPK silencing. Taken together, our data suggest that ACh improves cristae remodelling to defend against PA-induced myocardial hypertrophy, presumably by increasing mitofilin expression and activating AMPK to form the MICOS complex through MAChR. These results suggest new and promising therapeutic approaches targeting mitochondria to prevent lipotoxic cardiomyopathy.


Asunto(s)
Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Hipertrofia/tratamiento farmacológico , Mitocondrias/genética , Proteínas Mitocondriales/genética , Proteínas Musculares/genética , Proteínas Quinasas/genética , Quinasas de la Proteína-Quinasa Activada por el AMP , Acetilcolina/metabolismo , Animales , Animales Recién Nacidos/genética , Remodelación Atrial/efectos de los fármacos , Remodelación Atrial/genética , Modelos Animales de Enfermedad , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hipertrofia/inducido químicamente , Hipertrofia/metabolismo , Hipertrofia/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Proteínas Mitocondriales/metabolismo , Proteínas Musculares/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Obesidad/tratamiento farmacológico , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Palmitatos/toxicidad , Fosforilación , Mapas de Interacción de Proteínas/efectos de los fármacos , Transporte de Proteínas , ARN Interferente Pequeño/farmacología , Ratas , Nervio Vago/efectos de los fármacos , Nervio Vago/patología
18.
Am J Physiol Endocrinol Metab ; 317(2): E312-E326, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31211620

RESUMEN

Diabetic patients are more susceptible to myocardial ischemia damage than nondiabetic patients, with worse clinical outcomes and greater mortality. The mechanism may be related to glucose metabolism, mitochondrial homeostasis, and oxidative stress. Pyridostigmine may improve vagal activity to protect cardiac function in cardiovascular diseases. Researchers have not determined whether pyridostigmine regulates glucose metabolism and mitochondrial homeostasis to reduce myocardial vulnerability to injury in diabetic mice. In the present study, autonomic imbalance, myocardial damage, mitochondrial dysfunction, and oxidative stress were exacerbated in isoproterenol-stimulated diabetic mice, revealing the myocardial vulnerability of diabetic mice to injury compared with mice with diabetes or exposed to isoproterenol alone. Compared with normal mice, the expression of glucose transporters (GLUT)1/4 phosphofructokinase (PFK) FB3, and pyruvate kinase isoform (PKM) was decreased in diabetic mice, but increased in isoproterenol-stimulated normal mice. Following exposure to isoproterenol, the expression of (GLUT)1/4 phosphofructokinase (PFK) FB3, and PKM decreased in diabetic mice compared with normal mice. The downregulation of SIRT3/AMPK and IRS-1/Akt in isoproterenol-stimulated diabetic mice was exacerbated compared with that in diabetic mice or isoproterenol-stimulated normal mice. Pyridostigmine improved vagus activity, increased GLUT1/4, PFKFB3, and PKM expression, and ameliorated mitochondrial dysfunction and oxidative stress to reduce myocardial damage in isoproterenol-stimulated diabetic mice. Based on these results, it was found that pyridostigmine may reduce myocardial vulnerability to injury via the SIRT3/AMPK and IRS-1/Akt pathways in diabetic mice with isoproterenol-induced myocardial damage. This study may provide a potential therapeutic target for myocardial damage in diabetic patients.


Asunto(s)
Diabetes Mellitus Experimental , Cardiomiopatías Diabéticas/prevención & control , Glucosa/metabolismo , Mitocondrias Cardíacas/efectos de los fármacos , Isquemia Miocárdica/prevención & control , Bromuro de Piridostigmina/farmacología , Animales , Metabolismo de los Hidratos de Carbono/efectos de los fármacos , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/patología , Isoproterenol/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/fisiología , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Bromuro de Piridostigmina/uso terapéutico
19.
Sheng Li Xue Bao ; 71(2): 216-224, 2019 Apr 25.
Artículo en Chino | MEDLINE | ID: mdl-31008481

RESUMEN

Obesity is an important risk factor for cardiovascular diseases, which can lead to a variety of cardiovascular diseases including myocardial remodeling. Obesity may induce myocardial dysfunction by affecting hemodynamics, inducing autonomic imbalance, adipose tissue dysfunction, and mitochondrial dyshomeostasis. The key necessary biochemical functions for metabolic homeostasis are performed in mitochondria, and mitochondrial homeostasis is considered as one of the key determinants for cell viability. Mitochondrial homeostasis is regulated by dynamic regulation of mitochondrial fission and fusion, as well as mitochondrial cristae remodeling, biogenesis, autophagy, and oxidative stress. The mitochondrial fission-fusion and morphological changes of mitochondrial cristae maintain the integrity of the mitochondrial structure. The mitochondria maintain a "healthy" state by balancing biogenesis and autophagy, while reactive oxygen species can act as signaling molecules to regulate intracellular signaling. The excessive accumulation of lipids and lipid metabolism disorder in obesity leads to mitochondrial dyshomeostasis, which activate the apoptotic cascade and lead to myocardial remodeling. In this review, we provide an overview of the recent research progress on obesity-induced myocardial remodeling and its possible mechanism of mitochondrial dyshomeostasis.


Asunto(s)
Mitocondrias/patología , Dinámicas Mitocondriales , Miocardio/patología , Obesidad/fisiopatología , Humanos , Especies Reactivas de Oxígeno
20.
Free Radic Biol Med ; 135: 251-260, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30878647

RESUMEN

Accumulating evidence has elucidated that hyperlipidemia is closely associated with an increasing prevalence of CVDs (cardiovascular diseases) because of endothelial dysfunction. In the present study, we investigated the effect and mechanism of PU (Punicalagin), a major ellagitannin in pomegranate, on endothelial dysfunction both in vivo and in vitro. In vivo, PU significantly ameliorated hyperlipidemia-induced accumulation of serum triglyceride and cholesterol as well as endothelial and mitochondrial dysfunction of thoracic aorta. Intriguingly, the FoxO1 (forkhead box O1) pathway was activated, which may account for prevention of vascular dysfunction and mitochondrial loss via upregulating mitochondrial biogenesis. In line, through in vitro cell cultures, our study demonstrated that PU not only increased the total FoxO1 protein, but also enhanced its nuclear translocation. In addition, silencing of FoxO1 remarkably abolished the ability of PU to augment the mitochondrial biogenesis, eNOS (endothelial NO synthase) expression, and oxidative stress, implying the irreplaceable role of FoxO1 in regulating endothelial function in the presence of PU. Conversely, suppression of excessive ROS (reactive oxygen species) secured the PA (palmitate)-induced decrease of FoxO1 expression, implying that there was a cross-talk between FoxO1 pathway and ROS. Concomitantly, the inflammatory response in current study was primarily mediated via p38 MAPK/NF-κB signaling pathway besides of FoxO1 pathway. Taken together, our findings suggest that PU ameliorates endothelial dysfunction by activating FoxO1 pathway, a pivotal regulating switch of mitochondrial biogenesis.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Proteína Forkhead Box O1/genética , Taninos Hidrolizables/farmacología , Mitocondrias/efectos de los fármacos , Animales , Línea Celular , Células Endoteliales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Taninos Hidrolizables/química , Ratones , Mitocondrias/genética , FN-kappa B/genética , Estrés Oxidativo/efectos de los fármacos , Granada (Fruta)/química , Proteínas Quinasas p38 Activadas por Mitógenos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA