Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Ann Vasc Surg ; 105: 227-235, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38609009

RESUMO

BACKGROUND: Studies have linked matrix metalloproteinases (MMPs) to both thoracic aortic aneurysm and abdominal aortic aneurysm (TAA and AAA). The precise MMPs entailed in this procedure, however, were still unknown. This study used a two-sample Mendelian randomization (MR) analysis to look into the causal relationship between MMPs and the risk of TAA and AAA. METHODS: Eight MMPs, including MMP-1, MMP-2, MMP-3, MMP-8, MMP-9, MMP-10, MMP-12, and MMP-13, were found among people of European ancestry with accessible Genome-Wide Association Studies (GWAS). We employed the findings from Genome-Wide Association Studies (GWAS) for 8 MMPs, and TAA and AAA from the FinnGen consortiums (3,201 cases and 317,899 controls, respectively) were used in a two-sample MR analysis. The primary method of analysis for MR was the inverse variance weighted (IVW) method, along with analyses of heterogeneity and horizontal pleiotropy. 31 single-nucleotide polymorphisms connected to MMP were retrieved. RESULTS: IVW demonstrated a negative causal association between TAA and AAA and serum MMP-12 levels. The incidence of TAA decreased by 1.031% for every 1 ng/mL increase in serum MMP-12 [odds ratio (OR) = 0.897, 95% confidence interval (CI): 0.831-0.968, P = 0.005]. The incidence of AAA fell by 1.653% (OR = 0.835, 95% CI: 0.752-0.926, P = 0.001) for every 1 ng/mL increase in serum MMP-12. There was no horizontal pleiotropy or heterogeneity in the MR data (P > 0.05). CONCLUSIONS: The levels of TAA and AAA and serum MMP-12 are causally related. MMP-12 is a factor that reduces the risk of AAA and TTA. Our study suggested that MMP-12 level is causally associated with a decreased risk of TAA and AAA.


Assuntos
Aneurisma da Aorta Abdominal , Aneurisma da Aorta Torácica , Metaloproteinases da Matriz , Humanos , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/enzimologia , Aneurisma da Aorta Abdominal/diagnóstico por imagem , Aneurisma da Aorta Abdominal/sangue , Aneurisma da Aorta Abdominal/epidemiologia , Aneurisma da Aorta Torácica/genética , Aneurisma da Aorta Torácica/enzimologia , Aneurisma da Aorta Torácica/sangue , Aneurisma da Aorta Torácica/diagnóstico por imagem , Aneurisma da Aorta Torácica/epidemiologia , Estudos de Casos e Controles , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Incidência , Metaloproteinase 12 da Matriz/genética , Metaloproteinase 12 da Matriz/sangue , Metaloproteinases da Matriz/genética , Metaloproteinases da Matriz/sangue , Análise da Randomização Mendeliana , Fenótipo , Polimorfismo de Nucleotídeo Único , Fatores de Proteção , Medição de Risco , Fatores de Risco
2.
Front Cardiovasc Med ; 10: 1233147, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37790597

RESUMO

Background: The lactate to albumin ratio (LAR) has emerged as a promising prognostic marker in critically ill patients. Despite its potential utility, the prognostic value of LAR in septic myocardial injury (SMI) remains uncertain. Methods: This study aims to investigate the prognostic significance of LAR in SMI through a retrospective cohort analysis of data from the Medical Information Mart for Intensive Care III (MIMIC-III) (v1.4) database. The study included intensive care unit (ICU)-admitted patients (age ≥18 years) diagnosed with SMI. The primary endpoint was in-hospital mortality. Results: A total of 704 patients were included in the study, of which 59.10% were male. Hospital mortality and ICU mortality rates were recorded at 29.97% and 22.87%, respectively. After adjusting for confounding factors, multivariate Cox proportional risk analysis demonstrated that LAR was independently associated with an increased risk of both hospital mortality (HR, 1.39 [95% CI: 1.24-1.56] P < 0.001) and ICU mortality (HR, 1.46 [95% CI: 1.29-1.65] P < 0.001). Furthermore, the generalized additive model (GAM) and restricted cubic spline (RCS) model indicated a linear relationship between LAR and mortality rates in the ICU and hospital. Conclusions: The LAR may serve as a potential prognostic biomarker in critically ill patients with SMI. High LAR levels are associated with a higher risk of in-hospital mortality and can help identify individuals with high mortality rates. Overall, the findings emphasize the importance of using LAR as a tool for risk stratification and management of critically ill patients with SMI.

3.
Int J Mol Med ; 52(4)2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37654208

RESUMO

Necroptosis, which is distinct from apoptosis and necrosis, serves a crucial role in ontogeny and the maintenance of homeostasis. In the last decade, it has been demonstrated that the pathogenesis of cardiovascular diseases is also linked to necroptosis. Receptor interaction protein kinase (RIPK) 1, RIPK3 and mixed lineage kinase domain­like protein serve vital roles in necroptosis. In addition to the aforementioned necroptosis­related components, calcium/calmodulin­dependent protein kinase II (CaMKII) has been identified as a novel substrate for RIPK3 that promotes the opening of the mitochondrial permeability transition pore (mPTP), and thus, mediates necroptosis of myocardial cells through the RIPK3­CaMKII­mPTP signaling pathway. The present review provides an overview of the current knowledge of the RIPK3­CaMKII­mPTP­mediated necroptosis signaling pathway in cardiovascular diseases, focusing on the role of the RIPK3­CaMKII­mPTP signaling pathway in acute myocardial infarction, ischemia­reperfusion injury, heart failure, abdominal aortic aneurysm, atherosclerosis, diabetic cardiomyopathy, hypertrophic cardiomyopathy, atrial fibrillation, and the cardiotoxicity associated with antitumor drugs and other chemicals. Finally, the present review discusses the research status of drugs targeting the RIPK3­CaMKII­mPTP signaling pathway.


Assuntos
Doenças Cardiovasculares , Insuficiência Cardíaca , Humanos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Necroptose , Transdução de Sinais , Proteína Serina-Treonina Quinases de Interação com Receptores
5.
Int J Biol Sci ; 18(14): 5276-5290, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36147470

RESUMO

In diabetic cardiomyopathy (DCM), a major diabetic complication, the myocardium is structurally and functionally altered without evidence of coronary artery disease, hypertension or valvular disease. Although numerous anti-diabetic drugs have been applied clinically, specific medicines to prevent DCM progression are unavailable, so the prognosis of DCM remains poor. Mitochondrial ATP production maintains the energetic requirements of cardiomyocytes, whereas mitochondrial dysfunction can induce or aggravate DCM by promoting oxidative stress, dysregulated calcium homeostasis, metabolic reprogramming, abnormal intracellular signaling and mitochondrial apoptosis in cardiomyocytes. In response to mitochondrial dysfunction, the mitochondrial quality control (MQC) system (including mitochondrial fission, fusion, and mitophagy) is activated to repair damaged mitochondria. Physiological mitochondrial fission fragments the network to isolate damaged mitochondria. Mitophagy then allows dysfunctional mitochondria to be engulfed by autophagosomes and degraded in lysosomes. However, abnormal MQC results in excessive mitochondrial fission, impaired mitochondrial fusion and delayed mitophagy, causing fragmented mitochondria to accumulate in cardiomyocytes. In this review, we summarize the molecular mechanisms of MQC and discuss how pathological MQC contributes to DCM development. We then present promising therapeutic approaches to improve MQC and prevent DCM progression.


Assuntos
Diabetes Mellitus , Cardiomiopatias Diabéticas , Trifosfato de Adenosina/metabolismo , Cálcio/metabolismo , Diabetes Mellitus/metabolismo , Cardiomiopatias Diabéticas/tratamento farmacológico , Cardiomiopatias Diabéticas/metabolismo , Humanos , Mitocôndrias/metabolismo , Mitofagia
6.
Metabolism ; 137: 155313, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36126721

RESUMO

Mitochondrial dysfunction has been regarded as a hallmark of diabetic cardiomyopathy. In addition to their canonical metabolic actions, mitochondria influence various other aspects of cardiomyocyte function, including oxidative stress, iron regulation, metabolic reprogramming, intracellular signaling transduction and cell death. These effects depend on the mitochondrial quality control (MQC) system, which includes mitochondrial dynamics, mitophagy and mitochondrial biogenesis. Mitochondria are not static entities, but dynamic units that undergo fission and fusion cycles to maintain their structural integrity. Increased mitochondrial fission elevates the number of mitochondria within cardiomyocytes, a necessary step for cardiomyocyte metabolism. Enhanced mitochondrial fusion promotes communication and cooperation between pairs of mitochondria, thus facilitating mitochondrial genomic repair and maintenance. On the contrary, erroneous fission or reduced fusion promotes the formation of mitochondrial fragments that contain damaged mitochondrial DNA and exhibit impaired oxidative phosphorylation. Under normal/physiological conditions, injured mitochondria can undergo mitophagy, a degradative process that delivers poorly structured mitochondria to lysosomes. However, defective mitophagy promotes the accumulation of nonfunctional mitochondria, which may induce cardiomyocyte death. A decline in the mitochondrial population due to mitophagy can stimulate mitochondrial biogenesis), which generates new mitochondrial offspring to maintain an adequate mitochondrial number. Energy crises or ATP deficiency also increase mitochondrial biogenesis, because mitochondrial DNA encodes 13 subunits of the electron transport chain (ETC) complexes. Disrupted mitochondrial biogenesis diminishes the mitochondrial mass, accelerates mitochondrial senescence and promotes mitochondrial dysfunction. In this review, we describe the involvement of MQC in the pathogenesis of diabetic cardiomyopathy. Besides, the potential targeted therapies that could be applied to improve MQC during diabetic cardiomyopathy are also discussed and accelerate the development of cardioprotective drugs for diabetic patients.


Assuntos
Diabetes Mellitus , Cardiomiopatias Diabéticas , Humanos , Cardiomiopatias Diabéticas/metabolismo , Mitofagia/genética , Mitocôndrias/metabolismo , Dinâmica Mitocondrial/fisiologia , DNA Mitocondrial/genética , Diabetes Mellitus/metabolismo
8.
Drug Chem Toxicol ; 45(4): 1443-1448, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33108916

RESUMO

Prolonged survival and expanded treatment options in myeloma patients have led to adverse events associated with treatment getting increased attention. This systematic review and meta-analysis aimed to determine the incidence of ixazomib-associated cardiovascular adverse events (CVAEs) and to compare the rates of ixazomib-associated CVAEs and related therapies. CVAEs were defined as heart failure, hypertension, ischemia, and arrhythmia. All-grade and high-grade CVAEs and study characteristics were recorded. A total of 266 potentially relevant articles were identified, and 246 were excluded after review. Twenty studies of 1715 patients with multiple myeloma were thus considered in this study. The estimated rates of all-grade and high-grade ixazomib associated CVAEs were 11.2 and 3.7%, respectively. Subgroup analysis showed that median age ≥65 years, none phase 1 trial and combination regimen were associated with higher rates of high-grade ixazomib associated CVAEs. Ixazomib was association with increased high-grade CVAEs risk (RR = 1.679, 95% CI: 1.078-2.615, P = 0.022). Ixazomib was associated with a significant rate of high-grade CVAEs. Future studies are needed to identify patients at high risk for high-grade CVAEs.


Assuntos
Hipertensão , Mieloma Múltiplo , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica , Compostos de Boro/efeitos adversos , Glicina/efeitos adversos , Glicina/análogos & derivados , Humanos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/epidemiologia
9.
Oxid Med Cell Longev ; 2021: 5589612, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33763168

RESUMO

Endothelial dysfunction, which is characterized by damage to the endoplasmic reticulum (ER) and mitochondria, is involved in a variety of cardiovascular disorders. Here, we explored whether mitochondrial damage and ER stress are associated with endothelial dysfunction. We also examined whether and how melatonin protects against oxidized low-density lipoprotein- (ox-LDL-) induced damage in endothelial cells. We found that CHOP, GRP78, and PERK expressions, which are indicative of ER stress, increased significantly in response to ox-LDL treatment. ox-LDL also induced mitochondrial dysfunction as evidenced by decreased mitochondrial membrane potential, increased mitochondrial ROS levels, and downregulation of mitochondrial protective factors. In addition, ox-LDL inhibited antioxidative processes, as evidenced by decreased antioxidative enzyme activity and reduced Nrf2/HO-1 expression. Melatonin clearly reduced ER stress and promoted mitochondrial function and antioxidative processes in the presence of ox-LDL. Molecular investigation revealed that ox-LDL activated the JNK/Mff signaling pathway, and melatonin blocked this effect. These results demonstrate that ox-LDL induces ER stress and mitochondrial dysfunction and activates the JNK/Mff signaling pathway, thereby contributing to endothelial dysfunction. Moreover, melatonin inhibited JNK/Mff signaling and sustained ER homeostasis and mitochondrial function, thereby protecting endothelial cells against ox-LDL-induced damage.


Assuntos
Estresse do Retículo Endoplasmático/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Lipoproteínas LDL/farmacologia , Melatonina/farmacologia , Proteínas de Membrana/metabolismo , Proteínas Mitocondriais/metabolismo , Transdução de Sinais , Chaperona BiP do Retículo Endoplasmático , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
10.
Aging (Albany NY) ; 12(13): 13618-13632, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32645692

RESUMO

Mitochondria and the endoplasmic reticulum (ER) are known to promote cardiac ischemia/reperfusion (I/R) injury. Overexpression of yes-associated protein (YAP) and/or sarcoplasmic reticulum calcium ATPase 2a (SERCA2a) has been shown to protect cardiomyocytes against I/R-induced injury. Here, we show that activation of the YAP/SERCA2a pathway attenuated mitochondrial damage and ER stress (ERS) to maintain cardiomyocyte viability in the setting of I/R injury. Our results demonstrate that I/R treatment reduced the transcription and expression of YAP and SERCA2a, along with a decline in cardiomyocyte viability. The overexpression of YAP promoted SERCA2a transcription, whereas SERCA2a upregulation did not affect the YAP transcription, suggesting that YAP functions upstream of SERCA2a. Activation of the YAP/SERCA2a pathway suppressed mitochondrial damage by sustaining the mitochondrial redox balance and restoring mitochondrial bioenergetics. Additionally, its activation repressed ERS, reduced calcium overload, and eventually blocked caspase activation. The knockdown of SERCA2a suppressed the protective effects of YAP overexpression on mitochondrial damage and ERS. Overall, our findings reveal that the YAP/SERCA2a pathway attenuates the mitochondrial damage and ERS in response to cardiac I/R injury by regulating the mitochondria-ER communication.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ciclo Celular/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/patologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Transdução de Sinais/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Apoptose/genética , Proteínas de Ciclo Celular/genética , Hipóxia Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Retículo Endoplasmático/patologia , Estresse do Retículo Endoplasmático/fisiologia , Técnicas de Silenciamento de Genes , Camundongos , Mitocôndrias/patologia , Miocárdio/citologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Cultura Primária de Células , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Proteínas de Sinalização YAP
11.
Int Immunopharmacol ; 81: 106204, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32086130

RESUMO

BACKGROUND: Accumulating evidence has revealed the roles of microRNAs (miRs) in sepsis, hence, the aim of the present study was to investigate whether miR-208a-5p affects sepsis whilst attempting to elucidate the mechanisms by which the suppressors of cytokine signaling 2 (SOCS2)-mediated nuclear factor-kappaB/hypoxia-inducible factor-1α (NF-κB/HIF-1α) pathway is implicated in this process. METHODS: The sepsis model was established by cecal ligation and puncture in mice. Serum levels of myocardial enzyme cardiac Troponin-I (cTnI) and brain natriuretic peptide (BNP) in mice were measured. Malondialdehyde (MDA), lactate dehydrogenase (LDH) activity, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), NF-κB p65, HIF-1α and superoxidedismutase (SOD) activity in myocardial tissues were determined. Furthermore, the swelling degree of mitochondria and the apoptosis of cardiomyocytes was measured. The expression of miR-208a-5p, SOCS2, Bcl-2, Bax, NF-κB p65 and HIF-1α in myocardial tissues of mice were detected. RESULTS: Down-regulation of miR-208a-5p and up-regulation of SOCS2 raised the activity of SOD, while reduced the activity of LDH and MDA and the concentrations of cTnI, BNP, TNF-α, IL-6, NF-κB p65 and HIF-1α in mice with sepsis. Down-regulated miR-208a-5p and up-regulated SOCS2 reduced degree of mitochondria swelling, and suppressed cardiomyocytes apoptosis in mice with sepsis. MiR-208a-5p, NF-κB p65 and HIF-1α expression were raised while SOCS2 expression was depressed in myocardial tissues of mice with sepsis. CONCLUSION: This study suggests that high expression of SOCS2 or inhibition of miR-208a-5p alleviates the myocardial injury of sepsis mice via modulating NF-κB/HIF-1α pathway, which are potential candidate markers and therapeutic targets for sepsis mice.


Assuntos
MicroRNAs/genética , Miocárdio/metabolismo , Sepse/genética , Animais , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos , Terapia de Alvo Molecular , Miocárdio/patologia , NF-kappa B/metabolismo , Sepse/metabolismo , Sepse/terapia , Transdução de Sinais , Proteínas Supressoras da Sinalização de Citocina/metabolismo
12.
J Cell Physiol ; 235(10): 6647-6659, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-31976559

RESUMO

Despite significant advances in therapies in past decades, the mortality rate of septic cardiomyopathy remains high. The aim of this study is to explore the therapeutic effects of combined treatment using melatonin and irisin in a mouse model of lipopolysaccharide (LPS)-mediated septic cardiomyopathy. Our data found that melatonin and irisin could further attenuate LPS-induced myocardial depression. Molecular investigation illustrated that melatonin and irisin cotreatment sustained cardiomyocyte viability and improved mitochondrial function under LPS stress. Pathway analysis demonstrated that macrophage-stimulating 1 (Mst1), which was significantly activated by LPS, was drastically inhibited by melatonin/irisin cotreatment. Mechanically, Mst1 activated c-Jun N-terminal kinase (JNK) pathway and the latter induced oxidative stress, adenosine triphosphate metabolism disorder, mitochondrial membrane potential reduction, and cardiomyocyte death activation. Melatonin and irisin cotreatment effectively inhibited the Mst1-JNK pathway and, thus, promoted cardiomyocyte survival and mitochondrial homeostasis. Interestingly, Mst1 overexpression abolished the beneficial effects of melatonin and irisin in vivo and in vitro. Altogether, our results confirmed that melatonin and irisin combination treatment could protect heart against sepsis-induced myocardial depression via modulating the Mst1-JNK pathways.


Assuntos
Cardiomiopatias/induzido quimicamente , Cardiomiopatias/tratamento farmacológico , Fibronectinas/farmacologia , Fator de Crescimento de Hepatócito/metabolismo , Lipopolissacarídeos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Melatonina/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Cardiomiopatias/metabolismo , Células Cultivadas , Coração/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Sepse/tratamento farmacológico , Sepse/metabolismo , Transdução de Sinais/efeitos dos fármacos
13.
Cell Stress Chaperones ; 24(5): 991-1003, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31388827

RESUMO

Cardiac microvascular ischemia-reperfusion (IR) injury has been a neglected topic in recent decades. In the current study, we investigated the mechanism underlying microvascular IR injury, with a focus on mitochondrial homeostasis. We also explored the protective role of tanshinone IIA (Tan IIA) in microvascular protection in the context of IR injury. Through animal studies and cell experiments, we demonstrated that IR injury mediated microvascular wall destruction, lumen stenosis, perfusion defects, and cardiac microvascular endothelial cell (CMEC) apoptosis via inducing mitochondrial damage. In contrast, Tan IIA administration had the ability to sustain CMEC viability and microvascular homeostasis, finally attenuating microvascular IR injury. Function studies have confirmed that the SIRT1/PGC1α pathway is responsible for the microvascular protection from the Tan IIA treatment. SIRT1 activation by Tan IIA sustained the mitochondrial potential, alleviated the mitochondrial pro-apoptotic factor leakage, reduced the mPTP opening, and blocked mitochondrial apoptosis, providing a survival advantage for CMECs and preserving microvascular structure and function. By comparison, inhibiting SIRT1 abrogated the beneficial effects of Tan IIA on mitochondrial function, CMEC survival, and microvascular homeostasis. Collectively, this study indicated that Tan IIA should be considered a microvascular-protective drug that alleviates acute cardiac microcirculation IR injury via activating the SIRT1/PGC1α pathway and thereby blocking mitochondrial damage.


Assuntos
Abietanos/farmacologia , Apoptose/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Animais , Coração/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Sirtuína 1/metabolismo
14.
Redox Biol ; 26: 101287, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31386965

RESUMO

The basic pathophysiological mechanisms underlying septic cardiomyopathy have not yet been completely clarified. Disease-specific treatments are lacking, and care is still based on supportive modalities. The aim of our study was to assess the protective effects of melatonin on septic cardiomyopathy, with a focus on the interactions between receptor-interacting protein kinase 3 (Ripk3), the mitochondria, endoplasmic reticulum (ER) and cytoskeletal degradation in cardiomyocytes. Ripk3 expression was increased in heart samples challenged with LPS, followed by myocardial inflammation, cardiac dysfunction, myocardial breakdown and cardiomyocyte death. The melatonin treatment attenuated septic myocardial injury in a comparable manner to the genetic depletion of Ripk3. Molecular investigations revealed that Ripk3 intimately regulated mitochondrial function, ER stress, cytoskeletal homeostasis and cardioprotective signaling pathways. Melatonin-mediated inhibition of Ripk3 improved mitochondrial bioenergetics, reduced mitochondria-initiated oxidative damage, sustained mitochondrial dynamics, ameliorated ER stress, normalized calcium recycling, and activated cardioprotective signaling pathways (including AKT, ERK and AMPK) in cardiomyocytes. Interestingly, Ripk3 overexpression mediated resistance to melatonin therapy following the infection of LPS-treated hearts with an adenovirus expressing Ripk3. Altogether, our findings identify Ripk3 upregulation as a novel risk factor for the development of sepsis-related myocardial injury, and melatonin restores the physiological functions of the mitochondria, ER, contractile cytoskeleton and cardioprotective signaling pathways. Additionally, our data also reveal a new, potentially therapeutic mechanism by which melatonin protects the heart from sepsis-mediated dysfunction, possibly by targeting Ripk3.


Assuntos
Cardiomiopatias/etiologia , Cardiomiopatias/metabolismo , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Melatonina/farmacologia , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Sepse/complicações , Cardiomiopatias/tratamento farmacológico , Cardiomiopatias/patologia , Cardiotônicos/farmacologia , Citoesqueleto/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Deleção de Genes , Humanos , Lipopolissacarídeos/imunologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
15.
Curr Pharm Des ; 25(18): 2060-2070, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31284854

RESUMO

BACKGROUND: Sepsis and septic shock are known to prompt multiple organ failure including cardiac contractile dysfunction, which is typically referred to as septic cardiomyopathy. Among various theories postulated for the etiology of septic cardiomyopathy, mitochondrial injury (both morphology and function) in the heart is perceived as the main culprit for reduced myocardial performance and ultimately heart failure in the face of sepsis. METHODS: Over the past decades, ample of experimental and clinical work have appeared, focusing on myocardial mitochondrial changes and related interventions in septic cardiomyopathy. RESULTS AND CONCLUSION: Here we will briefly summarize the recent experimental and clinical progress on myocardial mitochondrial morphology and function in sepsis, and discuss possible underlying mechanisms, as well as the contemporary interventional options.


Assuntos
Cardiomiopatias/etiologia , Mitocôndrias Cardíacas/patologia , Sepse/complicações , Choque Séptico/complicações , Coração , Humanos
16.
J Mol Histol ; 50(5): 405-415, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31256303

RESUMO

Viral myocarditis has been found to be one of the leading causes of sudden death in young adults. However, no effective drugs have been developed to intervene the progression of myocarditis. Accordingly, the present study is carried out to explore the protective role played by melatonin in the setting of viral myocarditis with a focus on Mst1-Hippo pathway, mitochondrial dysfunction and ER stress. Cardiac function was determined via echocardiographic examination. Mitochondrial function and ER stress were detected via ELISA, western blots, and immunofluorescence. Our data demonstrated that virus injection induced cardiac dysfunction as evidenced by reduced contractile function in myocardium. Besides, LDH release assay and western blotting analysis demonstrated that cardiomyocyte death was activated by virus injection. Interestingly, melatonin treatment improved cardiac function and repressed virus-mediated cardiomyocyte apoptosis. At the molecular levels, mitochondrial dysfunction was induced by virus infection, as indicated by mitochondrial membrane potential reduction, mPTP opening rate elevation and caspase-9-related apoptosis activation. Besides, ER stress parameters were also elevated in virus-treated cardiomyocytes. Interestingly, melatonin treatment maintained mitochondrial dysfunction and repressed ER stress. To the end, we found that Mst1 was upregulated by virus infection; this effect was attenuated through supplementation with melatonin. However, Mst1 overexpression reduced the beneficial impact exerted by melatonin on cardiomyocyte viability, mitochondrial function and ER homeostasis. Our study illustrated that melatonin treatment attenuated viral myocarditis via sustaining cardiomyocyte viability, repressing mitochondrial dysfunction and inhibiting ER stress in a manner dependent on Mst1 inhibition.


Assuntos
Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Melatonina/farmacologia , Mitocôndrias/efeitos dos fármacos , Miocardite/prevenção & controle , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Animais , Sobrevivência Celular/efeitos dos fármacos , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Mitocôndrias/patologia , Miócitos Cardíacos/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , Vírus/patogenicidade
17.
Cell Stress Chaperones ; 24(3): 595-608, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30993599

RESUMO

Irisin plays a protective effect in acute and chronic myocardial damage, but its role in septic cardiomyopathy is unclear. The aim of our study was to explore the in vivo and in vitro effects of irisin using an LPS-induced septic cardiomyopathy model. Our results demonstrated that irisin treatment attenuated LPS-mediated cardiomyocyte death and myocardial dysfunction. At the molecular level, LPS application was associated with mitochondrial oxidative injury, cardiomyocyte ATP depletion and caspase-related apoptosis activation. In contrast, the irisin treatment sustained mitochondrial function by inhibiting DRP1-related mitochondrial fission and the reactivation of mitochondrial fission impaired the protective action of irisin on inflammation-attacked mitochondria and cardiomyocytes. Additionally, we found that irisin modulated DRP1-related mitochondrial fission through the JNK-LATS2 signaling pathway. JNK activation and/or LATS2 overexpression abolished the beneficial effects of irisin on LPS-mediated mitochondrial stress and cardiomyocyte death. Altogether, our results illustrate that LPS-mediated activation of DRP1-related mitochondrial fission through the JNK-LATS2 pathway participates in the pathogenesis of septic cardiomyopathy. Irisin could be used in the future as an effective therapy for sepsis-induced myocardial depression because it corrects DRP1-related mitochondrial fission and normalizes the JNK-LATS2 signaling pathway.


Assuntos
Cardiomiopatias/tratamento farmacológico , Dinaminas/metabolismo , Fibronectinas/farmacologia , Mitocôndrias/efeitos dos fármacos , Dinâmica Mitocondrial/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Animais , Cardiomiopatias/induzido quimicamente , Lipopolissacarídeos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Mitocôndrias/patologia , Miócitos Cardíacos/patologia
18.
Chemosphere ; 197: 241-250, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29353674

RESUMO

A environment-friendly 3D inorganic heteropoly blue (HPB) Ba2Na2 [HPWV4WVI8O40]·26H2O was directly synthesized by hydrothermal method and characterized by means of ICP, IR, XPS, X-ray single crystal and X-ray powder diffraction. It was an efficient heterogeneous photo-Fenton-like catalyst to degrade anionic dye methyl orange under visible light irradiation. It removed cationic dyes methylene blue in neutral environment and rhodamine B in acidic condition via flocculation. The removal efficiency of methylene blue and rhodamine B by flocculation was more than 95%. Moreover, it could degrade methyl orange and flocculate rhodamine B at the same time. For MO and MO-RhB solutions, the degradation rates of MO in 60 min were 85.5% and 49.1%, respectively. Furthermore, the possible pathways for the production of active species in the MO degradation reaction were discussed. This is the first HPB constructed with 4e-reduced phosphotungstate, Ba and Na ions, having the properties of photo-Fenton-like catalyst and flocculant.


Assuntos
Compostos Azo/química , Recuperação e Remediação Ambiental/métodos , Peróxido de Hidrogênio/química , Ferro/química , Azul de Metileno/química , Rodaminas/química , Compostos de Tungstênio/química , Poluentes Químicos da Água/química , Catálise , Corantes/química , Luz , Compostos de Tungstênio/síntese química , Poluentes Químicos da Água/análise , Difração de Raios X
19.
Cardiovasc J Afr ; 26(3): e1-3, 2015 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-26592989

RESUMO

Systemic inflammatory response syndrome (SIRS) complicated with ST-elevation myocardial infarction has rarely been reported, and the precise mechanisms of myocardial injury remain unclear. Here, we present a case involving a 45-year-old man who developed SIRS secondary to diabetes-induced infection, and who ultimately developed ST-elevation myocardial infarction with acute heart failure, fulminant diabetes, acute liver dysfunction, acute kidney dysfunction and rhabdomyolysis. The patient eventually recovered due to early detection, correct diagnosis and powerful treatment. Clinicians should be aware of this new type of myocardial infarction, which is induced by inflammatory injury, but is not due to a primary coronary event such as plaque erosion and/or rupture, fissuring or dissection.


Assuntos
Complicações do Diabetes/etiologia , Infarto do Miocárdio/etiologia , Pneumonia Bacteriana/etiologia , Síndrome de Resposta Inflamatória Sistêmica/etiologia , Angiografia Coronária , Complicações do Diabetes/diagnóstico , Complicações do Diabetes/terapia , Eletrocardiografia , Humanos , Masculino , Pessoa de Meia-Idade , Insuficiência de Múltiplos Órgãos/etiologia , Infarto do Miocárdio/diagnóstico , Infarto do Miocárdio/terapia , Pneumonia Bacteriana/diagnóstico , Pneumonia Bacteriana/terapia , Valor Preditivo dos Testes , Fatores de Risco , Síndrome de Resposta Inflamatória Sistêmica/diagnóstico , Síndrome de Resposta Inflamatória Sistêmica/terapia , Resultado do Tratamento , Ultrassonografia de Intervenção
20.
Int J Mol Med ; 35(3): 715-23, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25604880

RESUMO

Rosiglitazone has been found to have anti-atherogenic effects and to increase serum high-density lipoprotein (HDL) cholesterol (HDL-C) levels. However, in vivo studies investigating the regulation of adenosine triphosphate-binding cassette transporter A1 (ABCA1) and scavenger receptor class B type I (SR-BI) by rosiglitazone are limited. Moreover, the effects of rosiglitazone on the function and levels of HDL are unclear. In the present study, we investigated the effects of rosiglitazone on HDL function and its mechanisms of action in atherosclerotic rabbits. Our results revealed that rosiglitazone induced a significant increase in serum HDL-C levels, paraoxonase 1 (PON1) activity, [(3)H]cholesterol efflux rates, and the expression of ABCA1 and SR-BI in hepatocytes and peritoneal macrophages. The expression of ABCA1 was also increased in aortic lesions. Rosiglitazone markedly reduced serum myeloperoxidase (MPO) activity, aortic intima-media thickness (IMT) and the percentage of plaque area in the aorta. It can thus be concluded that in atherosclerotic rabbits, rosigitazone increases the levels of HDL-C and hinders atherosclerosis. Thus, it improves HDL quality and function, as well as the HDL-induced cholesterol efflux, exerting anti-inflammatory and antioxidant effects.


Assuntos
Aterosclerose/metabolismo , Hipoglicemiantes/farmacologia , Lipoproteínas HDL/metabolismo , Tiazolidinedionas/farmacologia , Transportador 1 de Cassete de Ligação de ATP/genética , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Animais , Arildialquilfosfatase/metabolismo , Aterosclerose/tratamento farmacológico , Aterosclerose/genética , Aterosclerose/patologia , Colesterol/metabolismo , Modelos Animais de Doenças , Ativação Enzimática/efeitos dos fármacos , Expressão Gênica , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Lipídeos/sangue , Lipoproteínas HDL/sangue , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/metabolismo , Masculino , Peroxidase/metabolismo , Coelhos , Rosiglitazona , Receptores Depuradores Classe B/genética , Receptores Depuradores Classe B/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...