Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 513
Filtrar
1.
J Cardiothorac Surg ; 19(1): 553, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39354539

RESUMO

OBJECTIVE: We aim to investigate the association between prognosis and outcomes following myocardial ischemia-reperfusion injury, as well as peripheral blood levels of NLRP3 and the triglyceride-glucose index (TyG). METHODS: A total of 100 patients who underwent emergency coronary intervention following myocardial infarction confirmed by coronary angiography at our hospital between October 2021 and May 2023 were included in this study. Patients were stratified into two groups based on their prognoses: the control group (n = 73), which did not experience new myocardial infarctions or require hospitalization for heart failure or suffer sudden cardiac death post-interventional treatment; and the observation group (n = 27), which experienced one or more cardiovascular events post-treatment. Patient demographics were obtained from clinical records while biochemical analyses assessed peripheral blood triglycerides, blood glucose levels, and TyG index. Additionally, ELISA measurements determined levels of NLRP3 as well as inflammatory factors IL-6, TNF-α, and CRP in peripheral blood samples. Cardiac function was evaluated according to NYHA standards. Univariable Cox regression analysis identified factors influencing patient prognosis while Pearson correlation analysis examined relationships among prognosis, outcomes following myocardial ischemia-reperfusion injury, TyG index, and peripheral blood NLRP3. RESULTS: No significant differences were observed in the general characteristics between the two patient groups (P > 0.05). However, the observation group exhibited higher levels of peripheral blood triglycerides, blood glucose, and TyG index compared to the control group (P < 0.05). Additionally, levels of NLRP3 and inflammatory factors IL-6, TNF-α, and CRP were elevated in the observation group compared to the control group (P < 0.05). Cardiac function impairment was more pronounced in the observation group (P < 0.05). Notably, TyG index and peripheral blood NLRP3 demonstrated higher risk ratios compared to other biomarkers (P < 0.05), indicating their significance in prognosis and outcomes. Elevated levels of NLRP3 and TyG index were associated with poorer recovery of cardiac function, increased rehospitalization rates, and higher mortality (P < 0.05). CONCLUSION: Elevated NLRP3 levels and an increased TyG index are strongly associated with impaired cardiac function and heightened risk of cardiovascular events. These findings suggest that these biomarkers may serve as crucial prognostic indicators following myocardial ischemia-reperfusion injury.


Assuntos
Glicemia , Traumatismo por Reperfusão Miocárdica , Proteína 3 que Contém Domínio de Pirina da Família NLR , Triglicerídeos , Humanos , Masculino , Feminino , Prognóstico , Proteína 3 que Contém Domínio de Pirina da Família NLR/sangue , Pessoa de Meia-Idade , Triglicerídeos/sangue , Traumatismo por Reperfusão Miocárdica/sangue , Glicemia/análise , Glicemia/metabolismo , Idoso , Biomarcadores/sangue
2.
BMC Cardiovasc Disord ; 24(1): 531, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39354361

RESUMO

BACKGROUND: Myocardial ischemia-reperfusion injury (MI/RI) is an unavoidable risk event for acute myocardial infarction, with ferroptosis showing close involvement. We investigated the mechanism of MI/RI inducing myocardial injury by inhibiting the ferroptosis-related SLC7A11/glutathione (GSH)/glutathione peroxidase 4 (GPX4) pathway and activating mitophagy. METHODS: A rat MI/RI model was established, with myocardial infarction area and injury assessed by TTC and H&E staining. Rat cardiomyocytes H9C2 were cultured in vitro, followed by hypoxia/reoxygenation (H/R) modeling and the ferroptosis inhibitor lipoxstatin-1 (Lip-1) treatment, or 3-Methyladenine or rapamycin treatment and overexpression plasmid (oe-SLC7A11) transfection during modeling. Cell viability and death were evaluated by CCK-8 and LDH assays. Mitochondrial morphology was observed by transmission electron microscopy. Mitochondrial membrane potential was detected by fluorescence dye JC-1. Levels of inflammatory factors, reactive oxygen species (ROS), Fe2+, malondialdehyde, lipid peroxidation, GPX4 enzyme activity, glutathione reductase, GSH and glutathione disulfide, and SLC7A11, GPX4, LC3II/I and p62 proteins were determined by ELISA kit, related indicator detection kits and Western blot. RESULTS: The ferroptosis-related SLC7A11/GSH/GPX4 pathway was repressed in MI/RI rat myocardial tissues, inducing myocardial injury. H/R affected GSH synthesis and inhibited GPX4 enzyme activity by down-regulating SLC7A11, thus promoting ferroptosis in cardiomyocytes, which was averted by Lip-1. SLC7A11 overexpression improved H/R-induced cardiomyocyte ferroptosis via the GSH/GPX4 pathway. H/R activated mitophagy in cardiomyocytes. Mitophagy inhibition reversed H/R-induced cellular ferroptosis. Mitophagy activation partially averted SLC7A11 overexpression-improved H/R-induced cardiomyocyte ferroptosis. H/R suppressed the ferroptosis-related SLC7A11/GSH/GPX4 pathway by inducing mitophagy, leading to cardiomyocyte injury. CONCLUSIONS: Increased ROS under H/R conditions triggered cardiomyocyte injury by inducing mitophagy to suppress the ferroptosis-related SLC7A11/GSH/GPX4 signaling pathway activation.


Assuntos
Sistema y+ de Transporte de Aminoácidos , Modelos Animais de Doenças , Ferroptose , Glutationa , Mitofagia , Traumatismo por Reperfusão Miocárdica , Miócitos Cardíacos , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Ratos Sprague-Dawley , Transdução de Sinais , Animais , Masculino , Ratos , Sistema y+ de Transporte de Aminoácidos/metabolismo , Sistema y+ de Transporte de Aminoácidos/genética , Linhagem Celular , Ferroptose/efeitos dos fármacos , Glutationa/metabolismo , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/patologia , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitofagia/efeitos dos fármacos , Infarto do Miocárdio/patologia , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/genética , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Miócitos Cardíacos/efeitos dos fármacos , Estresse Oxidativo , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/genética , Espécies Reativas de Oxigênio/metabolismo
3.
Mol Med Rep ; 30(6)2024 12.
Artigo em Inglês | MEDLINE | ID: mdl-39364741

RESUMO

The present study aimed to investigate the role of PI3K­mediated ferroptosis signaling induced by mild therapeutic hypothermia (MTH), which was defined as a temperature of 34˚C, in protecting against myocardial ischemia-reperfusion (I/R) injury (MIRI). To meet this aim, H9C2 cells underwent hypoxia­reperfusion (H/R) and/or MTH. The MTT assay was used to assess cell viability, cytotoxicity was measured using a lactate dehydrogenase cytotoxicity assay, and Annexin V­FITC/PI flow cytometric analysis was used to analyze early and late cell apoptosis. In addition, 84 healthy adult male Sprague­Dawley rats were randomly divided into seven groups (n=12), and underwent I/R and various treatments. Hemodynamics were monitored, and the levels of myocardial injury marker enzymes and oxidative stress markers in myocardial tissue were measured using ELISA. The expression levels of PI3K, AKT, transient receptor potential cation channel subfamily M member 7 (TRPM7), glutathione peroxidase 4 (GPX4) and acyl­CoA synthetase long chain family member 4 (ACSL4) in animals and cells were measured using western blot analysis. These experiments revealed that MTH could effectively reduce myocardial infarct size, improve hemodynamic performance following MIRI and suppress myocardial apoptosis, thereby contributing to the recovery from H/R injury. Mechanistically, MTH was revealed to be able to activate the PI3K/AKT signaling pathway in cells, upregulating GPX4, and downregulating the expression levels of TRPM7 and ACSL4. Treatment with 2­aminoethoxydiphenyl borate (an inhibitor of TRPM7) could further strengthen the myocardial protective effects of MTH, whereas treatment with erastin (promoter of ferroptosis) and wortmannin (inhibitor of PI3K) led to the effective elimination of the myocardial protective effects of MTH. Compared with in the I/R group, the PI3K/AKT activation level and the expression levels of GPX4 were both significantly increased, whereas the expression levels of TRPM7 and ACSL4 were significantly decreased in the I/R + MTH group. Taken together, the results of the present study indicated that MTH may activate the PI3K/AKT signaling pathway to inhibit TRPM7 and suppress ferroptosis induced by MIRI.


Assuntos
Ferroptose , Traumatismo por Reperfusão Miocárdica , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Ratos Sprague-Dawley , Transdução de Sinais , Canais de Cátion TRPM , Animais , Ferroptose/efeitos dos fármacos , Canais de Cátion TRPM/metabolismo , Canais de Cátion TRPM/antagonistas & inibidores , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Masculino , Ratos , Hipotermia Induzida/métodos , Proteínas Serina-Treonina Quinases/metabolismo , Linhagem Celular , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos
4.
Zhen Ci Yan Jiu ; 49(9): 902-908, 2024.
Artigo em Chinês | MEDLINE | ID: mdl-39401826

RESUMO

OBJECTIVES: To observe the effect of electroacupuncture(EA) on adenosine 5'-monophosphate-activated protein kinase (AMPK)/Kruppel-like factor 2 (KLF2) signaling pathway in ischemic myocardial tissues of rats, so as to explore the underlying mechanism of EA in attenuating myocardial ischemia-reperfusion injury (MIRI) through mediating angiogenesis. METHODS: Male SD rats were randomly divided into sham operation group, model group and EA group, with 10 rats in each group. The MIRI model was established by ligation of the anterior descending branch of the left coronary artery. Twenty-four hours after modeling, the rats in the EA group were given EA (2 Hz/100 Hz, 2 mA) at "Neiguan" (PC6) for 20 min each time, once a day for 5 consecutive days. Echocardiography was used to detect cardiac ejection fraction (EF) to evaluate cardiac function. HE staining was used to observe the morphological changes in rat myocardial tissue. Immunohistochemistry was used to detect the density of neovascularization in rat ischemic myocardium. Western blot and ELISA were used to detect the phosphorylated(p)-AMPK, AMPK, KLF2, vascular endothelial growth factor (VEGF) protein expression levels, and VEGF receptor 2 (VEGFR2) content in rat ischemic myocardial tissue, respectively. RESULTS: After modeling, compared with the sham operation group, rats in the model group had decreased EF(P<0.01), significant myocardial fiber damage with inflammatory cell infiltration, increased neovascular density (P<0.05), increased p-AMPK, AMPK, VEGF protein expression levels and VEGFR2 content in myocardial ischemic tissues(P<0.05, P<0.01), and decreased protein expression level of KLF2 (P<0.05). After EA intervention, compared with the model group, rats in the EA group had elevated EF(P<0.01), significantly reduced myocardial fiber damage, reduced inflammatory cell infiltration, increased neovascular density(P<0.01), and elevated p-AMPK, AMPK, KLF2, and VEGF protein expression levels and VEGFR2 content in the myocardial ischemic tissue (P<0.01). CONCLUSIONS: EA may promote angiogenesis, attenuate myocardial injury, and achieve cardioprotective effects in MIRI rats by regulating the expression of AMPK/KLF2 signaling pathway in myocardial tissues.


Assuntos
Proteínas Quinases Ativadas por AMP , Eletroacupuntura , Fatores de Transcrição Kruppel-Like , Traumatismo por Reperfusão Miocárdica , Ratos Sprague-Dawley , Transdução de Sinais , Animais , Masculino , Ratos , Fatores de Transcrição Kruppel-Like/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Humanos , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/terapia , Neovascularização Fisiológica , Pontos de Acupuntura , Miocárdio/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Angiogênese
5.
ACS Nano ; 18(41): 28154-28169, 2024 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-39373010

RESUMO

Myocardial ischemia-reperfusion (IR) injury is a severe rhythmic disease with a high prevalence in the early morning. IR injury has a significant circadian rhythm in reactive oxygen species (ROS) and inflammation levels. The development of rhythmic drugs has become a priority in myocardial IR injury. In this study, resveratrol (RES) and proanthocyanidins (OPC) were utilized to design nanoparticles (NPs), with hyaluronic acid (HA) as the core, grafted with MMP-targeting peptides to improve delivery to injured myocardial regions (HA-RES-OPC-MMP NPs). NPs significantly scavenged ROS, attenuated inflammation, and activated the rhythm gene. Notably, the difference in therapeutic effects on myocardial IR injury in mice at Zeitgeber time (ZT)1 and ZT13 confirms that NPs are rhythm-dependent drugs. At ZT13, echocardiographic and MRI confirm that IR injury in mice was not as severe as at ZT1, yet NPs were also less effective in treatment. Further, Per1/2 knockout mice confirmed the rhythm-dependent treatment of myocardial IR injury by NPs. Molecular studies have shown that rhythmic characteristics of inflammation and Sirt1 transcript levels are the main reasons for the different rhythmic therapeutic effects of NPs. Circadian rhythm-dependent treatment of HA-RES-OPC-MMP NPs has excellent potential for more precise treatment of myocardial IR injury in the future.


Assuntos
Ritmo Circadiano , Traumatismo por Reperfusão Miocárdica , Nanopartículas , Resveratrol , Animais , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Nanopartículas/química , Camundongos , Resveratrol/farmacologia , Resveratrol/química , Ritmo Circadiano/efeitos dos fármacos , Camundongos Knockout , Masculino , Proantocianidinas/química , Proantocianidinas/farmacologia , Ácido Hialurônico/química , Espécies Reativas de Oxigênio/metabolismo , Camundongos Endogâmicos C57BL , Sirtuína 1/metabolismo , Proteínas Circadianas Period/metabolismo , Proteínas Circadianas Period/genética , Polifenóis/química , Polifenóis/farmacologia
6.
Cardiovasc Toxicol ; 24(11): 1236-1252, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39264521

RESUMO

Uremic cardiomyopathy (UC) represents a complex syndrome characterized by different cardiac complications, including systolic and diastolic dysfunction, left ventricular hypertrophy, and diffuse fibrosis, potentially culminating in myocardial infarction (MI). Revascularization procedures are often necessary for MI management and can induce ischemia reperfusion injury (IR). Despite this clinical relevance, the role of fine particulate matter (PM2.5) in UC pathology and the underlying subcellular mechanisms governing this pathology remains poorly understood. Hence, we investigate the impact of PM2.5 exposure on UC susceptibility to IR injury. Using a rat model of adenine-induced chronic kidney disease (CKD), the animals were exposed to PM2.5 at 250 µg/m3 for 3 h daily over 21 days. Subsequently, hearts were isolated and subjected to 30 min of ischemia followed by 60 min of reperfusion to induce IR injury. UC hearts exposed to PM2.5 followed by IR induction (Adenine + PM_IR) exhibited significantly impaired cardiac function and increased cardiac injury (increased infarct size and apoptosis). Analysis at the subcellular level revealed reduced mitochondrial copy number, impaired mitochondrial bioenergetics, decreased expression of PGC1-α (a key regulator of mitochondrial biogenesis), and compromised mitochondrial quality control mechanisms. Additionally, increased mitochondrial oxidative stress and perturbation of the PI3K/AKT/AMPK signaling axis were evident. Our findings therefore collectively indicate that UC myocardium when exposed to PM2.5 is more vulnerable to IR-induced injury, primarily due to severe mitochondrial impairment.


Assuntos
Apoptose , Cardiomiopatias , Modelos Animais de Doenças , Metabolismo Energético , Mitocôndrias Cardíacas , Traumatismo por Reperfusão Miocárdica , Material Particulado , Transdução de Sinais , Uremia , Animais , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Traumatismo por Reperfusão Miocárdica/induzido quimicamente , Material Particulado/toxicidade , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/patologia , Mitocôndrias Cardíacas/efeitos dos fármacos , Masculino , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/fisiopatologia , Apoptose/efeitos dos fármacos , Uremia/metabolismo , Uremia/induzido quimicamente , Uremia/patologia , Uremia/complicações , Metabolismo Energético/efeitos dos fármacos , Miócitos Cardíacos/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Insuficiência Renal Crônica/patologia , Insuficiência Renal Crônica/induzido quimicamente , Insuficiência Renal Crônica/metabolismo , Poluentes Atmosféricos/toxicidade , Ratos Sprague-Dawley , Proteínas Proto-Oncogênicas c-akt/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Adenina/toxicidade , Adenina/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Função Ventricular Esquerda/efeitos dos fármacos , Infarto do Miocárdio/patologia , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/induzido quimicamente , Infarto do Miocárdio/fisiopatologia , Fosfatidilinositol 3-Quinase/metabolismo
7.
Int Immunopharmacol ; 142(Pt A): 113097, 2024 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-39260311

RESUMO

Hydroxyl Safflower Yellow A (HSYA) is the primary bioactive compound derived from Safflower, which has been scientifically proven to possess anti-inflammatory, anti-apoptotic, and ameliorative properties against mitochondrial damage during acute myocardial ischemia-reperfusion injury (MIRI); however, its effects during the recovery stage remain unknown. Angiogenesis plays a crucial role in the rehabilitation process. AIM OF THE STUDY: The objective of this study was to investigate the long-term angiogenic effect of HSYA and its contribution to recovery after myocardial ischemia, as well as explore its underlying mechanism using non-targeted metabolomics and network pharmacology. MATERIALS AND METHODS: The MIRI model in rat was established by ligating the left anterior descending branch of the coronary artery. The effect of HSYA was assessed based on myocardial infarction volume and histopathology. Immunofluorescence staining was employed to evaluate angiogenesis, while ELISA was used to detect markers of myocardial injury. Additionally, a rat myocardial microvascular endothelial cell (CMECs) injury model was established using oxygen-glucose deprivation/reoxygenation (OGD/R), followed by scratch assays, migration assays, and tube formation experiments to assess angiogenesis. Western blot analysis was conducted to validate the underlying mechanism. RESULTS: Our findings provide compelling evidence for the therapeutic efficacy of HSYA in reducing myocardial infarction size, facilitating cardiac functional recovery, and reversing pathological alterations within the heart. Furthermore, we elucidate that HSYA exerts its effects on promoting migration and generation of myocardial microvascular endothelial cells through activation of the HIF-1α-VEGFA-Notch1 signaling pathway. CONCLUSION: These results underscore how HSYA enhances cardiac function via angiogenesis promotion and activation of the aforementioned signaling cascade.


Assuntos
Chalcona , Subunidade alfa do Fator 1 Induzível por Hipóxia , Traumatismo por Reperfusão Miocárdica , Neovascularização Fisiológica , Quinonas , Ratos Sprague-Dawley , Receptor Notch1 , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular , Animais , Chalcona/análogos & derivados , Chalcona/farmacologia , Chalcona/uso terapêutico , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Quinonas/farmacologia , Quinonas/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Receptor Notch1/metabolismo , Ratos , Neovascularização Fisiológica/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Modelos Animais de Doenças , Células Cultivadas , Carthamus tinctorius , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia , Infarto do Miocárdio/metabolismo , Angiogênese
8.
J Control Release ; 375: 552-573, 2024 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-39276800

RESUMO

Organic nanocrystals, particularly those composed of conjugated molecules, hold immense potential for various applications. However, their practical utility is often hindered by the challenge of achieving stable aqueous dispersions, which are essential for biological compatibility and effective delivery. This study introduces a novel and versatile strategy for preparing stable aqueous organic nanocrystals using a modified reprecipitation method. We demonstrate the broad applicability of this approach by successfully preparing a diverse library of nanocrystals from 27 conjugated molecules. Our findings reveal a charge-balanced aggregation mechanism for nanocrystal formation, highlighting the crucial role of surface charge in controlling particle size and stability. Based on this mechanism, we establish a comprehensive molecular combination strategy that directly links molecular properties to colloidal behaviour, enabling the straightforward prediction and preparation of stable aqueous dispersions without the need for excipients. This strategy provides a practical workflow for tailoring the functionality of these nanocrystals for a wide range of applications. To illustrate their therapeutic potential, we demonstrate the enhanced efficacy of these nanocrystals in treating acute ulcerative colitis, myocardial ischemia/reperfusion injury, and cancer in mouse models. This work paves the way for developing next-generation nanomaterials with tailored functionalities for diverse biomedical applications.

9.
Lab Anim Res ; 40(1): 32, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39237965

RESUMO

Ischemic heart disease is the most prevalent cause of death worldwide affecting both the gender of all age groups. The high mortality rate is due to damage of myocardial tissue that emanates at the time of myocardial ischemia and re-oxygenation, thus averting reperfusion injury is recognized as a potential way to reduce acute cardiac injury and subsequent mortality. Flavonoids are polyphenol derivatives of plant origin and empirical shreds of evidence substantiate their numerous activities such as antioxidant, anti-inflammatory, anti-apoptotic, and anti-thrombotic activity, leading to their role in cardio protection. Recent investigations have unveiled the capacity of flavonoids to impede pivotal regulatory enzymes, signaling molecules, and transcription factors that orchestrate the mediators participating in the inflammatory cascade. The present comprehensive review, dwells on the preclinical studies on the effectiveness of flavonoids from the year 2007 to 2023, for the prevention and therapeutics for myocardial ischemia-reperfusion injury.

10.
Mol Med Rep ; 30(5)2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39301623

RESUMO

Following acute myocardial infarction, the recovery of blood flow leads to myocardial ischemia­reperfusion (MI/R) injury, which is primarily characterized by the activation of inflammatory signals, microvascular obstruction, increased oxidative stress and excessive Ca2+ overload. It has also been demonstrated that platelets can exacerbate MI/R injury by releasing reactive oxygen species, inflammatory factors and chemokines, while also obstructing microvessels through thrombus formation. As a bioactive molecule with proinflammatory and chemotactic properties, lipocalin 2 (LCN2) exhibits a positive correlation with obesity, hyperglycemia, hypertriglyceridemia and insulin resistance index, which are all significant risk factors for ischemic cardiomyopathy. Notably, the potential role of LCN2 in promoting atherosclerosis may be related to its influence on the function of macrophages, smooth muscle cells and endothelial cells, but its effect on platelet function has not yet been reported. In the present study, the effect of a high­fat diet (HFD) on LCN2 expression was determined by detecting LCN2 expression levels in the liver and serum samples of mice through reverse transcription­quantitative PCR and enzyme linked immunosorbent assay, respectively. The effect of LCN2 on platelet function was evaluated by examining whether LCN2 affected platelet activation, aggregation, adhesion, clot retraction and P­selectin expression. To determine whether LCN2 aggravated MI/R injury in HFD­fed mice by affecting platelet and inflammatory cell recruitment, wild­type and LCN2 knockout mice fed a HFD were subjected to MI/R injury, then hearts were collected for hematoxylin and eosin staining and 2,3,5­triphenyltetrazolium chloride staining, and immunohistochemistry was employed to detect the expression of CD42b, Ly6G, CD3 and B220. Based on observing the upregulation of LCN2 expression in mice fed a HFD, the present study further confirmed that LCN2 could accelerate platelet activation, aggregation and adhesion. Moreover, in vivo studies validated that knockout of LCN2 not only mitigated MI/R injury, but also inhibited the recruitment of platelets and inflammatory cells in myocardial tissue following ischemia­reperfusion. In conclusion, the current findings suggested that the effect of HFD­induced LCN2 on aggravating MI/R injury may totally or partially dependent on its promotion of platelet function.


Assuntos
Dieta Hiperlipídica , Lipocalina-2 , Traumatismo por Reperfusão Miocárdica , Ativação Plaquetária , Animais , Dieta Hiperlipídica/efeitos adversos , Lipocalina-2/metabolismo , Lipocalina-2/genética , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/etiologia , Traumatismo por Reperfusão Miocárdica/genética , Camundongos , Masculino , Plaquetas/metabolismo , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Camundongos Knockout
11.
J Biochem Mol Toxicol ; 38(9): e23816, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39185902

RESUMO

Reperfusion strategies, the standard therapy for acute myocardial infarction (AMI), may result in ischemia/reperfusion (I/R) damage. Suppressor of cytokine signaling1 (SOCS1) exerts a cardioprotective function in myocardial I/R damage. Here, we investigated epigenetic modulators that deregulate SOCS1 in cardiomyocytes under hypoxia/reoxygenation (H/R) conditions. Human AC16 cardiomyocytes were exposed to H/R conditions to generate a cell model of myocardial I/R damage. Expression of mRNA and protein was detected by quantitative PCR and western blot analysis, respectively. Cell migratory and invasive abilities were evaluated by transwell assay. Cell apoptosis and M2 macrophage polarization were assessed by flow cytometry. TNF-α, IL-1ß, and IL-6 levels were examined by ELISA. The interaction of KLF4 with SOCS1 was verified by chromatin immunoprecipitation and luciferase assays. SOCS1 and transcription factor KLF4 protein levels were underexpressed by 75% and 57%, respectively, in H/R-exposed AC16 cardiomyocytes versus control cells. Under H/R conditions, forced SOCS1 expression (2.7 times) induced cell migration (2.2 times) and invasion (1.9 times) and hindered cell apoptosis (by 45%) of AC16 cardiomyocytes as well as enhanced M2 macrophage polarization (4.6 times). Mechanistically, KLF4 upregulation promoted SOCS1 transcription (2.6 times) and expression (2.6 times) by binding to the SOCS1 promoter. Decrease of SOCS1 (by 51%) reversed the effects of KLF4 upregulation on cardiomyocyte migration, invasion and apoptosis, and M2 macrophage polarization under H/R conditions. Additionally, SOCS1 and KLF4 were underexpressed by 56% and 63%, respectively, in AMI serum. Our study indicates that KLF4-induced upregulation of SOCS1 can attenuate H/R-triggered apoptosis of AC16 cardiomyocytes and enhance M2 macrophage polarization.


Assuntos
Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like , Macrófagos , Traumatismo por Reperfusão Miocárdica , Miócitos Cardíacos , Proteína 1 Supressora da Sinalização de Citocina , Regulação para Cima , Proteína 1 Supressora da Sinalização de Citocina/metabolismo , Proteína 1 Supressora da Sinalização de Citocina/genética , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fatores de Transcrição Kruppel-Like/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Humanos , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Macrófagos/metabolismo , Linhagem Celular , Apoptose
12.
J Control Release ; 374: 639-652, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39208931

RESUMO

Delivering therapeutic agents efficiently to inflamed regions remains an intractable challenge following myocardial ischemia-reperfusion injury (MI/RI) due to the transient nature of the enhanced permeability and retention effect, which disappears after 24 h. Leveraging the inflammation-homing and plasticity properties of circulating monocytes (MN) as hitchhiking carriers and further inducing their polarization into anti-inflammatory phenotype macrophages upon reaching the inflamed sites is beneficial for MI/RI therapy. Herein, DSS/PB@BSP nanoparticles capable of clearing reactive oxygen species and inhibiting inflammation were developed by employing hollow Prussian blue nanoparticles (PB) as carriers to encapsulate betamethasone sodium phosphate (BSP) and further modified with dextran sulfate sodium (DSS), a targeting ligand for the scavenger receptor on MN. This formulation was internalized into MN as living cell drug depots, reprogramming them into anti-inflammation type macrophages to inhibit inflammation. In vitro assessments revealed the successful construction of the nanoparticle. In a murine MI/RI model, circulating MN laden with these nanoparticles significantly enhanced drug delivery and accumulation at the cardiac injury site, exhibiting favorable therapeutic ability and promoting M2-biased differentiation. Our study provides an effective approach with minimally invasion and biosecurity that makes this nanoplatform as a promising candidate for immunotherapy and clinical translation in the treatment of MI/RI.


Assuntos
Betametasona , Macrófagos , Camundongos Endogâmicos C57BL , Monócitos , Traumatismo por Reperfusão Miocárdica , Nanopartículas , Animais , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Macrófagos/efeitos dos fármacos , Nanopartículas/química , Monócitos/efeitos dos fármacos , Camundongos , Masculino , Betametasona/administração & dosagem , Betametasona/análogos & derivados , Ferrocianetos/química , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/farmacologia , Portadores de Fármacos/química , Reprogramação Celular/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
13.
Biomed Pharmacother ; 179: 117345, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39208667

RESUMO

This study investigates the protective effects of propofol on the myocardium by inhibiting the expression of SLC16A13 through in vivo animal experiments, while also exploring its mechanism in ferroptosis to provide new strategies for preventing perioperative myocardial ischemia-reperfusion injury. We randomly divided 30 rats into three groups (n=10 each): sham surgery group, ischemia-reperfusion (I/R) group, and propofol pretreatment group. The results showed that compared with the sham surgery group, the I/R group had a significant decrease in cardiac function and an increase in infarct size. Propofol pretreatment effectively alleviated the damage caused by ischemia-reperfusion (I/R). In the next phase of the study, we administered the PPARα agonist GW7647 to artificially increase the expression of SLC16A13. Fifty rats were randomly divided into five groups (n=10 each), with the GW7647 pretreatment group and propofol+GW7647 pretreatment group added based on the previous three groups. Afterwards, we validated the in vivo results using H9C2 and further explored the mechanism by which propofol inhibits ferroptosis. The study found that L-lactic acid in myocardial tissue of the GW7647 group was further increased compared to the I/R group, and the degree of ferroptosis was aggravated. In addition, upregulation of SLC16A13 significantly inhibited the phosphorylation of AMPK, weakened the protective mechanism of AMPK, and exacerbated cardiac damage. However, propofol pretreatment can effectively inhibit the expression of SLC16A13, maintain normal myocardial cell morphology, and protect cardiac function. These results indicate that propofol inhibits the expression of SLC16A13, alleviates myocardial cell ferroptosis via the AMPK/GPX4 pathway, and reverses damage caused by myocardial ischemia-reperfusion.


Assuntos
Proteínas Quinases Ativadas por AMP , Ferroptose , Transportadores de Ácidos Monocarboxílicos , Traumatismo por Reperfusão Miocárdica , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Propofol , Ratos Sprague-Dawley , Animais , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Ferroptose/efeitos dos fármacos , Transportadores de Ácidos Monocarboxílicos/metabolismo , Propofol/farmacologia , Ratos , Masculino , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Transdução de Sinais/efeitos dos fármacos , Linhagem Celular , Miocárdio/patologia , Miocárdio/metabolismo , Proteínas Musculares
14.
Int Immunopharmacol ; 140: 112921, 2024 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-39133953

RESUMO

Effective treatment of myocardial ischemia-reperfusion (MIR) injury remains an unmet clinical need. Cardiomyocyte apoptosis is common at this stage and poses a significant risk. Corylin, a flavonoid compound extracted from Psoralea corylifolia L., has been shown to have anti-inflammatory, anticancer, and antiatherosclerotic properties. However, whether and how corylin affects MIR injury remain unclear. In this study, we explored the mechanism of corylin as a potent therapeutic agent for MI/R injury, using a left anterior descending (LAD) coronary artery ligation and oxygen-glucose deprivation and reperfusion (OGD/R) model in vivo and in vitro. TUNEL, Annexin-V/PI double staining,Ki67 immunohistochemistry, western blot analysis, and immunofluorescence were used to validate cell apoptosis level and Raf-1/ASK1 complex activity. The interaction between corylin and Raf-1/ASK1 complex was detected using molecular docking, corylin-Raf-1 binding assays, and coimmunoprecipitation (Co-IP). Moreover, TTC staining, echocardiography, HE staining, Masson trichrome staining and serological testing were performed to assess the cardioprotective effects of corylin in vivo. These findings showed that corylin reduces MIR injury-induced cardiomyocyte apoptosis and improves cardiac function. Mechanistically, corylin can interact with Raf-1 and promote the formation of the Raf-1/ASK1 complex, thus inhibiting cardiomyocyte apoptosis. In conclusion, our results demonstrate that corylin ameliorated cardiac dysfunction after MIR injury by reducing myocardial apoptosis.


Assuntos
Apoptose , MAP Quinase Quinase Quinase 5 , Traumatismo por Reperfusão Miocárdica , Miócitos Cardíacos , Proteínas Proto-Oncogênicas c-raf , Apoptose/efeitos dos fármacos , Animais , Proteínas Proto-Oncogênicas c-raf/metabolismo , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , MAP Quinase Quinase Quinase 5/metabolismo , Flavonoides/farmacologia , Flavonoides/uso terapêutico , Camundongos Endogâmicos C57BL , Camundongos , Humanos , Psoralea/química , Modelos Animais de Doenças
15.
Cell Signal ; 123: 111370, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39216681

RESUMO

Acute myocardial infarction (AMI) is the leading cause of death worldwide, and reperfusion therapy is a critical therapeutic approach to reduce myocardial ischemic injury and minimize infarct size. However, ischemia/reperfusion (I/R) itself also causes myocardial injury, and inflammation is an essential mechanism by which it leads to myocardial injury, with macrophages as crucial immune cells in this process. Macrophages are innate immune cells that maintain tissue homeostasis, host defence during pathogen infection, and repair during tissue injury. During the acute phase of I/R, M1-type macrophages generate a pro-inflammatory milieu, clear necrotic myocardial tissue, and further recruit mononuclear (CCR2+) macrophages. Over time, the reparative (M2 type) macrophages gradually became dominant. In recent years, metabolic studies have shown a clear correlation between the metabolic profile of macrophages and their phenotype and function. M1-type macrophages are mainly characterized by glycolytic energy supply, and their tricarboxylic acid (TCA) cycle and mitochondrial oxidative phosphorylation (OXPHOS) processes are impaired. In contrast, M2 macrophages rely primarily on OXPHOS for energy. Changing the metabolic profile of macrophages can alter the macrophage phenotype. Altered energy pathways are also present in macrophages during I/R, and intervention in this process contributes to earlier and greater M2 macrophage infiltration, which may be a potential target for the treatment of myocardial I/R injury. Therefore, this paper mainly reviews the characteristics of macrophage energy metabolism alteration and phenotypic transition during I/R and its mechanism of mediating myocardial injury to provide a basis for further research in this field.


Assuntos
Macrófagos , Traumatismo por Reperfusão Miocárdica , Humanos , Macrófagos/metabolismo , Animais , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Fosforilação Oxidativa , Miocárdio/metabolismo , Miocárdio/patologia , Metabolismo Energético , Reprogramação Metabólica
16.
Biomed Pharmacother ; 178: 117268, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39116780

RESUMO

Gastric precancerous lesion (GPL) is a crucial stage in the development of gastric cancer, characterized by incomplete intestinal epithelial chemotaxis and heterogeneous hyperplasia with high malignant potential. Early intervention in GPL is vital for preventing gastric cancer. Additionally, there are shared risk factors and pathogenesis between tumors and coronary heart disease (CHD), with an increasing number of tumor patients GPL complicated with CHD due to improved survival rates. Reperfusion therapy in CHD can result in myocardial ischemia-reperfusion injury (MIRI). Traditional Chinese medicine (TCM) has demonstrated unique advantages in treating GPL and MIRI by promoting blood circulation and removing blood stasis. Panax ginseng total saponin (PNS), a component of TCM known for its blood circulation benefits, has shown positive effects in inhibiting tumor growth and improving myocardial ischemia. This study utilized a GPL-MIRI mouse model to investigate the effects of PNS in treatment. Results indicated that PNS significantly improved typical GPL lesions in mice, such as incomplete intestinal epithelialization and heteroplasia, and also reduced myocardial infarction. At the molecular level, PNS exhibited a bidirectional regulatory role in the GPL-MIRI model. It enhanced the autophagic process in gastric mucosal cells by inhibiting the PI3K/Akt/mTOR signaling pathway, while suppressed excessive autophagy in cardiomyocytes. These findings offer new insights and treatment strategies for managing GPL and MIRI using the TCM compound PNS.


Assuntos
Autofagia , Traumatismo por Reperfusão Miocárdica , Panax notoginseng , Saponinas , Transdução de Sinais , Neoplasias Gástricas , Animais , Masculino , Camundongos , Autofagia/efeitos dos fármacos , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Panax notoginseng/química , Fosfatidilinositol 3-Quinases/metabolismo , Lesões Pré-Cancerosas/tratamento farmacológico , Lesões Pré-Cancerosas/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Saponinas/farmacologia , Saponinas/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/patologia , Neoplasias Gástricas/metabolismo , Serina-Treonina Quinases TOR/metabolismo
17.
Exp Gerontol ; 195: 112540, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39122228

RESUMO

BACKGROUND: Myocardial ischemia-reperfusion (MI/R) can lead to structural and functional abnormalities in the hippocampal neurons of the brain. High-mobility group box-l (HMGB1) is implicated in the activation of immune cells and the stimulation of inflammatory responses. However, the specific role of HMGB1 in cognitive impairment induced by MI/R in elderly rats has yet to be elucidated. METHODS: Elderly rats underwent surgical procedures to induce MI/R. To evaluate the learning and memory abilities of these rats, a water maze test and a new-object recognition test were administered. Nissl staining was utilised to examine hippocampal neuron damage. Enzyme-linked immunosorbent assay, western blotting, and real-time quantitative polymerase chain reaction (RT-qPCR) analyses were conducted to measure the expression levels of HMGB1, inflammatory cytokines, and molecular pathways. RESULTS: The study found that MI/R induced cognitive impairment in elderly rats. There was an observed increase in serum HMGB1 levels, along with elevated concentrations of pro-inflammatory cytokines in the plasma and hippocampus, accompanied by a decrease in anti-inflammatory cytokines. Moreover, substantial damage was evident in the hippocampal neurons of rats exposed to MI/R. In the brains of these rats, there was an increased expression of HMGB1, the receptor for advanced glycation end products (RAGE), toll-like receptor 4 (TLR4), phosphorylated p65, interleukin-1ß (IL-1ß), IL-6, IL-23, tumour necrosis factor-α (TNF-α), caspase-3, and Bax. In contrast, the expression of B-cell lymphoma 2 was decreased. The RT-qPCR analyses indicated elevated levels of HMGB1, RAGE, TLR4, IL-1ß, IL-6, IL-23, TNF-α, caspase-3, and Bax mRNA. CONCLUSION: The increased concentration of serum and hippocampal inflammatory factors in the brains of elderly rats subjected to MI/R suggests that cognitive impairment may be induced through the activation of the HMGB1/TLR4/NF-κB signalling pathway.


Assuntos
Disfunção Cognitiva , Citocinas , Proteína HMGB1 , Hipocampo , Traumatismo por Reperfusão Miocárdica , Ratos Sprague-Dawley , Animais , Proteína HMGB1/metabolismo , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/patologia , Masculino , Hipocampo/metabolismo , Hipocampo/patologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Ratos , Citocinas/metabolismo , Receptor 4 Toll-Like/metabolismo , Aprendizagem em Labirinto , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Modelos Animais de Doenças , Neurônios/metabolismo , Neurônios/patologia , NF-kappa B/metabolismo , Envelhecimento , Transdução de Sinais
18.
Colloids Surf B Biointerfaces ; 243: 114159, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39137530

RESUMO

After myocardial ischemia/reperfusion injury (MI/RI), endothelial cell injury causes impaired angiogenesis and obstruction of microcirculation, resulting in an inflammatory outburst that exacerbates the damage. Therefore, synergistic blood vessel repair and inflammation inhibition are effective therapeutic strategies. In this study, we developed a platelet membrane (PM)-encapsulated baicalin nanocrystalline (BA NC) nanoplatform with a high drug load, BA NC@PM, which co-target to endothelial cells and macrophages through the transmembrane proteins of the PM to promote angiogenesis and achieve anti-inflammatory effects. In vitro cell scratch assays and transwell assay manifested that BA NC@PM could promote endothelial cell migration, as well as increase mRNA expression of CD31 and VEGF in the heart after treatment of MI/RI mice, suggesting its favorable vascular repair function. In addition, the preparation significantly reduced the expression of pro-inflammatory factors and increased the expression of anti-inflammatory factors in plasma, promoting the polarization of macrophages. Our study highlights a strategy for enhancing the treatment of MI/RI by promoting angiogenesis and regulating macrophage polarization via the biomimetic BA NC@PM nanoplatform.


Assuntos
Inflamação , Traumatismo por Reperfusão Miocárdica , Nanopartículas , Animais , Camundongos , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Inflamação/tratamento farmacológico , Inflamação/patologia , Inflamação/metabolismo , Nanopartículas/química , Flavonoides/farmacologia , Flavonoides/química , Materiais Biomiméticos/farmacologia , Materiais Biomiméticos/química , Humanos , Camundongos Endogâmicos C57BL , Masculino , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Movimento Celular/efeitos dos fármacos , Células RAW 264.7 , Tamanho da Partícula , Angiogênese
19.
Acta Biomater ; 2024 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-39122136

RESUMO

Sympathetic hyperactivation and inflammatory responses are the main causes of myocardial ischemia‒reperfusion (I/R) injury and myocardial I/R-related ventricular arrhythmias (VAs). Previous studies have demonstrated that light-emitting diodes (LEDs) could modulate post-I/R neuroinflammation, thus providing protection against myocardial I/R injury. Nevertheless, further applications of LEDs are constrained due to the low penetration depth (<1 cm) and potential phototoxicity. Low-intensity focused ultrasound (LIFU), an emerging noninvasive neuromodulation strategy with deeper penetration depth (∼10 cm), has been confirmed to modulate sympathetic nerve activity and inflammatory responses. Sonodynamic therapy (SDT), which combines LIFU with sonosensitizers, confers additional advantages, including superior therapeutic efficacy, precise localization of neuronal modulation and negligible side effects. Herein, LIFU and SDT were introduced to modulate post-myocardial I/R neuroinflammation to protect against myocardial I/R injury. The results indicated that LIFU and SDT inhibited sympathetic neural activity, suppressed the activation of astrocytes and microglia, and promoted microglial polarization towards the M2 phenotype, thereby attenuating myocardial I/R injury and preventing I/R-related malignant VAs. These insights suggest that LIFU and SDT inspire a noninvasive and efficient neuroinflammatory modulation strategy with great clinical translation potential thus benefiting more patients with myocardial I/R in the future. STATEMENT OF SIGNIFICANCE: Myocardial ischemia-reperfusion (I/R) may cause I/R injury and I/R-induced ventricular arrhythmias. Sympathetic hyperactivation and inflammatory response play an adverse effect in myocardial I/R injury. Previous studies have shown that light emitting diode (LED) can regulate I/R-induced neuroinflammation, thus playing a myocardial protective role. However, due to the low penetration depth and potential phototoxicity of LED, it is difficult to achieve clinical translation. Herein, we introduced sonodynamic modulation of neuroinflammation to protect against myocardial I/R injury, based on mitochondria-targeted nanosonosensitizers (CCNU980 NPs). We demonstrated that sonodynamic modulation could promote microglial autophagy, thereby preventing myocardial I/R injury and I/R-induced ventricular arrhythmias. This is the first example of sonodynamic modulation of myocardial I/R-induced neuroinflammation, providing a novel strategy for clinical translation.

20.
Imeta ; 3(4): e220, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39135700

RESUMO

Emerging evidence has demonstrated the profound impact of the gut microbiome on cardiovascular diseases through the production of diverse metabolites. Using an animal model of myocardial ischemia-reperfusion (I/R) injury, we found that the prophylactic administration of a well-known probiotic, Bifidobacterium infantis (B. infantis), exhibited cardioprotective effects in terms of preserving cardiac contractile function and preventing adverse cardiac remodeling following I/R and that these cardioprotective effects were recapitulated by its metabolite inosine. Transcriptomic analysis further revealed that inosine mitigated I/R-induced cardiac inflammation and cell death. Mechanistic investigations elucidated that inosine suppressed the production of pro-inflammatory cytokines and reduced the numbers of dendritic cells and natural killer cells, achieved through the activation of the adenosine A2A receptor (A2AR) that when inhibited abrogated the cardioprotective effects of inosine. Additionally, in vitro studies using C2C12 myoblasts revealed that inosine attenuated cell death by serving as an alternative carbon source for adenosine triphosphate (ATP) generation through the purine salvage pathway when subjected to oxygen-glucose deprivation/reoxygenation that simulated myocardial I/R injury. Likewise, inosine reversed the I/R-induced decrease in ATP levels in mouse hearts. Taken together, our findings indicate that B. infantis or its metabolite inosine exerts cardioprotective effects against I/R by suppressing cardiac inflammation and attenuating cardiac cell death, suggesting prophylactic therapeutic options for acute ischemic cardiac injury.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA