Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 159.485
Filtrar
1.
Nat Commun ; 15(1): 4632, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38951500

RESUMEN

ANKRD11 (Ankyrin Repeat Domain 11) is a chromatin regulator and a causative gene for KBG syndrome, a rare developmental disorder characterized by multiple organ abnormalities, including cardiac defects. However, the role of ANKRD11 in heart development is unknown. The neural crest plays a leading role in embryonic heart development, and its dysfunction is implicated in congenital heart defects. We demonstrate that conditional knockout of Ankrd11 in the murine embryonic neural crest results in persistent truncus arteriosus, ventricular dilation, and impaired ventricular contractility. We further show these defects occur due to aberrant cardiac neural crest cell organization leading to outflow tract septation failure. Lastly, knockout of Ankrd11 in the neural crest leads to impaired expression of various transcription factors, chromatin remodelers and signaling pathways, including mTOR, BMP and TGF-ß in the cardiac neural crest cells. In this work, we identify Ankrd11 as a regulator of neural crest-mediated heart development and function.


Asunto(s)
Cardiopatías Congénitas , Corazón , Ratones Noqueados , Cresta Neural , Proteínas Represoras , Animales , Cresta Neural/metabolismo , Cresta Neural/embriología , Ratones , Corazón/embriología , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Cardiopatías Congénitas/patología , Regulación del Desarrollo de la Expresión Génica , Cromatina/metabolismo , Transducción de Señal , Miocardio/metabolismo , Femenino
2.
Commun Biol ; 7(1): 795, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38951640

RESUMEN

The peroxisome is a versatile organelle that performs diverse metabolic functions. PEX3, a critical regulator of the peroxisome, participates in various biological processes associated with the peroxisome. Whether PEX3 is involved in peroxisome-related redox homeostasis and myocardial regenerative repair remains elusive. We investigate that cardiomyocyte-specific PEX3 knockout (Pex3-KO) results in an imbalance of redox homeostasis and disrupts the endogenous proliferation/development at different times and spatial locations. Using Pex3-KO mice and myocardium-targeted intervention approaches, the effects of PEX3 on myocardial regenerative repair during both physiological and pathological stages are explored. Mechanistically, lipid metabolomics reveals that PEX3 promotes myocardial regenerative repair by affecting plasmalogen metabolism. Further, we find that PEX3-regulated plasmalogen activates the AKT/GSK3ß signaling pathway via the plasma membrane localization of ITGB3. Our study indicates that PEX3 may represent a novel therapeutic target for myocardial regenerative repair following injury.


Asunto(s)
Membrana Celular , Integrina beta3 , Ratones Noqueados , Regeneración , Animales , Ratones , Integrina beta3/metabolismo , Integrina beta3/genética , Membrana Celular/metabolismo , Miocitos Cardíacos/metabolismo , Masculino , Plasmalógenos/metabolismo , Transducción de Señal , Miocardio/metabolismo , Miocardio/patología , Ratones Endogámicos C57BL , Lesiones Cardíacas/metabolismo , Lesiones Cardíacas/patología , Lesiones Cardíacas/genética , Proliferación Celular , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética
3.
J Nanobiotechnology ; 22(1): 385, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38951822

RESUMEN

BACKGROUND: Numerous studies have confirmed the involvement of extracellular vesicles (EVs) in various physiological processes, including cellular death and tissue damage. Recently, we reported that EVs derived from ischemia-reperfusion heart exacerbate cardiac injury. However, the role of EVs from healthy heart tissue (heart-derived EVs, or cEVs) on myocardial ischemia-reperfusion (MI/R) injury remains unclear. RESULTS: Here, we demonstrated that intramyocardial administration of cEVs significantly enhanced cardiac function and reduced cardiac damage in murine MI/R injury models. cEVs treatment effectively inhibited ferroptosis and maintained mitochondrial homeostasis in cardiomyocytes subjected to ischemia-reperfusion injury. Further results revealed that cEVs can transfer ATP5a1 into cardiomyocytes, thereby suppressing mitochondrial ROS production, alleviating mitochondrial damage, and inhibiting cardiomyocyte ferroptosis. Knockdown of ATP5a1 abolished the protective effects of cEVs. Furthermore, we found that the majority of cEVs are derived from cardiomyocytes, and ATP5a1 in cEVs primarily originates from cardiomyocytes of the healthy murine heart. Moreover, we demonstrated that adipose-derived stem cells (ADSC)-derived EVs with ATP5a1 overexpression showed much better efficacy on the therapy of MI/R injury compared to control ADSC-derived EVs. CONCLUSIONS: These findings emphasized the protective role of cEVs in cardiac injury and highlighted the therapeutic potential of targeting ATP5a1 as an important approach for managing myocardial damage induced by MI/R injury.


Asunto(s)
Vesículas Extracelulares , Ratones Endogámicos C57BL , ATPasas de Translocación de Protón Mitocondriales , Daño por Reperfusión Miocárdica , Miocitos Cardíacos , Animales , Vesículas Extracelulares/metabolismo , Ratones , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Masculino , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Miocardio/metabolismo , Miocardio/patología , Especies Reactivas de Oxígeno/metabolismo , Ferroptosis/efectos de los fármacos , Modelos Animales de Enfermedad
4.
J Nanobiotechnology ; 22(1): 382, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38951872

RESUMEN

Reperfusion therapy is critical for saving heart muscle after myocardial infarction, but the process of restoring blood flow can itself exacerbate injury to the myocardium. This phenomenon is known as myocardial ischemia-reperfusion injury (MIRI), which includes oxidative stress, inflammation, and further cell death. microRNA-146a (miR-146a) is known to play a significant role in regulating the immune response and inflammation, and has been studied for its potential impact on the improvement of heart function after myocardial injury. However, the delivery of miR-146a to the heart in a specific and efficient manner remains a challenge as extracellular RNAs are unstable and rapidly degraded. Milk exosomes (MEs) have been proposed as ideal delivery platform for miRNA-based therapy as they can protect miRNAs from RNase degradation. In this study, the effects of miR-146a containing MEs (MEs-miR-146a) on improvement of cardiac function were examined in a rat model of MIRI. To enhance the targeting delivery of MEs-miR-146a to the site of myocardial injury, the ischemic myocardium-targeted peptide IMTP was modified onto the surfaces, and whether the modified MEs-miR-146a could exert a better therapeutic role was examined by echocardiography, myocardial injury indicators and the levels of inflammatory factors. Furthermore, the expressions of miR-146a mediated NF-κB signaling pathway-related proteins were detected by western blotting and qRT-PCR to further elucidate its mechanisms. MiR-146 mimics were successfully loaded into the MEs by electroporation at a square wave 1000 V voltage and 0.1 ms pulse duration. MEs-miR-146a can be up-taken by cardiomyocytes and protected the cells from oxygen glucose deprivation/reperfusion induced damage in vitro. Oral administration of MEs-miR-146a decreased myocardial tissue apoptosis and the expression of inflammatory factors and improved cardiac function after MIRI. The miR-146a level in myocardium tissues was significantly increased after the administration IMTP modified MEs-miR-146a, which was higher than that of the MEs-miR-146a group. In addition, intravenous injection of IMTP modified MEs-miR-146a enhanced the targeting to heart, improved cardiac function, reduced myocardial tissue apoptosis and suppressed inflammation after MIRI, which was more effective than the MEs-miR-146a treatment. Moreover, IMTP modified MEs-miR-146a reduced the protein levels of IRAK1, TRAF6 and p-p65. Therefore, IMTP modified MEs-miR-146a exerted their anti-inflammatory effect by inhibiting the IRAK1/TRAF6/NF-κB signaling pathway. Taken together, our findings suggested miR-146a containing MEs may be a promising strategy for the treatment of MIRI with better outcome after modification with ischemic myocardium-targeted peptide, which was expected to be applied in clinical practice in future.


Asunto(s)
Exosomas , MicroARNs , Daño por Reperfusión Miocárdica , FN-kappa B , Ratas Sprague-Dawley , Transducción de Señal , Animales , MicroARNs/metabolismo , MicroARNs/genética , Daño por Reperfusión Miocárdica/metabolismo , Exosomas/metabolismo , FN-kappa B/metabolismo , Ratas , Masculino , Leche/química , Miocardio/metabolismo , Cardiotónicos/farmacología , Miocitos Cardíacos/metabolismo
5.
Cardiovasc Diabetol ; 23(1): 227, 2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-38951895

RESUMEN

In recent years, the incidence of diabetes has been increasing rapidly, posing a serious threat to human health. Diabetic cardiomyopathy (DCM) is characterized by cardiomyocyte hypertrophy, myocardial fibrosis, apoptosis, ventricular remodeling, and cardiac dysfunction in individuals with diabetes, ultimately leading to heart failure and mortality. However, the underlying mechanisms contributing to DCM remain incompletely understood. With advancements in molecular biology technology, accumulating evidence has shown that numerous non-coding RNAs (ncRNAs) crucial roles in the development and progression of DCM. This review aims to summarize recent studies on the involvement of three types of ncRNAs (micro RNA, long ncRNA and circular RNA) in the pathophysiology of DCM, with the goal of providing innovative strategies for the prevention and treatment of DCM.


Asunto(s)
Cardiomiopatías Diabéticas , ARN Circular , ARN Largo no Codificante , Humanos , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/fisiopatología , Cardiomiopatías Diabéticas/metabolismo , Animales , ARN Circular/genética , ARN Circular/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Regulación de la Expresión Génica , ARN no Traducido/genética , ARN no Traducido/metabolismo , Transducción de Señal , Miocardio/patología , Miocardio/metabolismo
6.
Clin Cardiol ; 47(7): e24307, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38953367

RESUMEN

BACKGROUND: We aim to provide a comprehensive review of the current state of knowledge of myocardial viability assessment in patients undergoing coronary artery bypass grafting (CABG), with a focus on the clinical markers of viability for each imaging modality. We also compare mortality between patients with viable myocardium and those without viability who undergo CABG. METHODS: A systematic database search with meta-analysis was conducted of comparative original articles (both observations and randomized controlled studies) of patients undergoing CABG with either viable or nonviable myocardium, in EMBASE, MEDLINE, Cochrane database, and Google Scholar, from inception to 2022. Imaging modalities included were dobutamine stress echocardiography (DSE), cardiac magnetic resonance (CMR), single-photon emission computed tomography (SPECT), and positron emission tomography (PET). RESULTS: A total of 17 studies incorporating a total of 2317 patients were included. Across all imaging modalities, the relative risk of death post-CABG was reduced in patients with versus without viability (random-effects model: odds ratio: 0.42; 95% confidence interval: 0.29-0.61; p < 0.001). Imaging for myocardial viability has significant clinical implications as it can affect the accuracy of the diagnosis, guide treatment decisions, and predict patient outcomes. Generally, based on local availability and expertise, either SPECT or DSE should be considered as the first step in evaluating viability, while PET or CMR would provide further evaluation of transmurality, perfusion metabolism, and extent of scar tissue. CONCLUSION: The assessment of myocardial viability is an essential component of preoperative evaluation in patients with ischemic heart disease undergoing surgical revascularization. Careful patient selection and individualized assessment of viability remain paramount.


Asunto(s)
Puente de Arteria Coronaria , Isquemia Miocárdica , Función Ventricular Izquierda , Humanos , Cardiomiopatías/fisiopatología , Cardiomiopatías/cirugía , Cardiomiopatías/diagnóstico , Cardiomiopatías/etiología , Puente de Arteria Coronaria/efectos adversos , Enfermedad de la Arteria Coronaria/cirugía , Enfermedad de la Arteria Coronaria/fisiopatología , Enfermedad de la Arteria Coronaria/diagnóstico , Enfermedad de la Arteria Coronaria/complicaciones , Ecocardiografía de Estrés/métodos , Isquemia Miocárdica/fisiopatología , Isquemia Miocárdica/cirugía , Isquemia Miocárdica/diagnóstico , Isquemia Miocárdica/complicaciones , Miocardio/patología , Supervivencia Tisular , Tomografía Computarizada de Emisión de Fotón Único , Disfunción Ventricular Izquierda/fisiopatología , Disfunción Ventricular Izquierda/etiología , Función Ventricular Izquierda/fisiología
7.
Cell Metab ; 36(7): 1456-1481, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38959861

RESUMEN

The heart is the most metabolically active organ in the human body, and cardiac metabolism has been studied for decades. However, the bulk of studies have focused on animal models. The objective of this review is to summarize specifically what is known about cardiac metabolism in humans. Techniques available to study human cardiac metabolism are first discussed, followed by a review of human cardiac metabolism in health and in heart failure. Mechanistic insights, where available, are reviewed, and the evidence for the contribution of metabolic insufficiency to heart failure, as well as past and current attempts at metabolism-based therapies, is also discussed.


Asunto(s)
Insuficiencia Cardíaca , Miocardio , Humanos , Miocardio/metabolismo , Insuficiencia Cardíaca/metabolismo , Animales , Corazón , Metabolismo Energético
8.
Nat Commun ; 15(1): 5565, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38956062

RESUMEN

Long-term treatment of myocardial infarction is challenging despite medical advances. Tissue engineering shows promise for MI repair, but implantation complexity and uncertain outcomes pose obstacles. microRNAs regulate genes involved in apoptosis, angiogenesis, and myocardial contraction, making them valuable for long-term repair. In this study, we find downregulated miR-199a-5p expression in MI. Intramyocardial injection of miR-199a-5p into the infarcted region of male rats revealed its dual protective effects on the heart. Specifically, miR-199a-5p targets AGTR1, diminishing early oxidative damage post-myocardial infarction, and MARK4, which influences long-term myocardial contractility and enhances cardiac function. To deliver miR-199a-5p efficiently and specifically to ischemic myocardial tissue, we use CSTSMLKAC peptide to construct P-MSN/miR199a-5p nanoparticles. Intravenous administration of these nanoparticles reduces myocardial injury and protects cardiac function. Our findings demonstrate the effectiveness of P-MSN/miR199a-5p nanoparticles in repairing MI through enhanced contraction and anti-apoptosis. miR199a-5p holds significant therapeutic potential for long-term repair of myocardial infarction.


Asunto(s)
MicroARNs , Infarto del Miocardio , Nanopartículas , MicroARNs/genética , MicroARNs/metabolismo , MicroARNs/administración & dosificación , Animales , Infarto del Miocardio/genética , Masculino , Ratas , Nanopartículas/administración & dosificación , Nanopartículas/química , Ratas Sprague-Dawley , Apoptosis/efectos de los fármacos , Miocardio/metabolismo , Miocardio/patología , Modelos Animales de Enfermedad , Contracción Miocárdica/efectos de los fármacos , Administración Intravenosa , Isquemia Miocárdica/genética , Isquemia Miocárdica/terapia , Isquemia Miocárdica/metabolismo
9.
Sci Rep ; 14(1): 15133, 2024 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-38956194

RESUMEN

The goal of this study was to evaluate the intensity of autophagy and ubiquitin-dependent proteolysis processes occurring in myocardium of left ventricle (LV) in subsequent stages of pulmonary arterial hypertension (PAH) to determine mechanisms responsible for LV mass loss in a monocrotaline-induced PAH rat model. LV myocardium samples collected from 32 Wistar rats were analyzed in an early PAH group (n = 8), controls time-paired (n = 8), an end-stage PAH group (n = 8), and their controls (n = 8). Samples were subjected to histological analyses with immunofluorescence staining, autophagy assessment by western blotting, and evaluation of ubiquitin-dependent proteolysis in the LV by immunoprecipitation of ubiquitinated proteins. Echocardiographic, hemodynamic, and heart morphometric parameters were assessed regularly throughout the experiment. Considerable morphological and hemodynamic remodeling of the LV was observed over the course of PAH. The end-stage PAH was associated with significantly impaired LV systolic function and a decrease in LV mass. The LC3B-II expression in the LV was significantly higher in the end-stage PAH group compared to the early PAH group (p = 0.040). The measured LC3B-II/LC3B-I ratios in the end-stage PAH group were significantly elevated compared to the controls (p = 0.039). Immunofluorescence staining showed a significant increase in the abundance of LC3 puncta in the end-stage PAH group compared to the matched controls. There were no statistically significant differences in the levels of expression of all ubiquitinated proteins when comparing both PAH groups and matched controls. Autophagy may be considered as the mechanism behind the LV mass loss at the end stage of PAH.


Asunto(s)
Autofagia , Ventrículos Cardíacos , Proteolisis , Hipertensión Arterial Pulmonar , Ratas Wistar , Ubiquitina , Animales , Ubiquitina/metabolismo , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Ratas , Masculino , Hipertensión Arterial Pulmonar/metabolismo , Hipertensión Arterial Pulmonar/patología , Modelos Animales de Enfermedad , Miocardio/metabolismo , Miocardio/patología , Ecocardiografía , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Remodelación Ventricular
10.
Georgian Med News ; (349): 54-59, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38963202

RESUMEN

Doxorubicin is the common chemotherapeutic agent that has been harnessed for the treatment of various types of malignancy including the treatment of soft tissue and osteosarcoma and cancers of the vital organs like breast, ovary, bladder, and thyroid. It is also used to treat leukaemia and lymphoma, however, this is an obstacle because of their prominent side effects including cardiotoxicity and lung fibrosis, we do aim to determine the role of CoQ10 as an antioxidant on the impeding the deleterious impacts of doxorubicin on tissue degenerative effects. To do so, 27 rats were subdivided into 3 groups of 9 each; CoQ10 exposed group, Doxorubicin exposed group, and CoQ10 plus Doxorubicin group. At the end of the study, the animals were sacrificed and lungs with hearts were harvested, and slides were prepared for examination under a microscope. The results indicated that doxorubicin induced abnormal cellular structure resulting in damaging cellular structures of the lung and heart while CoQ10 impeded these damaging effects and nearly restoring normal tissue structure. As a result, CoQ10 will maintain normal tissue of the lung and heart.


Asunto(s)
Doxorrubicina , Pulmón , Ubiquinona , Animales , Doxorrubicina/efectos adversos , Ubiquinona/análogos & derivados , Ubiquinona/farmacología , Ratas , Pulmón/efectos de los fármacos , Pulmón/patología , Antibióticos Antineoplásicos/efectos adversos , Antibióticos Antineoplásicos/toxicidad , Miocardio/patología , Masculino , Antioxidantes/farmacología , Cardiotoxicidad/etiología , Cardiotoxicidad/patología , Corazón/efectos de los fármacos
11.
Sci Rep ; 14(1): 15344, 2024 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-38961220

RESUMEN

Decreased myocardial capillary density has been reported as an important histopathological feature associated with various heart disorders. Quantitative assessment of cardiac capillarization typically involves double immunostaining of cardiomyocytes (CMs) and capillaries in myocardial slices. In contrast, single immunostaining of basement membrane protein is a straightforward approach to simultaneously label CMs and capillaries, presenting fewer challenges in background staining. However, subsequent image analysis always requires expertise and laborious manual work to identify and segment CMs/capillaries. Here, we developed an image analysis tool, AutoQC, for automatic identification and segmentation of CMs and capillaries in immunofluorescence images of basement membrane. Commonly used capillarization-related measurements can be derived from segmentation results. By leveraging the power of a pre-trained segmentation model (Segment Anything Model, SAM) via prompt engineering, the training of AutoQC required only a small dataset with bounding box annotations instead of pixel-wise annotations. AutoQC outperformed SAM (without prompt engineering) and YOLOv8-Seg, a state-of-the-art instance segmentation model, in both instance segmentation and capillarization assessment. Thus, AutoQC, featuring a weakly supervised algorithm, enables automatic segmentation and high-throughput, high-accuracy capillarization assessment in basement-membrane-immunostained myocardial slices. This approach reduces the training workload and eliminates the need for manual image analysis once AutoQC is trained.


Asunto(s)
Membrana Basal , Procesamiento de Imagen Asistido por Computador , Miocardio , Miocitos Cardíacos , Membrana Basal/metabolismo , Animales , Miocitos Cardíacos/metabolismo , Miocardio/metabolismo , Miocardio/patología , Procesamiento de Imagen Asistido por Computador/métodos , Capilares/metabolismo , Algoritmos , Ratones , Vasos Coronarios/metabolismo , Vasos Coronarios/patología
12.
BMC Cardiovasc Disord ; 24(1): 333, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38961333

RESUMEN

BACKGROUND: Oxidative stress may contribute to cardiac ryanodine receptor (RyR2) dysfunction in diabetic cardiomyopathy. Ginsenoside Rb1 (Rb1) is a major pharmacologically active component of ginseng to treat cardiovascular diseases. Whether Rb1 treat diabetes injured heart remains unknown. This study was to investigate the effect of Rb1 on diabetes injured cardiac muscle tissue and to further investigate its possible molecular pharmacology mechanisms. METHODS: Male Sprague-Dawley rats were injected streptozotocin solution for 2 weeks, followed 6 weeks Rb1 or insulin treatment. The activity of SOD, CAT, Gpx, and the levels of MDA was measured; histological and ultrastructure analyses, RyR2 activity and phosphorylated RyR2(Ser2808) protein expression analyses; and Tunel assay were performed. RESULTS: There was decreased activity of SOD, CAT, Gpx and increased levels of MDA in the diabetic group from control. Rb1 treatment increased activity of SOD, CAT, Gpx and decreased the levels of MDA as compared with diabetic rats. Neutralizing the RyR2 activity significantly decreased in diabetes from control, and increased in Rb1 treatment group from diabetic group. The expression of phosphorylation of RyR2 Ser2808 was increased in diabetic rats from control, and were attenuated with insulin and Rb1 treatment. Diabetes increased the apoptosis rate, and Rb1 treatment decreased the apoptosis rate. Rb1 and insulin ameliorated myocardial injury in diabetic rats. CONCLUSIONS: These data indicate that Rb1 could be useful for mitigating oxidative damage, reduced phosphorylation of RyR2 Ser2808 and decreased the apoptosis rate of cardiomyocytes in diabetic cardiomyopathy.


Asunto(s)
Antioxidantes , Apoptosis , Diabetes Mellitus Experimental , Cardiomiopatías Diabéticas , Ginsenósidos , Miocitos Cardíacos , Estrés Oxidativo , Ratas Sprague-Dawley , Canal Liberador de Calcio Receptor de Rianodina , Estreptozocina , Animales , Diabetes Mellitus Experimental/tratamiento farmacológico , Masculino , Estrés Oxidativo/efectos de los fármacos , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/efectos de los fármacos , Ginsenósidos/farmacología , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/tratamiento farmacológico , Cardiomiopatías Diabéticas/patología , Cardiomiopatías Diabéticas/fisiopatología , Cardiomiopatías Diabéticas/etiología , Apoptosis/efectos de los fármacos , Antioxidantes/farmacología , Fosforilación , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Miocitos Cardíacos/metabolismo , Miocardio/patología , Miocardio/metabolismo , Insulina , Malondialdehído/metabolismo
13.
J Cell Mol Med ; 28(13): e18493, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38963241

RESUMEN

Interleukin-5 (IL-5) has been reported to be involved in cardiovascular diseases, such as atherosclerosis and cardiac injury. This study aimed to investigate the effects of IL-5 on cardiac remodelling. Mice were infused with angiotensin II (Ang II), and the expression and source of cardiac IL-5 were analysed. The results showed that cardiac IL-5 expression was time- and dose-dependently decreased after Ang II infusion, and was mainly derived from cardiac macrophages. Additionally, IL-5-knockout (IL-5-/-) mice were used to observe the effects of IL-5 knockout on Ang II-induced cardiac remodelling. We found knockout of IL-5 significantly increased the expression of cardiac hypertrophy markers, elevated myocardial cell cross-sectional areas and worsened cardiac dysfunction in Ang II-infused mice. IL-5 deletion also promoted M2 macrophage differentiation and exacerbated cardiac fibrosis. Furthermore, the effects of IL-5 deletion on cardiac remodelling was detected after the STAT3 pathway was inhibited by S31-201. The effects of IL-5 on cardiac remodelling and M2 macrophage differentiation were reversed by S31-201. Finally, the effects of IL-5 on macrophage differentiation and macrophage-related cardiac hypertrophy and fibrosis were analysed in vitro. IL-5 knockout significantly increased the Ang II-induced mRNA expression of cardiac hypertrophy markers in myocardial cells that were co-cultured with macrophages, and this effect was reversed by S31-201. Similar trends in the mRNA levels of fibrosis markers were observed when cardiac fibroblasts and macrophages were co-cultured. In conclusions, IL-5 deficiency promote the differentiation of M2 macrophages by activating the STAT3 pathway, thereby exacerbating cardiac remodelling in Ang II-infused mice. IL-5 may be a potential target for the clinical prevention of cardiac remodelling.


Asunto(s)
Angiotensina II , Cardiomegalia , Fibrosis , Interleucina-5 , Macrófagos , Ratones Noqueados , Factor de Transcripción STAT3 , Transducción de Señal , Remodelación Ventricular , Animales , Angiotensina II/farmacología , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Remodelación Ventricular/efectos de los fármacos , Ratones , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Interleucina-5/metabolismo , Interleucina-5/genética , Cardiomegalia/metabolismo , Cardiomegalia/patología , Cardiomegalia/genética , Cardiomegalia/inducido químicamente , Masculino , Ratones Endogámicos C57BL , Diferenciación Celular , Miocardio/metabolismo , Miocardio/patología
14.
Sci Rep ; 14(1): 15416, 2024 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-38965270

RESUMEN

Zinc oxide nanoparticles (ZnO NPs) are one of the most abundantly used nanomaterials in cosmetics and topical products, and nowadays, they are explored in drug delivery and tissue engineering. Some recent data evidenced that they are responsible for cardiotoxic effects and systemic toxicity. The present study aimed to investigate the toxic effect of ZnO NPs (39 nm) on the heart of Wistar rats and to perform a dose-response relationship using three different dose levels (25, 50, 100 mg/kg bw) of ZnO NPs on the electrocardiogram (ECG) readings, the levels of biochemical function parameters of heart, and the oxidative stress and antioxidant biomarkers. Furthermore, zinc concentration level and histopathological examination of heart tissues were determined. ZnO NPs showed a dose-dependent effect, as the 100 mg/kg bw ZnO NPs treated group showed the most significant changes in ECGs parameters: R-R distance, P-R interval, R and T amplitudes, and increased levels of heart enzymes Creatine Kinase- MB (CK-MB) and Lactate dehydrogenase (LDH). On the other hand, elevated zinc concentration levels, oxidative stress biomarkers MDA and NO, and decreased GSH levels were found also in a dose-dependent manner, the results were supported by impairment in the histopathological structure of heart tissues. While the dose of 100 mg/kg bw of ZnO bulk group showed no significant effects on heart function. The present study concluded that ZnO NPs could induce cardiac dysfunctions and pathological lesions mainly in the high dose.


Asunto(s)
Electrocardiografía , Corazón , Estrés Oxidativo , Ratas Wistar , Óxido de Zinc , Animales , Óxido de Zinc/toxicidad , Óxido de Zinc/química , Masculino , Ratas , Estrés Oxidativo/efectos de los fármacos , Corazón/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Nanopartículas del Metal/química , Biomarcadores/metabolismo , Miocardio/metabolismo , Miocardio/patología , Antioxidantes/metabolismo , Antioxidantes/farmacología , Nanopartículas/toxicidad
15.
Theranostics ; 14(9): 3486-3508, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38948064

RESUMEN

Rationale: Device implantation frequently triggers cardiac remodeling and fibrosis, with monocyte-driven inflammatory responses precipitating arrhythmias. This study investigates the role of m6A modification enzymes METTL3 and METTL14 in these responses and explores a novel therapeutic strategy targeting these modifications to mitigate cardiac remodeling and fibrosis. Methods: Peripheral blood mononuclear cells (PBMCs) were collected from patients with ventricular septal defects (VSD) who developed conduction blocks post-occluder implantation. The expression of METTL3 and METTL14 in PBMCs was measured. METTL3 and METTL14 deficiencies were induced to evaluate their effect on angiotensin II (Ang II)-induced myocardial inflammation and fibrosis. m6A modifications were analyzed using methylated RNA immunoprecipitation followed by quantitative PCR. NF-κB pathway activity and levels of monocyte migration and fibrogenesis markers (CXCR2 and TGF-ß1) were assessed. An erythrocyte microvesicle-based nanomedicine delivery system was developed to target activated monocytes, utilizing the METTL3 inhibitor STM2457. Cardiac function was evaluated via echocardiography. Results: Significant upregulation of METTL3 and METTL14 was observed in PBMCs from patients with VSD occluder implantation-associated persistent conduction block. Deficiencies in METTL3 and METTL14 significantly reduced Ang II-induced myocardial inflammation and fibrosis by decreasing m6A modification on MyD88 and TGF-ß1 mRNAs. This disruption reduced NF-κB pathway activation, lowered CXCR2 and TGF-ß1 levels, attenuated monocyte migration and fibrogenesis, and alleviated cardiac remodeling. The erythrocyte microvesicle-based nanomedicine delivery system effectively targeted inflamed cardiac tissue, reducing inflammation and fibrosis and improving cardiac function. Conclusion: Inhibiting METTL3 and METTL14 in monocytes disrupts the NF-κB feedback loop, decreases monocyte migration and fibrogenesis, and improves cardiac function. Targeting m6A modifications of monocytes with STM2457, delivered via erythrocyte microvesicles, reduces inflammation and fibrosis, offering a promising therapeutic strategy for cardiac remodeling associated with device implantation.


Asunto(s)
Fibrosis , Metiltransferasas , Monocitos , FN-kappa B , Humanos , Metiltransferasas/metabolismo , Metiltransferasas/genética , Monocitos/metabolismo , Masculino , Animales , FN-kappa B/metabolismo , Eritrocitos/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo , Femenino , Metilación , Ratones , Factor de Crecimiento Transformador beta1/metabolismo , Micropartículas Derivadas de Células/metabolismo , Leucocitos Mononucleares/metabolismo , Angiotensina II/metabolismo , Receptores de Interleucina-8B/metabolismo , Receptores de Interleucina-8B/genética , Remodelación Ventricular , Miocardio/metabolismo , Miocardio/patología , Nanomedicina/métodos
16.
J Clin Invest ; 134(13)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38949027

RESUMEN

Biological sex is an important modifier of physiology and influences pathobiology in many diseases. While heart disease is the number one cause of death worldwide in both men and women, sex differences exist at the organ and cellular scales, affecting clinical presentation, diagnosis, and treatment. In this Review, we highlight baseline sex differences in cardiac structure, function, and cellular signaling and discuss the contribution of sex hormones and chromosomes to these characteristics. The heart is a remarkably plastic organ and rapidly responds to physiological and pathological cues by modifying form and function. The nature and extent of cardiac remodeling in response to these stimuli are often dependent on biological sex. We discuss organ- and molecular-level sex differences in adaptive physiological remodeling and pathological cardiac remodeling from pressure and volume overload, ischemia, and genetic heart disease. Finally, we offer a perspective on key future directions for research into cardiac sex differences.


Asunto(s)
Caracteres Sexuales , Remodelación Ventricular , Humanos , Femenino , Masculino , Animales , Cardiopatías/patología , Cardiopatías/metabolismo , Cardiopatías/fisiopatología , Cardiopatías/genética , Hormonas Esteroides Gonadales/metabolismo , Corazón/fisiopatología , Corazón/fisiología , Miocardio/patología , Miocardio/metabolismo
17.
Vet Microbiol ; 295: 110154, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38959808

RESUMEN

Porcine reproductive and respiratory syndrome (PRRS) is one of the costliest diseases to pork producers worldwide. We tested samples from the pregnant gilt model (PGM) to better understand the fetal response to in-utero PRRS virus (PRRSV) infection. Our goal was to identify critical tissues and genes associated with fetal resilience or susceptibility. Pregnant gilts (N=22) were infected with PRRSV on day 86 of gestation. At 21 days post maternal infection, the gilts and fetuses were euthanized, and fetal tissues collected. Fetuses were characterized for PRRS viral load in fetal serum and thymus, and preservation status (viable or meconium stained: VIA or MEC). Fetuses (N=10 per group) were compared: uninfected (UNIF; <1 log/µL PRRSV RNA), resilient (HV_VIA, >5 log virus/µL but viable), and susceptible (HV_MEC, >5 log virus/µL with MEC). Gene expression in fetal heart, kidney, and liver was investigated using NanoString transcriptomics. Gene categories investigated were hypothesized to be involved in fetal response to PRRSV infection: renin- angiotensin-aldosterone, inflammatory, transporter and metabolic systems. Following PRRSV infection, CCL5 increased expression in heart and kidney, and ACE2 decreased expression in kidney, each associated with fetal PRRS susceptibility. Liver revealed the most significant differential gene expression: CXCL10 decreased and IL10 increased indicative of immune suppression. Increased liver gene expression indicated potential associations with fetal PRRS susceptibility on several systems including blood pressure regulation (AGTR1), energy metabolism (SLC16A1 and SLC16A7), tissue specific responses (KL) and growth modulation (TGFB1). Overall, analyses of non-lymphoid tissues provided clues to mechanisms of fetal compromise following maternal PRRSV infection.


Asunto(s)
Resistencia a la Enfermedad , Feto , Síndrome Respiratorio y de la Reproducción Porcina , Transcriptoma , Síndrome Respiratorio y de la Reproducción Porcina/inmunología , Virus del Síndrome Respiratorio y Reproductivo Porcino/inmunología , Resistencia a la Enfermedad/genética , Resistencia a la Enfermedad/inmunología , Embarazo , Animales , Porcinos , Femenino , Feto/inmunología , Feto/virología , Regulación de la Expresión Génica/inmunología , Miocardio/inmunología , Hígado/inmunología , Susceptibilidad a Enfermedades/inmunología , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/veterinaria , Riñón/inmunología
18.
Medicine (Baltimore) ; 103(27): e38817, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38968493

RESUMEN

A cross-sectional study to explore the correlation between cardiac and hepatic iron overload and its impact on the quality of life in children diagnosed with severe beta-thalassemia major (ß-TM). A cohort of 55 pediatric patients with ß-TM, diagnosed via genetic testing at the Affiliated Hospital of Guangdong Medical University from January 2015 to January 2022, was included in this study. The assessment of cardiac and hepatic iron overload was conducted using the magnetic resonance imaging T2* technique. The Chinese version of the Pediatric Quality of Life Inventory (PedsQL) 4.0. Pearson correlation analysis was utilized to assess the relationships between the cardiac and hepatic T2* values and between these T2* values and the total scores of PedsQL 4.0. Analysis showed no significant correlation between cardiac and hepatic T2* values. However, a significant relationship was observed between cardiac T2* values and PedsQL 4.0 total scores (r = 0.313, P < .05), indicating that cardiac, but not hepatic, iron overload is associated with the quality of life. This study highlights the absence of correlation between cardiac and hepatic iron overload levels and demonstrates a significant impact of cardiac iron overload on the quality of life in children with ß-TM. These findings suggest the need for a focused approach to cardiac health in managing ß-TM.


Asunto(s)
Sobrecarga de Hierro , Hígado , Imagen por Resonancia Magnética , Calidad de Vida , Talasemia beta , Humanos , Talasemia beta/psicología , Talasemia beta/complicaciones , Estudios Transversales , Sobrecarga de Hierro/diagnóstico por imagen , Masculino , Femenino , Niño , Imagen por Resonancia Magnética/métodos , Hígado/diagnóstico por imagen , Hígado/metabolismo , Preescolar , Adolescente , Miocardio/metabolismo
19.
Mediators Inflamm ; 2024: 8237681, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38974599

RESUMEN

Electroacupuncture (EA) at the Neiguan acupoint (PC6) has shown significant cardioprotective effects. Sympathetic nerves play an important role in maintaining cardiac function after myocardial infarction (MI). Previous studies have found that EA treatment may improve cardiac function by modulating sympathetic remodeling after MI. However, the mechanism in how EA affects sympathetic remodeling and improves cardiac function remains unclear. The aim of this study is to investigate the cardioprotective mechanism of EA after myocardial ischemic injury by improving sympathetic remodeling and promoting macrophage M2 polarization. We established a mouse model of MI by occluding coronary arteries in male C57/BL6 mice. EA treatment was performed at the PC6 with current intensity (1 mA) and frequency (2/15 Hz). Cardiac function was evaluated using echocardiography. Heart rate variability in mice was assessed via standard electrocardiography. Myocardial fibrosis was evaluated by Sirius red staining. Levels of inflammatory factors were assessed using RT-qPCR. Sympathetic nerve remodeling was assessed through ELISA, western blotting, immunohistochemistry, and immunofluorescence staining. Macrophage polarization was evaluated using flow cytometry. Our results indicated that cardiac systolic function improved significantly after EA treatment, with an increase in fractional shortening and ejection fraction. Myocardial fibrosis was significantly mitigated in the EA group. The sympathetic nerve marker tyrosine hydroxylase and the nerve sprouting marker growth-associated Protein 43 were significantly reduced in the EA group, indicating that sympathetic remodeling was significantly reduced. EA treatment also promoted macrophage M2 polarization, reduced levels of inflammatory factors TNF-α, IL-1ß, and IL-6, and decreased macrophage-associated nerve growth factor in myocardial tissue. To sum up, our results suggest that EA at PC6 attenuates sympathetic remodeling after MI to promote macrophage M2 polarization and improve cardiac function.


Asunto(s)
Electroacupuntura , Macrófagos , Ratones Endogámicos C57BL , Infarto del Miocardio , Animales , Masculino , Infarto del Miocardio/terapia , Ratones , Macrófagos/metabolismo , Sistema Nervioso Simpático , Ecocardiografía , Corazón/fisiopatología , Miocardio/metabolismo , Miocardio/patología
20.
Int J Mol Sci ; 25(13)2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-39000409

RESUMEN

Cardiac fibrosis is a severe outcome of Chagas disease (CD), caused by the protozoan Trypanosoma cruzi. Clinical evidence revealed a correlation between fibrosis levels with impaired cardiac performance in CD patients. Therefore, we sought to analyze the effect of inhibitors of TGF-ß (pirfenidone), p38-MAPK (losmapimod) and c-Jun (SP600125) on the modulation of collagen deposition in cardiac fibroblasts (CF) and in vivo models of T. cruzi chronic infection. Sirius Red/Fast Green dye was used to quantify both collagen expression and total protein amount, assessing cytotoxicity. The compounds were also used to treat C57/Bl6 mice chronically infected with T. cruzi, Brazil strain. We identified an anti-fibrotic effect in vitro for pirfenidone (TGF-ß inhibitor, IC50 114.3 µM), losmapimod (p38 inhibitor, IC50 17.6 µM) and SP600125 (c-Jun inhibitor, IC50 3.9 µM). This effect was independent of CF proliferation since these compounds do not affect T. cruzi-induced host cell multiplication as measured by BrdU incorporation. Assays of chronic infection of mice with T. cruzi have shown a reduction in heart collagen by pirfenidone. These results propose a novel approach to fibrosis therapy in CD, with the prospect of repurposing pirfenidone to prevent the onset of ECM accumulation in the hearts of the patients.


Asunto(s)
Cardiomiopatía Chagásica , Fibrosis , Ratones Endogámicos C57BL , Piridonas , Animales , Piridonas/farmacología , Piridonas/uso terapéutico , Cardiomiopatía Chagásica/tratamiento farmacológico , Cardiomiopatía Chagásica/parasitología , Cardiomiopatía Chagásica/metabolismo , Cardiomiopatía Chagásica/patología , Ratones , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/parasitología , Miocardio/patología , Miocardio/metabolismo , Colágeno/metabolismo , Trypanosoma cruzi/efectos de los fármacos , Humanos , Enfermedad Crónica , Factor de Crecimiento Transformador beta/metabolismo , Modelos Animales de Enfermedad , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Masculino , Antracenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...