Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.244
Filtrar
1.
Open Vet J ; 14(7): 1668-1676, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39175975

RESUMEN

Background: Doxorubicin (DOX) is a chemotherapeutic drug applied clinically for the remedy of cancer, but its possibly life-threatening cardiotoxicity effects remain a concern. Aim: After that, this study evaluates the cardioprotective impacts of Lagenaria siceraria (LSS) oil on DOX induced cardiomyopathy in rats. Methods: Wistar male rats (n = 28, weighting 190-210 g) were arbitrarily allocated into four equal groups. Group 1 control group (CTR) received normal saline orally (1 ml/kg); group 2 (DOX) received DOX (10 mg/kg); group 3 (DOLS) received DOX + 3 g of Lagenaria siceraria seeds oil/kg; group 4 (LSSO) received LSSO (3 g/kg) daily for 18 days. The serum samples were collected to determine the creatine kinase-MB (CK-MB) isoenzyme, lactate dehydrogenase (LDH), aspartate aminotransferase (AST), and Troponin I activity. At the same time, the catalase, malondialdehyde (MDA), and reduced glutathione (GSH) were assessed in heart tissues. Additionally, histopathological investigations for the heart tissue were performed. Results: Results revealed no significant change in CK-MB levels between the DOLS group compared to the CTR group (p > 0.05). DOX group confirmed a substantial increase in AST, LDH, and Troponin1 serum levels compared to DOLS and LLSO groups (p < 0.05). The study demonstrated the antioxidant activity of LSS oil against DOX-induced toxicity. The DOX group significantly reduced GSH and catalase levels, with an increase in MDA levels compared to DOLS and LLSO groups. Histopathological analysis showed protective properties of LSS oil against myocardial damage caused by DOX. Conclusion: This study highlights the favorable impacts of LSS oil in mitigating DOX-triggered cardiotoxicity in a rat model.


Asunto(s)
Cardiomiopatías , Doxorrubicina , Ratas Wistar , Animales , Doxorrubicina/efectos adversos , Masculino , Cardiomiopatías/inducido químicamente , Cardiomiopatías/veterinaria , Cardiomiopatías/prevención & control , Ratas , Cucurbitaceae/química , Antibióticos Antineoplásicos , Cardiotoxicidad/prevención & control , Cardiotoxicidad/etiología , Cardiotoxicidad/veterinaria , Extractos Vegetales/farmacología , Extractos Vegetales/administración & dosificación , Aceites de Plantas/farmacología , Aceites de Plantas/administración & dosificación , Aceites de Plantas/uso terapéutico
3.
Cardiovasc Toxicol ; 24(10): 1116-1124, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39068603

RESUMEN

Quercetin (QUE) has been found to inhibit the progression of sepsis-related diseases, including sepsis-induced cardiomyopathy (SIC). More information about the role and mechanism of QUE in SIC progression deserves further exploration. Human cardiomyocytes (AC16) were induced with LPS to mimic SIC cell models. Cell proliferation and apoptosis were determined using CCK8 assay, EdU assay, and flow cytometry. Cell inflammation and ferroptosis were evaluated by detecting IL-1ß, TNF-α, Fe2+, ROS, GSH, and GPX4 levels. 5-lipoxygenase (ALOX5) expression was examined by quantitative real-time PCR and western blot. LPS treatment reduced AC16 cell proliferation, while enhanced apoptosis, inflammation, and ferroptosis. QUE repressed LPS-induced AC16 cell apoptosis, inflammation, and ferroptosis. ALOX5 was upregulated in SIC patients, and its expression was reduced by QUE. ALOX5 knockdown restrained LPS-induced apoptosis, inflammation, and ferroptosis in AC16 cells. The inhibitory effect of QUE on LPS-induced myocardial injury could be reversed by ALOX5 overexpression. QUE promoted the activity of PI3K/AKT pathway by reducing ALOX5 expression. QUE could alleviate LPS-induced myocardial injury by regulating ALOX5/PI3K/AKT pathway, suggesting that QUE might be used for treating SIC.


Asunto(s)
Apoptosis , Cardiomiopatías , Ferroptosis , Lipopolisacáridos , Miocitos Cardíacos , Quercetina , Sepsis , Transducción de Señal , Femenino , Humanos , Masculino , Persona de Mediana Edad , Antiinflamatorios/farmacología , Apoptosis/efectos de los fármacos , Araquidonato 5-Lipooxigenasa/metabolismo , Araquidonato 5-Lipooxigenasa/genética , Cardiomiopatías/inducido químicamente , Cardiomiopatías/metabolismo , Cardiomiopatías/prevención & control , Cardiomiopatías/patología , Cardiomiopatías/enzimología , Línea Celular , Proliferación Celular/efectos de los fármacos , Ferroptosis/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/toxicidad , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quercetina/farmacología , Sepsis/inducido químicamente , Sepsis/tratamiento farmacológico , Sepsis/metabolismo , Transducción de Señal/efectos de los fármacos
4.
Vascul Pharmacol ; 156: 107411, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39029855

RESUMEN

INTRODUCTION: Tafamidis is a molecular chaperone that stabilizes the transthyretin (TTR) homo-tetramer, preventing its dissociation and consequent deposition as amyloid fibrils in organ tissues. Tafamidis reduces mortality and the incidence of hospitalization for cardiovascular causes in patients with TTR amyloid (ATTR) cardiomyopathy. As ATTR cardiomyopathy is associated with a high risk of thromboembolic complications, we hypothesized that tafamidis may have a direct ancillary anti-thrombotic effect. METHODS: Primary human aortic endothelial cells (HAECs) were treated with tafamidis at clinically relevant concentrations and with plasma of patients, before and after the initiation of treatment with tafamidis. The expression of TF was induced by incubation with Tumor Necrosis Factor α (TNFα). Intracellular expression of tissue factor (TF) was measured by western blot. TF activity was measured by a colorimetric assay. Gene expressions of TF were measured by quantitative polymerase chain reaction. RESULTS: Treatment with tafamidis dose-dependently reduced the expression and activity of TNFα-induced TF. This effect was confirmed in cells treated with patients' plasma. Signal Transducer and Activator of Transcription 3 (STAT3) phosphorylation was significantly inhibited by tafamidis. Incubation of HAECs with tafamidis and the STAT3 activator colivelin partially rescued the expression of TF. CONCLUSIONS: Treatment with tafamidis lowers the thrombotic potential in human primary endothelial cells by reducing TF expression and activity. This previously unknown off-target effect may provide a novel mechanistic explanation for the lower number of thromboembolic complications in ATTR cardiomyopathy patients treated with tafamidis.


Asunto(s)
Neuropatías Amiloides Familiares , Benzoxazoles , Cardiomiopatías , Células Endoteliales , Factor de Transcripción STAT3 , Tromboplastina , Factor de Necrosis Tumoral alfa , Humanos , Cardiomiopatías/metabolismo , Cardiomiopatías/tratamiento farmacológico , Cardiomiopatías/prevención & control , Cardiomiopatías/patología , Cardiomiopatías/genética , Benzoxazoles/farmacología , Células Endoteliales/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Células Cultivadas , Neuropatías Amiloides Familiares/tratamiento farmacológico , Neuropatías Amiloides Familiares/metabolismo , Neuropatías Amiloides Familiares/genética , Neuropatías Amiloides Familiares/patología , Tromboplastina/metabolismo , Tromboplastina/genética , Factor de Transcripción STAT3/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Fibrinolíticos/farmacología , Fosforilación , Relación Dosis-Respuesta a Droga , Prealbúmina/metabolismo , Prealbúmina/genética , Masculino , Transducción de Señal/efectos de los fármacos , Femenino , Aorta/metabolismo , Aorta/efectos de los fármacos , Aorta/patología , Anciano , Persona de Mediana Edad
5.
J Pharmacol Sci ; 156(1): 9-18, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39068035

RESUMEN

Necroptosis, necrosis characterized by RIPK3-MLKL activation, has been proposed as a mechanism of doxorubicin (DOX)-induced cardiomyopathy. We showed that rapamycin, an mTORC1 inhibitor, attenuates cardiomyocyte necroptosis. Here we examined role of MLKL in DOX-induced myocardial damage and protective effects of rapamycin. Cardiomyopathy was induced in mice by intraperitoneal injections of DOX (10 mg/kg, every other day) and followed for 7 days. DOX-treated mice showed a significant decline in LVEF assessed by cardiac MRI (45.5 ± 5.1% vs. 65.4 ± 4.2%), reduction in overall survival rates, and increases in myocardial RIPK3 and MLKL expression compared with those in vehicle-treated mice, and those changes were prevented by administration of rapamycin (0.25 mg/kg) before DOX injection. In immunohistochemical analyses, p-MLKL signals were detected in the cardiomyocytes of DOX-treated mice, and the signals were reduced by rapamycin. Mlkl+/- and Mlkl-/- mice were similarly resistant to DOX-induced cardiac dysfunction, indicating that a modest reduction in MLKL level is sufficient to prevent the development of DOX-induced cardiomyopathy. However, evidence of cardiomyocyte necrosis assessed by C9 immunostaining, presence of replacement fibrosis, and electron microscopic analyses was negligible in the myocardium of DOX-treated mice. Thus, MLKL-mediated signaling contributes to DOX-induced cardiac dysfunction primarily by a necrosis-independent mechanism, which is inhibitable by rapamycin.


Asunto(s)
Cardiomiopatías , Doxorrubicina , Ratones Endogámicos C57BL , Miocitos Cardíacos , Necroptosis , Proteínas Quinasas , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Sirolimus , Animales , Doxorrubicina/efectos adversos , Proteínas Quinasas/metabolismo , Sirolimus/farmacología , Cardiomiopatías/inducido químicamente , Cardiomiopatías/prevención & control , Cardiomiopatías/patología , Cardiomiopatías/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Necroptosis/efectos de los fármacos , Masculino , Ratones , Antibióticos Antineoplásicos/efectos adversos , Antibióticos Antineoplásicos/toxicidad
6.
Curr Probl Cardiol ; 49(10): 102743, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39053681

RESUMEN

BACKGROUND: Chemotherapy associated with breast cancer often induces cardiotoxicity, which compromises patients' health and quality of life. OBJECTIVE: To verify the effect of physical exercise on chemotherapy-induced cardiotoxicity, through the assessment of cardiac function in patients with breast cancer. METHODS: A systematic review and meta-analysis of clinical studies was conducted to evaluate the effectiveness of physical training in chemotherapy-induced cardiomyopathy in the PubMed, Web of Sciences and Scopus databases. Thirteen studies were included in the systematic review and eleven studies in the data meta-analysis. RESULTS: Global longitudinal strain presents a cardioprotective effect when compared to the control group (Heterogeneity: Chi² = 12.81, df = 10 (p = 0.23); I² = 22 %.) Test for global effect: Z = 2, 13 (p = 0.03). Physical training is more effective (test for subgroup differences, p = 0.031) in attenuating the impairment of %GLS induced by chemotherapy if performed concomitantly with exposure to chemotherapy (95 % CI; Heterogeneity: Chi² = 7.49, gl = 5 (p = 0.19); I² = 33 %; Test for global effect: Z = 2.33 (p = 0.02) when compared after chemotherapy treatment, or in the long term (for 12 months or more). However, without benefits in LVEF (Heterogeneity: Chi² = 42.14, df = 10 (p < 0.00001); I² = 76 %) Test for global effect: Z = 2.51 (p = 0.01) Conclusion: Exercise training is a cardioprotective approach in breast cancer patients who experience chemotherapy-induced cardiotoxicity. Exercise during exposure to chemotherapy has greater effects on preserving cardiac function.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Cardiotoxicidad , Terapia por Ejercicio , Femenino , Humanos , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Cardiomiopatías/fisiopatología , Cardiomiopatías/prevención & control , Cardiomiopatías/inducido químicamente , Cardiotoxicidad/prevención & control , Cardiotoxicidad/etiología , Terapia por Ejercicio/métodos , Calidad de Vida
7.
Mol Cell Biochem ; 479(7): 1817-1831, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38696001

RESUMEN

Doxorubicin (DOX) is a potent chemotherapeutic drug; however, its clinical use is limited due to its cardiotoxicity. Mitochondrial dysfunction plays a vital role in the pathogenesis of DOX-induced cardiomyopathy. Follistatin-like protein 1 (FSTL1) is a potent cardiokine that protects the heart from diverse cardiac diseases, such as myocardial infarction, cardiac ischemia/reperfusion injury, and heart failure. However, its role in DOX-induced cardiomyopathy is unclear. Therefore, the present study investigated whether administering recombinant FSTL1 could mitigate DOX-induced cardiomyopathy and clarified the underlying molecular mechanisms. FSTL1 treatment attenuated DOX-induced cardiac dysfunction, cardiac fibrosis, and cellular apoptosis by inhibiting excess mitochondrial matrix protein methionine sulfoxide reductase B2 (MsrB2)-mediated mitophagy. Furthermore, FSTL1 administration reduced the expression of apoptotic proteins, including MsrB2, Bax, caspase 3, mitochondrial Parkin, and LC3-II, increased myocardial ATP content, and decreased cardiac malondialdehyde levels, thus protecting mitochondrial function against DOX-induced cardiac injury. Furthermore, FSTL1 treatment protected the contractile properties of adult cardiomyocytes against DOX-induced injury in vitro. Furthermore, carbonyl cyanide m-chlorophenylhydrazone, a mitophagy inducer, impaired the protective effects of FSTL1 in DOX-treated H9c2 cardiomyocytes. In conclusion, these results show that FSTL1 is a novel therapeutic agent against DOX-induced cardiotoxicity that improves mitochondrial function and decreases mitophagy.


Asunto(s)
Cardiomiopatías , Doxorrubicina , Proteínas Relacionadas con la Folistatina , Mitofagia , Miocitos Cardíacos , Mitofagia/efectos de los fármacos , Animales , Doxorrubicina/efectos adversos , Cardiomiopatías/inducido químicamente , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Cardiomiopatías/prevención & control , Ratas , Proteínas Relacionadas con la Folistatina/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Masculino , Línea Celular , Apoptosis/efectos de los fármacos
8.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167208, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38701956

RESUMEN

OBJECTIVE: This study aims to investigate the cardiac protective effects and molecular mechanisms of electroacupuncture (EA) pre-treatment in lipopolysaccharide (LPS)-Induced Cardiomyopathy. METHODS AND RESULTS: Pre-treatment with EA was performed 30 min before intraperitoneal injection of LPS. Cardiac function changes in mice of the EA + LPS group were observed using electrocardiography, echocardiography, and enzyme linked immunosorbent assay (ELISA) and compared with the LPS group. The results demonstrated that EA pre-treatment significantly improved the survival rate of septic mice, alleviated the severity of endotoxemia, and exhibited notable cardiac protective effects. These effects were characterized by a reduction in ST-segment elevation on electrocardiography, an increase in ejection fraction (EF) and fraction shortening (FS) on echocardiography and a decrease in the expression of serum cardiac troponin I (cTn-I) levels. Serum exosomes obtained after EA pre-treatment were extracted and administered to septic mice, revealing significant cardiac protective effects of EA-derived exosomes. Furthermore, the antagonism of circulating exosomes in mice markedly suppressed the cardiac protective effects conferred by EA pre-treatment. Analysis of serum exosomes using quantitative reverse transcription-polymerase chain reaction (qRT-PCR) revealed a significant upregulation of miR-381 expression after EA pre-treatment. Inhibition or overexpression of miR-381 through serotype 9 adeno-associated virus (AAV9)-mediated gene delivery demonstrated that overexpression of miR-381 exerted a cardiac protective effect, while inhibition of miR-381 significantly attenuated the cardiac protective effects conferred by EA pre-treatment. CONCLUSIONS: Our research findings have revealed a novel endogenous cardiac protection mechanism, wherein circulating exosomes derived from EA pre-treatment mitigate LPS-induced cardiac dysfunction via miR-381.


Asunto(s)
Cardiomiopatías , Electroacupuntura , Exosomas , Lipopolisacáridos , MicroARNs , Animales , MicroARNs/genética , MicroARNs/metabolismo , Exosomas/metabolismo , Exosomas/genética , Electroacupuntura/métodos , Ratones , Cardiomiopatías/inducido químicamente , Cardiomiopatías/metabolismo , Cardiomiopatías/terapia , Cardiomiopatías/patología , Cardiomiopatías/genética , Cardiomiopatías/prevención & control , Lipopolisacáridos/toxicidad , Masculino , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad
9.
Phytomedicine ; 130: 155719, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-38763013

RESUMEN

BACKGROUND: Myocardial fibrosis is a risk factor that contributes to the increase in the incidence of cardiovascular disease and death, posing a significant threat to human health. Zhen-wu-tang (ZWT) is a classical Chinese medicinal recipe that has been extensively used to manage cardiovascular disorders throughout history. However, the fundamental processes involved in its effects were not clear. OBJECTIVE: This study examined the therapeutic effects of ZWT on myocardial fibrosis induced by isoproterenol (ISO) in mice, the effect of regulation and underlying mechanism on the polarization of M1 macrophage. METHODS: In vivo, a myocardial fibrosis mouse model was induced via intraperitoneal infusion of isoproterenol (ISO). ZWT or captopril (CAP) was administered intragastrically for 30 days. Cardiac function was evaluated by electrocardiogram (ECG) and echocardiography. By analysing myocardial fibrosis pathomorphologically and identifying fibrosis-related indicators, the protective effect of the ZWT on the heart was evaluated. A model of macrophage polarization was established in vitro by activating RAW264.7 cells with lipopolysaccharide (LPS). The regulatory effects of ZWT on macrophage polarization and the signalling pathways involved were examined by immunofluorescence staining, Western blotting (WB), quantitative real-time PCR (qRT-PCR) and siRNA transfection. RESULTS: ZWT improved cardiac function; reduced fibrotic deposition in cardiac tissues; decreased α-SMA, collagen I, and collagen III levels; and inhibited myocardial fibrosis in mice with ISO-induced myocardial fibrosis. Furthermore, the results showed that ZWT could suppress M1 macrophage polarization by downregulating the expression of CD86 and iNOS in vitro and in vivo. Finally, the results confirmed that ZWT could significantly reduce TLR4/NF-κB signalling pathway activation. CONCLUSION: ZWT showed therapeutic effects on ISO-induced myocardial fibrosis mice, and reduced M1 macrophages polarization through inhibiting TLR4/NF-κB pathway, suggesting that ZWT is a promising drug for myocardial fibrosis treatment.


Asunto(s)
Medicamentos Herbarios Chinos , Fibrosis , Isoproterenol , Macrófagos , Miocardio , FN-kappa B , Transducción de Señal , Receptor Toll-Like 4 , Animales , Ratones , Medicamentos Herbarios Chinos/farmacología , Receptor Toll-Like 4/metabolismo , Macrófagos/efectos de los fármacos , Células RAW 264.7 , Masculino , Transducción de Señal/efectos de los fármacos , FN-kappa B/metabolismo , Miocardio/patología , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Cardiomiopatías/prevención & control , Cardiomiopatías/tratamiento farmacológico
10.
Immun Inflamm Dis ; 12(5): e1229, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38775678

RESUMEN

BACKGROUND: Dioscin has many pharmacological effects; however, its role in sepsis-induced cardiomyopathy (SIC) is unknown. Accordingly, we concentrate on elucidating the mechanism of Dioscin in SIC rat model. METHODS: The SIC rat and H9c2 cell models were established by lipopolysaccharide (LPS) induction. The heart rate (HR), left ventricle ejection fraction (LVEF), mean arterial blood pressure (MAP), and heart weight index (HWI) of rats were evaluated. The myocardial tissue was observed by hematoxylin and eosin staining. 4-Hydroxy-2-nonenal (4-HNE) level in myocardial tissue was detected by immunohistochemistry. Superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH) activities in serum samples of rats and H9c2 cells were determined by colorimetric assay. Bax, B-cell lymphoma-2 (Bcl-2), toll-like receptor 4 (TLR4), myeloid differentiation primary response 88 (MyD88), phosphorylated-p65 (p-p65), and p65 levels in myocardial tissues of rats and treated H9c2 cells were measured by quantitative real-time PCR and Western blot. Viability and reactive oxygen species (ROS) accumulation of treated H9c2 cells were assayed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and dihydroethidium staining assays. RESULTS: Dioscin decreased HR and HWI, increased LVEF and MAP, alleviated the myocardial tissue damage, and reduced 4-HNE level in SIC rats. Dioscin reversed LPS-induced reduction on SOD, CAT, GSH, and Bcl-2 levels, and increment on Bax and TLR4 levels in rats and H9c2 cells. Overexpressed TLR4 attenuated the effects of Dioscin on promoting viability, as well as dwindling TLR4, ROS and MyD88 levels, and p-p65/p65 value in LPS-induced H9c2 cells. CONCLUSION: Protective effects of Dioscin against LPS-induced SIC are achieved via regulation of TLR4/MyD88/p65 signal pathway.


Asunto(s)
Cardiomiopatías , Diosgenina , Factor 88 de Diferenciación Mieloide , Sepsis , Transducción de Señal , Receptor Toll-Like 4 , Animales , Diosgenina/análogos & derivados , Diosgenina/farmacología , Diosgenina/uso terapéutico , Receptor Toll-Like 4/metabolismo , Ratas , Factor 88 de Diferenciación Mieloide/metabolismo , Sepsis/complicaciones , Sepsis/tratamiento farmacológico , Sepsis/metabolismo , Transducción de Señal/efectos de los fármacos , Masculino , Cardiomiopatías/tratamiento farmacológico , Cardiomiopatías/etiología , Cardiomiopatías/metabolismo , Cardiomiopatías/prevención & control , Línea Celular , Ratas Sprague-Dawley , Factor de Transcripción ReIA/metabolismo , Estrés Oxidativo/efectos de los fármacos , Lipopolisacáridos , Modelos Animales de Enfermedad , Apoptosis/efectos de los fármacos
11.
Nutrients ; 16(7)2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38612980

RESUMEN

Recently, we reported that during the hypertrophic phase (230 days old) of hereditary cardiomyopathy of the hamster (HCMH), short-term treatment (20 days) with 250 mg/kg/day of taurine prevents the development of hypertrophy in males but not in females. However, the mortality rate in non-treated animals was higher in females than in males. To verify whether the sex-dependency effect of taurine is due to the difference in the disease's progression, we treated the 230-day-old animals for a longer time period of 122 days. Our results showed that long-term treatment with low and high concentrations of taurine significantly prevents cardiac hypertrophy and early death in HCMH males (p < 0.0001 and p < 0.05, respectively) and females (p < 0.01 and p < 0.0001, respectively). Our results demonstrate that the reported sex dependency of short-term treatments with taurine is due to a higher degree of heart remodeling in females when compared to males and not to sex dependency. In addition, sex-dependency studies should consider the differences between the male and female progression of the disease. Thus, long-term taurine therapies are recommended to prevent remodeling and early death in hereditary cardiomyopathy.


Asunto(s)
Cardiomiopatías , Mortalidad Prematura , Femenino , Masculino , Animales , Cricetinae , Cardiomiopatías/prevención & control , Corazón , Taurina/farmacología , Taurina/uso terapéutico , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/prevención & control
12.
Environ Toxicol ; 39(8): 4134-4147, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38651543

RESUMEN

Doxorubicin (DOX) is a broad-spectrum antibiotic with potent anti-cancer activity. Nevertheless, despite having effective anti-neoplasm activity, its use has been clinically restricted due to its life-threatening side effects, such as cardiotoxicity. It is evident that betaine has anti-oxidant, and anti-inflammatory activity and has several beneficial effects, such as decreasing the amyloid-ß generation, reducing obesity, improving steatosis and fibrosis, and activating AMP-activated protein kinase (AMPK). However, whether betaine could mitigate DOX-induced cardiomyopathy is still unexplored. Cardiomyopathy was induced in male Sprague Dawley rats using DOX (4 mg/kg dose with a cumulative dose of 20 mg/kg, i.p.). Further, betaine (200 and 400 mg/kg) was co-treated with DOX through oral gavage for 28 days. After the completion of the study, several biochemical, oxidative stress parameters, histopathology, western blotting, and qRT-PCR were performed. Betaine treatment significantly reduced CK-MB, LDH, SGOT, and triglyceride levels, which are associated with cardiotoxicity. DOX-induced increased oxidative stress was also mitigated by betaine intervention as the SOD, catalase, MDA, and nitrite levels were restored. The histopathological investigation also confirmed the cardioprotective effect of betaine against DOX-induced cardiomyopathy as the tissue injury was reversed. Further, molecular analysis revealed that betaine suppressed the DOX-induced increased expression of phospho-p53, phospho-p38 MAPK, NF-kB p65, and PINK 1 with an upregulation of AMPK and downregulation of Nrf2 expression. Interestingly, qRT-PCR experiments show that betaine treatment alleviates the DOX-induced increase in inflammatory (TNF-α, NLRP3, and IL-6) and fibrosis (TGF-ß and Acta2) related gene expression, halting the cardiac injury. Interestingly, betaine also improves the mRNA expression of Nrf2, thus modulating the expression of antioxidant proteins and preventing oxidative damage. Here, we provide the first evidence that betaine treatment prevents DOX-induced cardiomyopathy by inhibiting oxidative stress, inflammation, and fibrosis by regulating AMPK/Nrf2/TGF-ß expression. We believe that betaine can be utilized as a potential novel therapeutic strategy for preventing DOX-induced cardiotoxicity.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Betaína , Cardiomiopatías , Doxorrubicina , Fibrosis , Inflamación , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta , Animales , Betaína/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Masculino , Doxorrubicina/toxicidad , Estrés Oxidativo/efectos de los fármacos , Cardiomiopatías/inducido químicamente , Cardiomiopatías/prevención & control , Cardiomiopatías/patología , Cardiomiopatías/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Ratas , Factor de Crecimiento Transformador beta/metabolismo , Antibióticos Antineoplásicos/toxicidad
13.
Cardiovasc Res ; 120(9): 1024-1036, 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-38646672

RESUMEN

AIMS: The anthracycline family of anticancer agents such as doxorubicin (DOX) can induce apoptotic death of cardiomyocytes and cause cardiotoxicity. We previously reported that DOX-induced apoptosis is accompanied by cardiomyocyte cell cycle re-entry. Cell cycle progression requires cyclin-dependent kinase 7 (CDK7)-mediated activation of downstream cell cycle CDKs. This study aims to determine whether CDK7 can be targeted for cardioprotection during anthracycline chemotherapy. METHODS AND RESULTS: DOX exposure induced CDK7 activation in mouse heart and isolated cardiomyocytes. Cardiac-specific ablation of Cdk7 attenuated DOX-induced cardiac dysfunction and fibrosis. Treatment with the covalent CDK7 inhibitor THZ1 also protected against DOX-induced cardiomyopathy and apoptosis. DOX treatment induced activation of the proapoptotic CDK2-FOXO1-Bim axis in a CDK7-dependent manner. In response to DOX, endogenous CDK7 directly bound and phosphorylated CDK2 at Thr160 in cardiomyocytes, leading to full CDK2 kinase activation. Importantly, inhibition of CDK7 further suppressed tumour growth when used in combination with DOX in an immunocompetent mouse model of breast cancer. CONCLUSION: Activation of CDK7 is necessary for DOX-induced cardiomyocyte apoptosis and cardiomyopathy. Our findings uncover a novel proapoptotic role for CDK7 in cardiomyocytes. Moreover, this study suggests that inhibition of CDK7 attenuates DOX-induced cardiotoxicity but augments the anticancer efficacy of DOX. Therefore, combined administration of CDK7 inhibitor and DOX may exhibit diminished cardiotoxicity but superior anticancer activity.


Asunto(s)
Apoptosis , Cardiotoxicidad , Quinasa 2 Dependiente de la Ciclina , Quinasa Activadora de Quinasas Ciclina-Dependientes , Quinasas Ciclina-Dependientes , Doxorrubicina , Ratones Endogámicos C57BL , Miocitos Cardíacos , Inhibidores de Proteínas Quinasas , Animales , Doxorrubicina/toxicidad , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Miocitos Cardíacos/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Femenino , Fenilendiaminas/farmacología , Transducción de Señal/efectos de los fármacos , Fosforilación , Ratones Noqueados , Cardiomiopatías/inducido químicamente , Cardiomiopatías/enzimología , Cardiomiopatías/prevención & control , Cardiomiopatías/patología , Cardiomiopatías/metabolismo , Antibióticos Antineoplásicos/toxicidad , Pirimidinas/farmacología , Humanos , Fibrosis , Línea Celular Tumoral , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/metabolismo , Función Ventricular Izquierda/efectos de los fármacos
14.
Life Sci ; 346: 122611, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38580195

RESUMEN

Sepsis is a life-threatening condition manifested by organ dysfunction caused by a dysregulated host response to infection. Lung, brain, liver, kidney, and heart are among the affected organs. Sepsis-induced cardiomyopathy is a common cause of death among septic patients. Sepsis-induced cardiomyopathy is characterized by an acute and reversible significant decline in biventricular both systolic and diastolic function. This is accompanied by left ventricular dilatation. The pathogenesis underlying sepsis-induced cardiomyopathy is multifactorial. Hence, targeting an individual pathway may not be effective in halting the extensive dysregulated immune response. Despite major advances in sepsis management strategies, no effective pharmacological strategies have been shown to treat or even reverse sepsis-induced cardiomyopathy. Melatonin, namely, N-acetyl-5-methoxytryptamine, is synthesized in the pineal gland of mammals and can also be produced in many cells and tissues. Melatonin has cardioprotective, neuroprotective, and anti-tumor activity. Several literature reviews have explored the role of melatonin in preventing sepsis-induced organ failure. Melatonin was found to act on different pathways that are involved in the pathogenesis of sepsis-induced cardiomyopathy. Through its antimicrobial, anti-inflammatory, and antioxidant activity, it offers a potential role in sepsis-induced cardiomyopathy. Its antioxidant activity is through free radical scavenging against reactive oxygen and nitrogen species and modulating the expression and activity of antioxidant enzymes. Melatonin anti-inflammatory activities control the overactive immune system and mitigate cytokine storm. Also, it mitigates mitochondrial dysfunction, a major mechanism involved in sepsis-induced cardiomyopathy, and thus controls apoptosis. Therefore, this review discusses melatonin as a promising drug for the management of sepsis-induced cardiomyopathy.


Asunto(s)
Antioxidantes , Cardiomiopatías , Melatonina , Sepsis , Melatonina/farmacología , Melatonina/uso terapéutico , Sepsis/complicaciones , Sepsis/tratamiento farmacológico , Humanos , Cardiomiopatías/etiología , Cardiomiopatías/tratamiento farmacológico , Cardiomiopatías/prevención & control , Animales , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Cardiotónicos/farmacología , Cardiotónicos/uso terapéutico
15.
Biomed Pharmacother ; 174: 116534, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38565062

RESUMEN

The isoproterenol (ISO)-induced myocardial fibrosis is considered a reliable and repeatable experimental model characterized by a relatively low mortality rate. Although is well-known that ISO stimulates the ß1 adrenergic receptors at the myocardial level, a high degree of heterogeneity emerges around the doses and duration of the treatment generating unclear results. Therefore, we propose to gain insights into the progression of ISO-induced myocardial fibrosis, in order to critically analyze and optimize the experimental model. Male Wistar rats (12-14-week-old) were submitted to subcutaneous injection of ISO, in particular, two doses were selected: the commonly used dose of 5 mg/kg and a lower dose of 1 mg/kg, administered for 3 and 6 days. Biochemical and histological examinations were conducted either immediately after the last administration or after a recovering period of 7 or 14 days from the initial administration. Noteworthy, from our investigation emerged that even the lower dose of ISO was able to induce the maximal biochemical and histological alterations, suggesting that lower doses should be considered to control the progression of the damage more precisely and to identify a prodromic phase in which intervention with pharmacological or nutraceutical tools can be effectively attempted.


Asunto(s)
Fibrosis , Isoproterenol , Miocardio , Ratas Wistar , Animales , Masculino , Miocardio/patología , Miocardio/metabolismo , Ratas , Cardiomiopatías/inducido químicamente , Cardiomiopatías/patología , Cardiomiopatías/metabolismo , Cardiomiopatías/prevención & control , Relación Dosis-Respuesta a Droga , Modelos Animales de Enfermedad
16.
Int Heart J ; 65(2): 173-179, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38556328

RESUMEN

Keshan disease (KD) is a type of endemic cardiomyopathy with an unknown cause. It is primarily found in areas in China with low selenium levels, from northeast to southwest. The nutritional biogeochemical etiology hypothesis suggests that selenium deficiency is a major factor in KD development. Selenium is important in removing free radicals and protecting cells and tissues from peroxide-induced damage. Thus, low environmental selenium may affect the selenium level within the human body, and selenium level differences are commonly observed between healthy people in KD and nonKD areas. From the 1970s to the 1990s, China successfully reduced KD incidence in endemic KD areas through a selenium supplementation program. After years of implementing prevention and control measures, the selenium level of the population in the KD areas has gradually increased, and the prevalence of KD in China has remained low and stable in recent years. Currently, the pathogenesis of KD remains vague, and the effect of selenium supplementation on the prognosis of KD still needs further study. This paper comprehensively reviews selenium deficiency and its connection to KD. Thus, this study aims to offer novel ideas and directions to effectively prevent and treat KD in light of the current situation.


Asunto(s)
Cardiomiopatías , Infecciones por Enterovirus , Desnutrición , Selenio , Humanos , Selenio/análisis , Cardiomiopatías/epidemiología , Cardiomiopatías/etiología , Cardiomiopatías/prevención & control , Infecciones por Enterovirus/complicaciones , Infecciones por Enterovirus/epidemiología , Infecciones por Enterovirus/prevención & control , China/epidemiología
17.
J Cardiovasc Pharmacol ; 84(2): 175-187, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38547523

RESUMEN

ABSTRACT: Sepsis-induced myocardial dysfunction commonly occurs in individuals with sepsis and is a severe complication with high morbidity and mortality rates. This study aimed to investigate the effects and potential mechanisms of the natural steroidal sapogenin ruscogenin (RUS) against lipopolysaccharide (LPS)-induced myocardial injury in septic mice. We found that RUS effectively alleviated myocardial pathological damage, normalized cardiac function, and increased survival in septic mice. RNA sequencing demonstrated that RUS administration significantly inhibited the activation of the NOD-like receptor signaling pathway in the myocardial tissues of septic mice. Subsequent experiments further confirmed that RUS suppressed myocardial inflammation and pyroptosis during sepsis. In addition, cultured HL-1 cardiomyocytes were challenged with LPS, and we observed that RUS could protect these cells against LPS-induced cytotoxicity by suppressing inflammation and pyroptosis. Notably, both the in vivo and in vitro findings indicated that RUS inhibited NOD-like receptor protein-3 (NLRP3) upregulation in cardiomyocytes stimulated with LPS. As expected, knockdown of NLRP3 blocked the LPS-induced activation of inflammation and pyroptosis in HL-1 cells. Furthermore, the cardioprotective effects of RUS on HL-1 cells under LPS stimulation were abolished by the novel NLRP3 agonist BMS-986299. Taken together, our results suggest that RUS can alleviate myocardial injury during sepsis, at least in part by suppressing NLRP3-mediated inflammation and pyroptosis, highlighting the potential of this molecule as a promising candidate for sepsis-induced myocardial dysfunction therapy.


Asunto(s)
Antiinflamatorios , Modelos Animales de Enfermedad , Lipopolisacáridos , Ratones Endogámicos C57BL , Miocitos Cardíacos , Proteína con Dominio Pirina 3 de la Familia NLR , Piroptosis , Sepsis , Transducción de Señal , Espirostanos , Animales , Lipopolisacáridos/toxicidad , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Miocitos Cardíacos/metabolismo , Espirostanos/farmacología , Sepsis/tratamiento farmacológico , Sepsis/complicaciones , Sepsis/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Piroptosis/efectos de los fármacos , Masculino , Transducción de Señal/efectos de los fármacos , Línea Celular , Antiinflamatorios/farmacología , Ratones , Cardiomiopatías/prevención & control , Cardiomiopatías/patología , Cardiomiopatías/metabolismo , Cardiomiopatías/tratamiento farmacológico , Cardiomiopatías/inducido químicamente , Mediadores de Inflamación/metabolismo
19.
Circ Res ; 134(5): 505-525, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38422177

RESUMEN

BACKGROUND: Chronic overconsumption of lipids followed by their excessive accumulation in the heart leads to cardiomyopathy. The cause of lipid-induced cardiomyopathy involves a pivotal role for the proton-pump vacuolar-type H+-ATPase (v-ATPase), which acidifies endosomes, and for lipid-transporter CD36, which is stored in acidified endosomes. During lipid overexposure, an increased influx of lipids into cardiomyocytes is sensed by v-ATPase, which then disassembles, causing endosomal de-acidification and expulsion of stored CD36 from the endosomes toward the sarcolemma. Once at the sarcolemma, CD36 not only increases lipid uptake but also interacts with inflammatory receptor TLR4 (Toll-like receptor 4), together resulting in lipid-induced insulin resistance, inflammation, fibrosis, and cardiac dysfunction. Strategies inducing v-ATPase reassembly, that is, to achieve CD36 reinternalization, may correct these maladaptive alterations. For this, we used NAD+ (nicotinamide adenine dinucleotide)-precursor nicotinamide mononucleotide (NMN), inducing v-ATPase reassembly by stimulating glycolytic enzymes to bind to v-ATPase. METHODS: Rats/mice on cardiomyopathy-inducing high-fat diets were supplemented with NMN and for comparison with a cocktail of lysine/leucine/arginine (mTORC1 [mechanistic target of rapamycin complex 1]-mediated v-ATPase reassembly). We used the following methods: RNA sequencing, mRNA/protein expression analysis, immunofluorescence microscopy, (co)immunoprecipitation/proximity ligation assay (v-ATPase assembly), myocellular uptake of [3H]chloroquine (endosomal pH), and [14C]palmitate, targeted lipidomics, and echocardiography. To confirm the involvement of v-ATPase in the beneficial effects of both supplementations, mTORC1/v-ATPase inhibitors (rapamycin/bafilomycin A1) were administered. Additionally, 2 heart-specific v-ATPase-knockout mouse models (subunits V1G1/V0d2) were subjected to these measurements. Mechanisms were confirmed in pharmacologically/genetically manipulated cardiomyocyte models of lipid overload. RESULTS: NMN successfully preserved endosomal acidification during myocardial lipid overload by maintaining v-ATPase activity and subsequently prevented CD36-mediated lipid accumulation, CD36-TLR4 interaction toward inflammation, fibrosis, cardiac dysfunction, and whole-body insulin resistance. Lipidomics revealed C18:1-enriched diacylglycerols as lipid class prominently increased by high-fat diet and subsequently reversed/preserved by lysine/leucine/arginine/NMN treatment. Studies with mTORC1/v-ATPase inhibitors and heart-specific v-ATPase-knockout mice further confirmed the pivotal roles of v-ATPase in these beneficial actions. CONCLUSION: NMN preserves heart function during lipid overload by preventing v-ATPase disassembly.


Asunto(s)
Cardiomiopatías , Resistencia a la Insulina , Animales , Ratones , Ratas , Adenosina Trifosfatasas , Arginina , Cardiomiopatías/inducido químicamente , Cardiomiopatías/prevención & control , Antígenos CD36/genética , Fibrosis , Inflamación , Leucina , Lípidos , Lisina , Diana Mecanicista del Complejo 1 de la Rapamicina , Miocitos Cardíacos , Mononucleótido de Nicotinamida , Receptor Toll-Like 4/genética
20.
BMC Anesthesiol ; 24(1): 72, 2024 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-38395800

RESUMEN

BACKGROUND: Approximately 40 to 60% of patients with sepsis develop sepsis-induced cardiomyopathy (SIC), which is associated with a substantial increase in mortality. We have found that molecular hydrogen (H2) inhalation improved the survival rate and cardiac injury in septic mice. However, the mechanism remains unclear. This study aimed to explore the regulatory mechanism by which hydrogen modulates autophagy and its role in hydrogen protection of SIC. METHODS: Cecal ligation and puncture (CLP) was used to induce sepsis in adult C57BL/6J male mice. The mice were randomly divided into 4 groups: Sham, Sham + 2% hydrogen inhalation (H2), CLP, and CLP + H2 group. The 7-day survival rate was recorded. Myocardial pathological scores were calculated. Myocardial troponin I (cTnI) levels in serum were detected, and the levels of autophagy- and mitophagy-related proteins in myocardial tissue were measured. Another four groups of mice were also studied: CLP, CLP + Bafilomycin A1 (BafA1), CLP + H2, and CLP + H2 + BafA1 group. Mice in the BafA1 group received an intraperitoneal injection of the autophagy inhibitor BafA1 1 mg/kg 1 h after operation. The detection indicators remained the same as before. RESULTS: The survival rate of septic mice treated with H2 was significantly improved, myocardial tissue inflammation was improved, serum cTnI level was decreased, autophagy flux was increased, and mitophagy protein content was decreased (P < 0.05). Compared to the CLP + H2 group, the CLP + H2 + BafA1 group showed a decrease in autophagy level and 7-day survival rate, an increase in myocardial tissue injury and cTnI level, which reversed the protective effect of hydrogen (P < 0.05). CONCLUSION: Hydrogen exerts protective effect against SIC, which may be achieved through the promotion of autophagy and mitophagy.


Asunto(s)
Cardiomiopatías , Sepsis , Humanos , Ratones , Masculino , Animales , Ratones Endogámicos C57BL , Autofagia , Cardiomiopatías/etiología , Cardiomiopatías/prevención & control , Sepsis/complicaciones , Sepsis/patología , Hidrógeno/farmacología , Hidrógeno/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA