Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Heart Circ Physiol ; 326(6): H1406-H1419, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38607343

RESUMEN

Cardiovascular research relies heavily on the veracity of in vitro cardiomyocyte models, with H9c2 and HL-1 cell lines at the forefront due to their cardiomyocyte-like properties. However, the variability stemming from nonstandardized culturing and transfection methods poses a significant challenge to data uniformity and reliability. In this study, we introduce meticulously crafted protocols to enhance the culture and transfection of H9c2 and HL-1 cells, emphasizing the reduction of cytotoxic effects while improving transfection efficiency. Through the examination of polymer-based and lipid-based transfection methods, we offer a comparative analysis that underscores the heightened efficiency and reduced toxicity of these approaches. Our research provides an extensive array of step-by-step procedures designed to foster robust cell cultures and outlines troubleshooting practices to rectify issues of low transfection rates. We discuss the merits and drawbacks of both transfection techniques, equipping researchers with the knowledge to choose the most fitting method for their experimental goals. By offering a definitive guide to these cell lines' culturing and transfection, our work seeks to set a new standard in procedural consistency, ensuring that the cardiovascular research community can achieve more dependable and reproducible results, thereby pushing the boundaries of current methodologies toward impactful clinical applications.NEW & NOTEWORTHY We have developed standardized protocols that significantly reduce cytotoxicity and enhance transfection efficiency in H9c2 and HL-1 cardiomyocyte cell lines. Our detailed comparative analysis of polymer-based and lipid-based transfection methods has identified optimized approaches with superior performance. Accompanying these protocols are comprehensive troubleshooting strategies to address common issues related to low transfection rates. Implementing these protocols is expected to yield more consistent and reproducible results, driving the field of cardiovascular research toward impactful clinical breakthroughs.


Asunto(s)
Lípidos , Miocitos Cardíacos , Transfección , Miocitos Cardíacos/metabolismo , Línea Celular , Animales , Lípidos/toxicidad , Lípidos/química , Ratas , Supervivencia Celular , Polímeros/toxicidad , Ratones
2.
Can J Physiol Pharmacol ; 102(3): 196-205, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-37992301

RESUMEN

Although the collagenase enzyme activity of matrix metalloproteinase-9 (MMP9) is well-documented, its non-enzymatic functions remain less understood. The interaction between intracellular superoxide dismutase-1 (SOD1) and MMP9 is known, with SOD1 suppressing MMP9. However, the mechanism by which MMP9, a secretory protein, influences the extracellular antioxidant superoxide dismutase-3 (SOD3) is not yet clear. To explore MMP9's regulatory impact on SOD3, we employed human embryonic kidney-293 cells, transfecting them with MMP9 overexpresssion and catalytic-site mutant plasmids. Additionally, MMP9 overexpressing cells were treated with an MMP9 activator and inhibitor. Analyses of both cell lysates and culture medium provided insights into MMP9's intracellular and extracellular regulatory roles. In-silico analysis and experimental approaches like proximal ligation assay and co-immunoprecipitation were utilized to delineate the protein-protein interactions between MMP9 and SOD3. Our findings indicate that activated MMP9 enhances SOD3 levels, a regulation not hindered by MMP9 inhibitors. Intriguingly, catalytically inactive MMP9 appeared to reduce SOD3 levels, likely due to MMP9's binding with SOD3, leading to their proteolytic degradation. This MMP9 influence on SOD3 was consistent in both intracellular and extracellular environments, suggesting a parallel in MMP9-SOD3 interactions across these domains. Ultimately, this study unveils a novel interaction between MMP9 and SOD3, highlighting the unique regulatory role of catalytically inactive MMP9 in diminishing SOD3 levels, contrasting its usual upregulation by active MMP9.


Asunto(s)
Metaloproteinasa 9 de la Matriz , Superóxido Dismutasa , Humanos , Superóxido Dismutasa-1/genética , Antioxidantes , Bioensayo
3.
Am J Physiol Regul Integr Comp Physiol ; 325(6): R665-R681, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37746707

RESUMEN

Ferroptosis is a newly identified myocardial cell death mechanism driven by iron-dependent lipid peroxidation. The presence of elevated intramyocardial lipid levels and excessive iron in patients with diabetes suggest a predominant role of ferroptosis in diabetic cardiomyopathy. As myocardial cell death is a precursor of heart failure, and intensive glycemic control cannot abate the increased risk of heart failure in patients with diabetes, targeting myocardial cell death via ferroptosis is a promising therapeutic avenue to prevent and/or treat diabetic cardiomyopathy. This review provides updated and comprehensive molecular mechanisms underpinning ferroptosis, clarifies several misconceptions about ferroptosis, emphasizes the importance of ferroptosis in diabetes-induced myocardial cell death, and offers valuable approaches to evaluate and target ferroptosis in the diabetic heart. Furthermore, basic concepts and ideas presented in this review, including glutathione peroxidase-4-independent and mitochondrial mechanisms of ferroptosis, are also important for investigating ferroptosis in other diabetic organs, as well as nondiabetic and metabolically compromised hearts.


Asunto(s)
Diabetes Mellitus , Cardiomiopatías Diabéticas , Ferroptosis , Insuficiencia Cardíaca , Humanos , Hierro/metabolismo , Muerte Celular/fisiología , Peroxidación de Lípido
4.
Adv Exp Med Biol ; 1396: 103-114, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36454462

RESUMEN

Differential gene expression is associated with diabetic cardiomyopathy (DMCM) and culminates in adverse remodeling in the diabetic heart. Genome editing is a technology utilized to alter endogenous genes. Genome editing also provides an option to induce cardioprotective genes or inhibit genes linked to adverse cardiac remodeling and thus has promise in ameliorating DMCM. Non-coding genes have emerged as novel regulators of cellular signaling and may serve as potential therapeutic targets for DMCM. Specifically, there is a widespread change in the gene expression of fetal cardiac genes and microRNAs, termed genetic reprogramming, that promotes pathological remodeling and contributes to heart failure in diabetes. This genetic reprogramming of both coding and non-coding genes varies with the progression and severity of DMCM. Thus, genetic editing provides a promising option to investigate the role of specific genes/non-coding RNAs in DMCM initiation and progression as well as developing therapeutics to mitigate cardiac remodeling and ameliorate DMCM. This chapter will summarize the research progress in genome editing and DMCM and provide future directions for utilizing genome editing as an approach to prevent and/or treat DMCM.


Asunto(s)
Diabetes Mellitus , Cardiomiopatías Diabéticas , Insuficiencia Cardíaca , Humanos , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/terapia , Edición Génica , Remodelación Ventricular , Corazón
5.
Am J Physiol Heart Circ Physiol ; 322(5): H867-H879, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35333113

RESUMEN

Peripheral artery disease (PAD) is an atherosclerotic disease that impairs blood flow and muscle function in the lower limbs. A skeletal muscle myopathy characterized by mitochondrial dysfunction and oxidative damage is present in PAD; however, the underlying mechanisms are not well established. We investigated the impact of chronic ischemia on skeletal muscle microcirculatory function and its association with leg skeletal muscle mitochondrial function and oxygen delivery and utilization capacity in PAD. Gastrocnemius samples and arterioles were harvested from patients with PAD (n = 10) and age-matched controls (Con, n = 11). Endothelium-dependent and independent vasodilation was assessed in response to flow (30 µL·min-1), acetylcholine, and sodium nitroprusside (SNP). Skeletal muscle mitochondrial respiration was quantified by high-resolution respirometry, microvascular oxygen delivery, and utilization capacity (tissue oxygenation index, TOI) were assessed by near-infrared spectroscopy. Vasodilation was attenuated in PAD (P < 0.05) in response to acetylcholine (Con: 71.1 ± 11.1%, PAD: 45.7 ± 18.1%) and flow (Con: 46.6 ± 20.1%, PAD: 29.3 ± 10.5%) but not SNP (P = 0.30). Complex I + II state 3 respiration (P < 0.01) and TOI recovery rate were impaired in PAD (P < 0.05). Both flow and acetylcholine-mediated vasodilation were positively associated with complex I + II state 3 respiration (r = 0.5 and r = 0.5, respectively, P < 0.05). Flow-mediated vasodilation and complex I + II state 3 respiration were positively associated with TOI recovery rate (r = 0.8 and r = 0.7, respectively, P < 0.05). These findings suggest that chronic ischemia attenuates skeletal muscle arteriole endothelial function, which may be a key mediator for mitochondrial and microcirculatory dysfunction in the PAD leg skeletal muscle. Targeting microvascular dysfunction may be an effective strategy to prevent and/or reverse disease progression in PAD.NEW & NOTEWORTHY Ex vivo skeletal muscle arteriole endothelial function is impaired in claudicating patients with PAD, and this is associated with attenuated skeletal muscle mitochondrial respiration. In vivo skeletal muscle oxygen delivery and utilization capacity is compromised in PAD, and this may be due to microcirculatory and mitochondrial dysfunction. These results suggest that targeting skeletal muscle arteriole function may lead to improvements in skeletal muscle mitochondrial respiration and oxygen delivery and utilization capacity in claudicating patients with PAD.


Asunto(s)
Oxígeno , Enfermedad Arterial Periférica , Acetilcolina/metabolismo , Arteriolas , Humanos , Isquemia/metabolismo , Microcirculación , Mitocondrias , Músculo Esquelético/irrigación sanguínea , Oxígeno/metabolismo , Enfermedad Arterial Periférica/diagnóstico , Enfermedad Arterial Periférica/metabolismo , Enfermedad Arterial Periférica/terapia , Respiración
6.
Am J Physiol Heart Circ Physiol ; 323(1): H176-H200, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35657616

RESUMEN

Diabetes is a major risk factor for cardiovascular diseases, including diabetic cardiomyopathy, atherosclerosis, myocardial infarction, and heart failure. As cardiovascular disease represents the number one cause of death in people with diabetes, there has been a major emphasis on understanding the mechanisms by which diabetes promotes cardiovascular disease, and how antidiabetic therapies impact diabetic heart disease. With a wide array of models to study diabetes (both type 1 and type 2), the field has made major progress in answering these questions. However, each model has its own inherent limitations. Therefore, the purpose of this guidelines document is to provide the field with information on which aspects of cardiovascular disease in the human diabetic population are most accurately reproduced by the available models. This review aims to emphasize the advantages and disadvantages of each model, and to highlight the practical challenges and technical considerations involved. We will review the preclinical animal models of diabetes (based on their method of induction), appraise models of diabetes-related atherosclerosis and heart failure, and discuss in vitro models of diabetic heart disease. These guidelines will allow researchers to select the appropriate model of diabetic heart disease, depending on the specific research question being addressed.


Asunto(s)
Aterosclerosis , Diabetes Mellitus Tipo 2 , Cardiomiopatías Diabéticas , Insuficiencia Cardíaca , Infarto del Miocardio , Animales , Diabetes Mellitus Tipo 2/complicaciones , Cardiomiopatías Diabéticas/complicaciones , Insuficiencia Cardíaca/etiología , Humanos , Hipoglucemiantes , Infarto del Miocardio/complicaciones
7.
Stem Cells ; 39(4): 497-506, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33438302

RESUMEN

Epigenetic reprogramming and autophagy have critical roles in differentiation of stem cells. However, very little is known about how epigenetic modifications are mediated and how they contribute to autophagy and differentiation in human cardiac stem cells (hCSCs). Previously, we have reported that intracellular matrix metalloproteinase-9 (MMP9), a collagenase, mediates cell death in hCSCs. Here, we investigated whether intracellular MMP9 mediates epigenetic modifications and autophagy in hCSCs. We created MMP9KO hCSCs and treated them with 5-azacytidine, an inhibitor of DNA methylation, and bafilomycin A1, an inhibitor of autophagosome degradation, and evaluated epigenetic modifications, autophagic flux, and differentiation. Our results showed compromised epigenetic modifications, reduced autophagy, and impaired differentiation in MMP9KO hCSCs. Remarkably, paracrine MMP9 supplementation restored epigenetic modifications but further reduced autophagy in MMP9KO hCSCs. We conclude that intracellular MMP9 is a critical mediator of epigenetic modifications and autophagy in hCSCs. Furthermore, the endocrine and paracrine effects of MMP9 vary for regulating autophagy in hCSCs. These novel roles of MMP9 are valuable for stem cell therapy.


Asunto(s)
Autofagia/genética , Epigénesis Genética , Metaloproteinasa 9 de la Matriz/genética , Miocitos Cardíacos/metabolismo , Células Madre/metabolismo , Autofagosomas/efectos de los fármacos , Autofagosomas/metabolismo , Autofagia/efectos de los fármacos , Azacitidina/farmacología , Sistemas CRISPR-Cas , Diferenciación Celular/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Metilación de ADN/efectos de los fármacos , ADN Metiltransferasa 3A/genética , ADN Metiltransferasa 3A/metabolismo , Eliminación de Gen , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Macrólidos/farmacología , Metaloproteinasa 9 de la Matriz/deficiencia , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Comunicación Paracrina/efectos de los fármacos , Transducción de Señal , Sirtuina 1/genética , Sirtuina 1/metabolismo , Células Madre/citología , Células Madre/efectos de los fármacos
8.
Int J Mol Sci ; 23(3)2022 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-35163316

RESUMEN

Metabolic remodeling is at the heart of diabetic cardiomyopathy. High glycemic fluctuations increase metabolic stress in the type 1 diabetes mellitus (T1DM) heart. There is a lack of understanding on how metabolites and genes affect metabolic remodeling in the T1DM heart. We hypothesize that differential expression of metabolic genes and metabolites synergistically influence metabolic remodeling preceding T1DM cardiomyopathy. To test our hypothesis, we conducted high throughput analysis of hearts from adult male hyperglycemic Ins2+/- (Akita) and littermate normoglycemic Ins2+/+ (WT) mice. The Akita mouse is a spontaneous, genetic model of T1DM that develops increased levels of consistent glycemic variability without the off-target cardiotoxic effects present in chemically- induced models of T1DM. After validating the presence of a T1DM phenotype, we conducted metabolomics via LC-MS analysis and genomics via next-generation sequencing in left ventricle tissue from the Akita heart. Ingenuity Pathway Analyses revealed that 108 and 30 metabolic pathways were disrupted within the metabolomics and genomics datasets, respectively. Notably, a comparison between the two analyses showed 15 commonly disrupted pathways, including ketogenesis, ketolysis, cholesterol biosynthesis, acetyl CoA hydrolysis, and fatty acid biosynthesis and beta-oxidation. These identified metabolic pathways predicted by the differential expression of metabolites and genes provide the foundation for understanding metabolic remodeling in the T1DM heart. By limited experiment, we revealed a predicted disruption in the metabolites and genes behind T1DM cardiac metabolic derangement. Future studies targeting these genes and metabolites will unravel novel therapies to prevent/improve metabolic remodeling in the T1DM heart.


Asunto(s)
Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Corazón/fisiología , Miocardio/metabolismo , Animales , Glucemia/genética , Glucemia/metabolismo , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/metabolismo , Modelos Animales de Enfermedad , Hiperglucemia/genética , Hiperglucemia/metabolismo , Insulina/genética , Insulina/metabolismo , Masculino , Redes y Vías Metabólicas/genética , Metabolómica/métodos , Ratones , Oxidación-Reducción
9.
Curr Diab Rep ; 21(12): 52, 2021 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-34902085

RESUMEN

PURPOSE OF REVIEW: Insulin is at the heart of diabetes mellitus (DM). DM alters cardiac metabolism causing cardiomyopathy, ultimately leading to heart failure. Polyamines, organic compounds synthesized by cardiomyocytes, have an insulin-like activity and effect on glucose metabolism, making them metabolites of interest in the DM heart. This review sheds light on the disrupted microRNA network in the DM heart in relation to developing novel therapeutics targeting polyamine biosynthesis to prevent/mitigate diabetic cardiomyopathy. RECENT FINDINGS: Polyamines prevent DM-induced upregulation of glucose and ketone body levels similar to insulin. Polyamines also enhance mitochondrial respiration and thereby regulate all major metabolic pathways. Non-coding microRNAs regulate a majority of the biological pathways in our body by modulating gene expression via mRNA degradation or translational repression. However, the role of miRNA in polyamine biosynthesis in the DM heart remains unclear. This review discusses the regulation of polyamine synthesis and metabolism, and its impact on cardiac metabolism and circulating levels of glucose, insulin, and ketone bodies. We provide insights on potential roles of polyamines in diabetic cardiomyopathy and putative miRNAs that could regulate polyamine biosynthesis in the DM heart. Future studies will unravel the regulatory roles these miRNAs play in polyamine biosynthesis and will open new doors in the prevention/treatment of adverse cardiac remodeling in diabetic cardiomyopathy.


Asunto(s)
Diabetes Mellitus , Cardiomiopatías Diabéticas , MicroARNs , Cardiomiopatías Diabéticas/genética , Humanos , Insulina , MicroARNs/genética , Miocitos Cardíacos , Poliaminas
10.
Am J Physiol Heart Circ Physiol ; 319(3): H604-H609, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32762561

RESUMEN

Coronavirus disease 2019 (COVID-19) and diabetes outcomes (CORONADO) trial revealed that 10.6% of patients with diabetes mellitus hospitalized for COVID-19 (COVID-19) die within 7 days. Several studies from New York, Italy, and China confirm that patients with diabetes are at a much higher risk for mortality due to COVID-19. Besides respiratory illness, COVID-19 increases cardiac injury and diabetic ketoacidosis. In the absence of specific guidelines for the prevention and treatment of COVID-19 for patients with diabetes, they remain at higher risk and are more susceptible to COVID-19. Furthermore, there is a scarcity of basic knowledge on how diabetes affects pathogenesis of severe acute respiratory coronavirus (SARS-CoV-2) infection. In patients with diabetes, impaired glucose use alters metabolic and consequently biological processes instigating pathological remodeling, which has detrimental effects on cardiovascular systems. A majority of biological processes are regulated by noncoding microRNAs (miRNAs), which have emerged as a promising therapeutic candidate for several diseases. In consideration of the higher risk of mortality in patients with diabetes and COVID-19, novel diagnostic test and treatment strategy are urgently warranted in post-COVID-19 era. Here, we describe potential roles of miRNA as a biomarker and therapeutic candidate, especially for heart failure, in patients with diabetes and COVID-19.


Asunto(s)
Infecciones por Coronavirus/metabolismo , Complicaciones de la Diabetes/epidemiología , MicroARNs/genética , Neumonía Viral/metabolismo , Enzima Convertidora de Angiotensina 2 , Animales , Biomarcadores/metabolismo , COVID-19 , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/patología , Humanos , MicroARNs/metabolismo , Pandemias , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/epidemiología , Neumonía Viral/patología
11.
Am J Physiol Heart Circ Physiol ; 319(2): H456-H467, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32706261

RESUMEN

Peripheral artery disease (PAD) is a manifestation of atherosclerosis in the leg arteries, which causes claudication. This may be in part due to vascular mitochondrial dysfunction and excessive reactive oxygen species (ROS) production. A mitochondrial-targeted antioxidant (MitoQ) has been shown to improve vascular mitochondrial function that, in turn, led to improved vascular function in older adults and animal models. However, the roles of vascular mitochondria in vascular function including endothelial function and arterial stiffness in patients with PAD are unknown; therefore, with the use of acute MitoQ intake, this study examined the roles of vascular mitochondria in endothelial function, arterial stiffness, exercise tolerance, and skeletal muscle function in patients with PAD. Eleven patients with PAD received either MitoQ or placebo in a randomized crossover design. At each visit, blood samples, brachial and popliteal artery flow-mediated dilation (FMD), peripheral and central pulse-wave velocity (PWV), blood pressure (BP), maximal walking capacity, time to claudication (COT), and oxygen utility capacity were measured pre- and-post-MitoQ and placebo. There were significant group by time interactions (P < 0.05) for brachial and popliteal FMD that both increased by Δ2.6 and Δ3.3%, respectively, and increases superoxide dismutase (Δ0.03 U/mL), maximal walking time (Δ73.8 s), maximal walking distance (Δ49.3 m), and COT (Δ44.2 s). There were no changes in resting heart rate, BP, malondialdehyde, total antioxidant capacity, PWV, or oxygen utility capacity (P > 0.05). MitoQ intake may be an effective strategy for targeting the vascular mitochondrial environment, which may be useful for restoring endothelial function, leg pain, and walking time in patients with PAD.NEW & NOTEWORTHY The results of this study reveal for the first time that acute oral intake of mitochondrial-targeted antioxidant (MitoQ, 80 mg) is effective for improving vascular endothelial function and superoxide dismutase in patients with peripheral artery disease (PAD). Acute MitoQ intake is also effective for improving maximal walking capacity and delaying the onset of claudication in patients with PAD. These findings suggest that the acute oral intake of MitoQ-mediated improvements in vascular mitochondria play a pivotal role for improving endothelial function, the redox environment, and skeletal muscle performance in PAD.


Asunto(s)
Antioxidantes/uso terapéutico , Arteria Braquial/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Tolerancia al Ejercicio/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Claudicación Intermitente/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Compuestos Organofosforados/uso terapéutico , Enfermedad Arterial Periférica/tratamiento farmacológico , Arteria Poplítea/efectos de los fármacos , Ubiquinona/análogos & derivados , Anciano , Antioxidantes/metabolismo , Presión Arterial/efectos de los fármacos , Arteria Braquial/metabolismo , Arteria Braquial/fisiopatología , Estudios Cruzados , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Femenino , Humanos , Claudicación Intermitente/diagnóstico , Claudicación Intermitente/metabolismo , Claudicación Intermitente/fisiopatología , Masculino , Persona de Mediana Edad , Mitocondrias/metabolismo , Contracción Muscular/efectos de los fármacos , Nebraska , Compuestos Organofosforados/metabolismo , Enfermedad Arterial Periférica/diagnóstico , Enfermedad Arterial Periférica/metabolismo , Enfermedad Arterial Periférica/fisiopatología , Arteria Poplítea/metabolismo , Arteria Poplítea/fisiopatología , Recuperación de la Función , Factores de Tiempo , Resultado del Tratamiento , Ubiquinona/metabolismo , Ubiquinona/uso terapéutico , Rigidez Vascular/efectos de los fármacos , Caminata
12.
J Immunol ; 200(2): 523-537, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29229678

RESUMEN

Sarcoplasmic/endoplasmic reticulum Ca2+ adenosine triphosphatase (SERCA)2a, a critical regulator of calcium homeostasis, is known to be decreased in heart failure. Patients with myocarditis or dilated cardiomyopathy develop autoantibodies to SERCA2a suggesting that they may have pathogenetic significance. In this report, we describe epitope mapping analysis of SERCA2a in A/J mice that leads us to make five observations: 1) SERCA2a contains multiple T cell epitopes that induce varying degrees of myocarditis. One epitope, SERCA2a 971-990, induces widespread atrial inflammation without affecting noncardiac tissues; the cardiac abnormalities could be noninvasively captured by echocardiography, electrocardiography, and magnetic resonance microscopy imaging. 2) SERCA2a 971-990-induced disease was associated with the induction of CD4 T cell responses and the epitope preferentially binds MHC class II/IAk rather than IEk By creating IAk/and IEk/SERCA2a 971-990 dextramers, the T cell responses were determined by flow cytometry to be Ag specific. 3) SERCA2a 971-990-sensitized T cells produce both Th1 and Th17 cytokines. 4) Animals immunized with SERCA2a 971-990 showed Ag-specific Abs with enhanced production of IgG2a and IgG2b isotypes, suggesting that SERCA2a 971-990 can potentially act as a common epitope for both T cells and B cells. 5) Finally, SERCA2a 971-990-sensitized T cells were able to transfer disease to naive recipients. Together, these data indicate that SERCA2a is a critical autoantigen in the mediation of atrial inflammation in mice and that our model may be helpful to study the inflammatory events that underlie the development of conditions such as atrial fibrillation in humans.


Asunto(s)
Mapeo Epitopo , Epítopos/inmunología , Miocarditis/inmunología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/inmunología , Alelos , Animales , Proteínas Bacterianas , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Mapeo Epitopo/métodos , Epítopos de Linfocito B/inmunología , Técnica del Anticuerpo Fluorescente , Expresión Génica , Atrios Cardíacos/inmunología , Atrios Cardíacos/metabolismo , Atrios Cardíacos/patología , Ventrículos Cardíacos/inmunología , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Epítopos Inmunodominantes/inmunología , Inmunohistoquímica , Ratones , Ratones Endogámicos , Miocarditis/diagnóstico por imagen , Miocarditis/patología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Péptidos/inmunología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
13.
Am J Physiol Heart Circ Physiol ; 316(6): H1237-H1252, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30925069

RESUMEN

The death of cardiomyocytes is a precursor for the cascade of hypertrophic and fibrotic remodeling that leads to cardiomyopathy. In diabetes mellitus (DM), the metabolic environment of hyperglycemia, hyperlipidemia, and oxidative stress causes cardiomyocyte cell death, leading to diabetic cardiomyopathy (DMCM), an independent cause of heart failure. Understanding the roles of the cell death signaling pathways involved in the development of cardiomyopathies is crucial to the discovery of novel targeted therapeutics and biomarkers for DMCM. Recent evidence suggests that hydrogen sulfide (H2S), an endogenous gaseous molecule, has cardioprotective effects against cell death. However, very little is known about signaling by which H2S and its downstream targets regulate myocardial cell death in the DM heart. This review focuses on H2S in the signaling of apoptotic, autophagic, necroptotic, and pyroptotic cell death in DMCM and other cardiomyopathies, abnormalities in H2S synthesis in DM, and potential H2S-based therapeutic strategies to mitigate myocardial cell death to ameliorate DMCM.


Asunto(s)
Apoptosis , Autofagia , Cardiomiopatías Diabéticas/prevención & control , Sulfuro de Hidrógeno/metabolismo , Miocardio/metabolismo , Necroptosis , Remodelación Ventricular , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Fármacos Cardiovasculares/uso terapéutico , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/patología , Cardiomiopatías Diabéticas/fisiopatología , Humanos , Sulfuro de Hidrógeno/uso terapéutico , Miocardio/patología , Necroptosis/efectos de los fármacos , Piroptosis , Transducción de Señal , Remodelación Ventricular/efectos de los fármacos
14.
Am J Physiol Heart Circ Physiol ; 317(5): H891-H922, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31418596

RESUMEN

Cell death is a fundamental process in cardiac pathologies. Recent studies have revealed multiple forms of cell death, and several of them have been demonstrated to underlie adverse cardiac remodeling and heart failure. With the expansion in the area of myocardial cell death and increasing concerns over rigor and reproducibility, it is important and timely to set a guideline for the best practices of evaluating myocardial cell death. There are six major forms of regulated cell death observed in cardiac pathologies, namely apoptosis, necroptosis, mitochondrial-mediated necrosis, pyroptosis, ferroptosis, and autophagic cell death. In this article, we describe the best methods to identify, measure, and evaluate these modes of myocardial cell death. In addition, we discuss the limitations of currently practiced myocardial cell death mechanisms.


Asunto(s)
Investigación Biomédica/normas , Enfermedades Cardiovasculares/patología , Muerte Celular , Guías como Asunto/normas , Miocitos Cardíacos/patología , Animales , Biomarcadores/metabolismo , Enfermedades Cardiovasculares/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Miocitos Cardíacos/metabolismo , Reproducibilidad de los Resultados , Transducción de Señal
15.
Am J Physiol Heart Circ Physiol ; 312(5): H968-H979, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28283551

RESUMEN

An activated renin-angiotensin system (RAS) within the central nervous system has been implicated in sympathoexcitation during various disease conditions including congestive heart failure (CHF). In particular, activation of the RAS in the paraventricular nucleus (PVN) of the hypothalamus has been recognized to augment sympathoexcitation in CHF. We observed a 2.6-fold increase in angiotensinogen (AGT) in the PVN of CHF. To elucidate the molecular mechanism for increased expression of AGT, we performed in silico analysis of the 3'-untranslated region (3'-UTR) of AGT and found a potential binding site for microRNA (miR)-133a. We hypothesized that decreased miR-133a might contribute to increased AGT in the PVN of CHF rats. Overexpression of miR-133a in NG108 cells resulted in 1.4- and 1.5-fold decreases in AGT and angiotensin type II (ANG II) type 1 receptor (AT1R) mRNA levels, respectively. A luciferase reporter assay performed on NG108 cells confirmed miR-133a binding to the 3'-UTR of AGT. Consistent with these in vitro data, we observed a 1.9-fold decrease in miR-133a expression with a concomitant increase in AGT and AT1R expression within the PVN of CHF rats. Furthermore, restoring the levels of miR-133a within the PVN of CHF rats with viral transduction resulted in a significant reduction of AGT (1.4-fold) and AT1R (1.5-fold) levels with a concomitant decrease in basal renal sympathetic nerve activity (RSNA). Restoration of miR-133a also abrogated the enhanced RSNA responses to microinjected ANG II within the PVN of CHF rats. These results reveal a novel and potentially unique role for miR-133a in the regulation of ANG II within the PVN of CHF rats, which may potentially contribute to the commonly observed sympathoexcitation in CHF.NEW & NOTEWORTHY Angiotensinogen (AGT) expression is upregulated in the paraventricular nucleus of the hypothalamus through posttranscriptional mechanism interceded by microRNA-133a in heart failure. Understanding the mechanism of increased expression of AGT in pathological conditions leading to increased sympathoexcitation may provide the basis for the possible development of new therapeutic agents with enhanced specificity.


Asunto(s)
Insuficiencia Cardíaca/genética , MicroARNs/genética , Sistema Renina-Angiotensina/genética , Regiones no Traducidas 3' , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Angiotensinógeno/metabolismo , Animales , Línea Celular Tumoral , Insuficiencia Cardíaca/fisiopatología , Riñón/inervación , Losartán/uso terapéutico , Masculino , MicroARNs/biosíntesis , Núcleo Hipotalámico Paraventricular/fisiopatología , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1/biosíntesis , Receptor de Angiotensina Tipo 1/genética
16.
Am J Physiol Heart Circ Physiol ; 310(7): H802-12, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26801305

RESUMEN

Despite an obnoxious smell and toxicity at a high dose, hydrogen sulfide (H2S) is emerging as a cardioprotective gasotransmitter. H2S mitigates pathological cardiac remodeling by regulating several cellular processes including fibrosis, hypertrophy, apoptosis, and inflammation. These encouraging findings in rodents led to initiation of a clinical trial using a H2S donor in heart failure patients. However, the underlying molecular mechanisms by which H2S mitigates cardiac remodeling are not completely understood. Empirical evidence suggest that H2S may regulate signaling pathways either by directly influencing a gene in the cascade or interacting with nitric oxide (another cardioprotective gasotransmitter) or both. Recent studies revealed that H2S may ameliorate cardiac dysfunction by up- or downregulating specific microRNAs. MicroRNAs are noncoding, conserved, regulatory RNAs that modulate gene expression mostly by translational inhibition and are emerging as a therapeutic target for cardiovascular disease (CVD). Few microRNAs also regulate H2S biosynthesis. The inter-regulation of microRNAs and H2S opens a new avenue for exploring the H2S-microRNA crosstalk in CVD. This review embodies regulatory mechanisms that maintain the physiological level of H2S, exogenous H2S donors used for increasing the tissue levels of H2S, H2S-mediated regulation of CVD, H2S-microRNAs crosstalk in relation to the pathophysiology of heart disease, clinical trials on H2S, and future perspectives for H2S as a therapeutic agent for heart failure.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Sulfuro de Hidrógeno/metabolismo , MicroARNs/metabolismo , Transducción de Señal , Animales , Enfermedades Cardiovasculares/genética , Humanos , MicroARNs/genética
17.
Am J Physiol Regul Integr Comp Physiol ; 306(2): R138-48, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24305061

RESUMEN

The study was conducted to investigate the role for dopamine in the centrally mediated sympathoexcitatory response in rats with Type 2 diabetes (T2D). T2D was induced by a combination of high-fat diet (HFD) and low-dose streptozotocin (STZ). HFD/STZ treatment for 12-14 wk resulted in significant increase in the number of FosB-positive cells in the paraventricular nucleus (PVN) and rostral ventrolateral medulla (RVLM). In anesthetized rats, administration of exogenous dopamine (dopamine hydrochloride, 20 mM) in the PVN, but not in the RVLM, elicited decreases in renal sympathetic nerve activity (RSNA) and mean arterial pressure (MAP) in control rats and but not in the T2D rats. Blocking the endogenous dopamine with dopamine D1/D5 receptor antagonist SCH39166 (2 mM) in the PVN and RVLM, resulted in increases in RSNA, MAP, and heart rate (HR) in both control and T2D rats. These responses were significantly attenuated in T2D rats compared with control rats (PVN - ΔRSNA: 21 ± 10 vs. 44 ± 2%; ΔMAP: 7 ± 3 vs. 19 ± 6 mmHg, ΔHR: 17 ± 5 vs. 32 ± 4 bpm, P < 0.05). There were no significant increases in response to dopamine D2/D3 receptor antagonist raclopride application in the PVN and RVLM of both control and T2D rats. Furthermore, there were decreased dopamine D1 receptor and D2 receptor expressions in the PVN of T2D rats. Taken together, these data suggest that reduced endogenous dopaminergic tone within the PVN may contribute to the sympathoexcitation in T2D.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Dopamina/metabolismo , Núcleo Hipotalámico Paraventricular/fisiología , Sistema Nervioso Simpático/fisiología , Animales , Benzazepinas/farmacología , Glucemia , Antagonistas de Dopamina/farmacología , Regulación de la Expresión Génica , Racloprida/farmacología , Ratas , Receptores Dopaminérgicos/genética , Receptores Dopaminérgicos/metabolismo , Sistema Nervioso Simpático/efectos de los fármacos
18.
Cell Death Discov ; 10(1): 268, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38824159

RESUMEN

Diabetes significantly increases the risk of heart failure by inducing myocardial cell death, potentially through ferroptosis-an iron-dependent, non-apoptotic cell death pathway characterized by lipid peroxidation. The role of cardiac ferroptosis in human heart failure, however, remains poorly understood. In this study, we compared cardiac ferroptosis in humans with diabetic heart failure to that in healthy controls. Our findings reveal that diabetes not only intensifies myocardial cell death but also upregulates markers of ferroptosis in human hearts. This is linked to decreased transcription and activity of glutathione peroxidase-4 (GPX4), influenced by reduced levels of activating transcription factor-4 (ATF4) and nuclear factor erythroid-2-related factor-2 (NRF2), and downregulation of glutathione reductase (GSR). Additionally, diabetic hearts showed an increased labile iron pool due to enhanced heme metabolism by heme oxygenase-1 (HMOX1), elevated iron import via divalent metal transporter-1 (DMT1), reduced iron storage through ferritin light chain (FLC), and decreased iron export via ferroportin-1 (FPN1). The reduction in FPN1 levels likely results from decreased stabilization by amyloid precursor protein (APP) and diminished NRF2-mediated transcription. Furthermore, diabetes upregulates lysophosphatidylcholine acyltransferase-3 (LPCAT3), facilitating the integration of polyunsaturated fatty acids (PUFA) into phospholipid membranes, and downregulates acyl-CoA thioesterase-1 (ACOT1), which further promotes ferroptosis. LC-MS/MS analysis identified several novel proteins implicated in diabetes-induced cardiac ferroptosis, including upregulated ceruloplasmin, which enhances iron metabolism, and cytochrome b-245 heavy chain (CYBB), a key component of NADPH oxidase that aids in the production of reactive oxygen species (ROS), along with downregulated voltage-dependent anion-selective channel protein-2 (VDAC2), essential for maintaining mitochondrial membrane potential. In conclusion, our study not only confirms the presence and potentially predominant role of cardiac ferroptosis in humans with diabetic heart failure but also elucidates its molecular mechanisms, offering potential therapeutic targets to mitigate heart failure complications in diabetic patients.

19.
Cell Death Discov ; 9(1): 111, 2023 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-37012234

RESUMEN

Cytomegalovirus (CMV) is a widely prevalent herpesvirus that reaches seroprevalence rates of up to 95% in several parts of the world. The majority of CMV infections are asymptomatic, albeit they have severe detrimental effects on immunocompromised individuals. Congenital CMV infection is a leading cause of developmental abnormalities in the USA. CMV infection is a significant risk factor for cardiovascular diseases in individuals of all ages. Like other herpesviruses, CMV regulates cell death for its replication and establishes and maintains a latent state in the host. Although CMV-mediated regulation of cell death is reported by several groups, it is unknown how CMV infection affects necroptosis and apoptosis in cardiac cells. Here, we infected primary cardiomyocytes, the contractile cells in the heart, and primary cardiac fibroblasts with wild-type and cell-death suppressor deficient mutant CMVs to determine how CMV regulates necroptosis and apoptosis in cardiac cells. Our results reveal that CMV infection prevents TNF-induced necroptosis in cardiomyocytes; however, the opposite phenotype is observed in cardiac fibroblasts. CMV infection also suppresses inflammation, reactive oxygen species (ROS) generation, and apoptosis in cardiomyocytes. Furthermore, CMV infection improves mitochondrial biogenesis and viability in cardiomyocytes. We conclude that CMV infection differentially affects the viability of cardiac cells.

20.
Am J Physiol Cell Physiol ; 303(1): C41-51, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22517358

RESUMEN

Hydrogen sulfide (H(2)S) has recently been identified as a regulator of various physiological events, including vasodilation, angiogenesis, antiapoptotic, and cellular signaling. Endogenously, H(2)S is produced as a metabolite of homocysteine (Hcy) by cystathionine ß-synthase (CBS), cystathionine γ-lyase (CSE), and 3-mercaptopyruvate sulfurtransferase (3MST). Although Hcy is recognized as vascular risk factor at an elevated level [hyperhomocysteinemia (HHcy)] and contributes to vascular injury leading to renovascular dysfunction, the exact mechanism is unclear. The goal of the current study was to investigate whether conversion of Hcy to H(2)S improves renovascular function. Ex vivo renal artery culture with CBS, CSE, and 3MST triple gene therapy generated more H(2)S in the presence of Hcy, and these arteries were more responsive to endothelial-dependent vasodilation compared with nontransfected arteries treated with high Hcy. Cross section of triple gene-delivered renal arteries immunostaining suggested increased expression of CD31 and VEGF and diminished expression of the antiangiogenic factor endostatin. In vitro endothelial cell culture demonstrated increased mitophagy during high levels of Hcy and was mitigated by triple gene delivery. Also, dephosphorylated Akt and phosphorylated FoxO3 in HHcy were reversed by H(2)S or triple gene delivery. Upregulated matrix metalloproteinases-13 and downregulated tissue inhibitor of metalloproteinase-1 in HHcy were normalized by overexpression of triple genes. Together, these results suggest that H(2)S plays a key role in renovasculopathy during HHcy and is mediated through Akt/FoxO3 pathways. We conclude that conversion of Hcy to H(2)S by CBS, CSE, or 3MST triple gene therapy improves renovascular function in HHcy.


Asunto(s)
Cistationina betasintasa/genética , Cistationina gamma-Liasa/genética , Terapia Genética , Sulfuro de Hidrógeno/metabolismo , Hiperhomocisteinemia/terapia , Sulfurtransferasas/genética , Animales , Células Cultivadas , Cistationina betasintasa/metabolismo , Cistationina gamma-Liasa/metabolismo , Endostatinas/biosíntesis , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Homocisteína/metabolismo , Hiperhomocisteinemia/genética , Hiperhomocisteinemia/metabolismo , Hipertensión Renovascular/genética , Hipertensión Renovascular/terapia , Metaloproteinasa 13 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/biosíntesis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Arteria Renal/metabolismo , Sulfurtransferasas/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Lesiones del Sistema Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA