Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Biomed Pharmacother ; 165: 115247, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37516018

ABSTRACT

Mitochondrial dysfunction, especially in terms of mitochondrial dynamics, has been reported to be closely associated with neuronal outcomes and neurological impairment in cerebral ischemia/hypoxia injury. Dynamin-related protein 1 (Drp1) is a cytoplasmic GTPase that mediates mitochondrial fission and participates in neuronal cell death, calcium signaling, and oxidative stress. The neuroprotective role of Drp1 inhibition has been confirmed in several central nervous system disease models, demonstrating that targeting Drp1 may shed light on novel approaches for the treatment of cerebral ischemia/hypoxia injury. In this review, we aimed to highlight the roles of Drp1 in programmed cell death, oxidative stress, mitophagy, and mitochondrial function to provide a better understanding of mitochondrial disturbances in cerebral ischemia/hypoxia injury, and we also summarize the advances in novel chemical compounds targeting Drp1 to provide new insights into potential therapies for cerebral ischemia/hypoxia injury.


Subject(s)
Brain Ischemia , Neurons , Humans , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Dynamins/metabolism , Hypoxia/metabolism , Mitochondria/metabolism , Neurons/metabolism , Animals
2.
J Physiol Biochem ; 79(2): 327-339, 2023 May.
Article in English | MEDLINE | ID: mdl-36786974

ABSTRACT

Flap expansion has become an important method widely used in wound repair and organ reconstruction. However, distal skin flap ischemic necrosis remains a problematic complication. In this study, integrative bioinformatics analyses indicated the upregulation of C-C motif chemokine ligand 2 (CCL2) and C-C motif chemokine receptor 2 (CCR2) in reperfusion-exposed skin flap tissues. In adipose-derived stem cells (ADSCs, CD90-positive, CD29-positive, CD34-negative, and CD106-negative) exposed to hypoxia, HIF-1α and CCL2 levels were significantly elevated. Conditioned medium (CM) from hypoxia-stimulated ADSCs promoted HDMEC proliferation, migration, and tube formation, partially inhibited by sh-CCL2-induced CCL2 knockdown or neutralized antibody-induced CCL2 depletion in ADSCs. Consistently, CCL2, CCR2, TNF-α, TLR2, and TLR4 protein levels in HDMECs were significantly increased by hypoxia-treated ADSCs CM, and partially decreased by sh-CCL2-induced CCL2 knockdown or neutralizing antibody-induced CCL2 knockdown in ADSCs. In the flap expansion model, ADSCs transplantation significantly improved flap survival and angiogenesis by endothelial cells in flap tissues, whereas CCL2 knockdown in ADSCs partially eliminated the improvement by ADSCs transplantation; overexpression of CCL2 in ADSCs further promoted the effects of ADSCs transplantation on skin flap. In conclusion, the CCL2/CCR2 axis in ADSCs could be induced by hypoxia, promoting HDMEC proliferation, migration, and tube formation and improving flap survival and angiogenesis in flap tissues.


Subject(s)
Adipose Tissue , Endothelial Cells , Humans , Endothelial Cells/metabolism , Adipose Tissue/metabolism , Ligands , Hypoxia/metabolism , Stem Cells/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Receptors, CCR2/genetics , Receptors, CCR2/metabolism
3.
Phytother Res ; 37(5): 1787-1805, 2023 May.
Article in English | MEDLINE | ID: mdl-36437582

ABSTRACT

Ischemia/hypoxia (I/H)-induced myocardial injury has a large burden worldwide. Hesperetin (HSP) has a cardioprotective effect, but the molecular mechanism underlying this is not clearly established. Here, we focused on the protective mechanisms of HSP against I/H-induced myocardium injury. H9c2 cardiomyocytes were challenged with CoCl2 for 22 h to imitate hypoxia after treatment groups received HSP for 4 h. The viability of H9c2 cardiomyocytes was evaluated, and cardiac function indices, reactive oxygen species, apoptosis, mitochondrial membrane potential (MMP), and intracellular Ca2+ concentration ([Ca2+ ]i ) were measured. L-type Ca2+ current (ICa-L ), myocardial contraction, and Ca2+ transients in isolated ventricular myocytes were also recorded. We found that HSP significantly increased the cell viability, and MMP while significantly decreasing cardiac impairment, oxidative stress, apoptosis, and [Ca2+ ]i caused by CoCl2 . Furthermore, HSP markedly attenuated ICa-L , myocardial contraction, and Ca2+ transients in a concentration-dependent manner. Our findings suggest a protective mechanism of HSP on I/H-induced myocardium injury by restoring oxidative balance, inhibiting apoptosis, improving mitochondrial function, and reducing Ca2+ influx via L-type Ca2+ channels (LTCCs). These data provide a new direction for HSP applied research as a LTCC inhibitor against I/H-induced myocardium injury.


Subject(s)
Myocytes, Cardiac , Oxidative Stress , Humans , Hypoxia , Homeostasis , Ischemia/metabolism , Apoptosis
4.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-973637

ABSTRACT

Background The exposure to diesel particulate matter (DPM) and its polycyclic aromatic hydrocarbons (PAH) is closely related to the morbidity and mortality of ischemic heart disease (IHD). However, it is unclear what key components and targets of DPM exposure involve in myocardial ischemia-hypoxia injury and associated mechanisms. Objective To identify key PAH components of DPM that act on myocardial hypoxic injury, andclarify the role of oxygen sensors-regulated anaerobic metabolism in DPM and key components-induced hypoxic injury and the targets of the key PAH components. Methods Human cardiomyocyte cell line AC16 cells were exposed to 0, 1, 5, and 10 μg·mL−1 DPM in a high glucose DMEM medium with 10% fetal bovine serum (FBS) (HGM) or low FBS (0.5%) in high glucose DMEM medium (LFM), for 12 h under 2% O2, and expression of hypoxia-inducible factor-1α (HIF-1α), Bax, and Cleaved-caspase3 was determined by Western blotting. Under normal condition, the cell viability was detected after PAH exposure for 12 h. Under the condition of ischemia-hypoxia model, cells were exposed to 0, 0.005, 0.5, and 5 µg·mL−1 PAH for 12 h, and the protein expression of HIF-1α, Bax, and Cleaved-caspase3 was determined. After exposure to DPM or PAH for 12 h, the contents of pyruvate and lactate in cells were detected. Pretreatment with glycolysis inhibitor GSK2837808A was used to explore the role of glycolysis in DPM and benzo[a]pyrene (BaP)-induced hypoxia injury. A molecular docking technique was used to analyze the binding affinity between PAH and oxygen sensors (prolyl hydroxylase domain-containing protein 2, PHD2, and factor-inhibiting hypoxia-inducible factor 1, FIH1), and the protein levels of PHD2, FIH1, and hydroxyl-HIF-1-alpha (OH-HIF-1α) after the DPM or BaP treatment were further determined. Results Under hypoxia, DPM exposure in the LFM induced the expression of HIF-1α, Bax, and Cleaved-caspase3 (P<0.01). Therefore, hypoxia and LFM were selected as the basic ischemia and hypoxia condition. Except for anthracene (Ant) (P>0.05), other PAH decreased cell viability when the concentration was above 1 μg·mL−1 (P<0.05). All concentrations of BaP induced the expression of HIF-1α protein (P<0.05), and the protein levels of Bax and Cleaved-caspase3 were up-regulated after the 0.5 and 5 µg·mL−1 BaP exposure (P<0.01). After exposure to DPM (1, 5 and 10 μg·mL−1) or BaP (0.5 and 5 μg·mL−1), the intracellular pyruvate and lactate contents increased (P<0.05). The glycolysis inhibitor co-treatment decreased the levels of HIF-1α, Bax, and Cleaved-caspase3 proteins compared with the DPM or BaP exposure group for 12 h (P<0.05). The binding abilities of the five PAHs to the oxygen sensors PHD2 and FIH1 were strong, and BaP was the strongest. Although the DPM or BaP exposure had no effects on the protein levels of PHD2 and FIH1 in AC16 cells (P<0.05), the protein level of OH-HIF-1α was decreased (P<0.01). Conclusion BaP exposure can promote hypoxia and injury of myocardial cells and is the key PAH component of DPM that induces myocardial ischemia and hypoxia injury. BaP exposure inhibits the hydroxylation function of PHD2 on HIF-1α by combining with PHD2, decreases the level of OH-HIF-1α and induces HIF-1α accumulation. And then HIF-1α promotes anaerobic metabolism and accelerates ischemia and hypoxia injury of myocardial cells.

5.
Int Heart J ; 63(2): 327-337, 2022.
Article in English | MEDLINE | ID: mdl-35354753

ABSTRACT

Emerging literature suggests that pyroptosis plays a critical role in ischemia/hypoxia (I/R) -induced myocardial injury. Melatonin has been implicated in attenuating I/R-induced injury of cardiomyocytes. Nevertheless, whether melatonin inhibits I/R-induced pyroptosis of cardiomyocytes and the underlying molecular mechanisms remain unexploited.H9c2 cardiomyocytes were cultured under oxygen-glucose deprivation/reperfusion (OGD/R) condition to establish a myocardial pyroptosis model in vitro. OGD/R-induced pyroptosis was evaluated by CCK-8 assay, IL-1ß and IL-18 release, and western blotting. Luciferase reporter assay was utilized to validate the association between miR-155 and Forkhead box O3a (FOXO3a).Melatonin could inhibit OGD/R-induced pyroptosis of H9c2 cells and upregulation of FOXO3a contributed to the antipyroptotic effect of melatonin. Melatonin reduced miR-155 expression, which led to FOXO3a upregulation and inhibition of pyroptosis in OGD/R-exposed H9c2 cells. miR-155 inhibitor enhanced the antipyroptotic effect of melatonin in OGD/R-exposed H9c2 cells. Melatonin-induced downregulation of miR-155 and upregulation of FOXO3a were reversed by melatonin receptor 2 (MT2) siRNA. Melatonin treatment also led to an increased level of apoptosis repressor with caspase recruitment domain (ARC), which was inhibited by FOXO3a siRNA. Moreover, silencing ARC by siRNA significantly blocked the antipyroptotic actions of melatonin, whereas ARC overexpression enhanced the antipyroptotic actions of melatonin in OGD/R-exposed H9c2 cells.Our findings demonstrated that melatonin prevented OGD/R-induced pyroptosis via regulating the MT2/miR-155/FOXO3a/ARC axis in cardiomyocytes.


Subject(s)
Melatonin , MicroRNAs , Myocardial Reperfusion Injury , Glucose/metabolism , Humans , Melatonin/metabolism , Melatonin/pharmacology , MicroRNAs/genetics , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Oxygen/metabolism , Oxygen/pharmacology , Pyroptosis/genetics , Reperfusion
6.
Front Cardiovasc Med ; 9: 819454, 2022.
Article in English | MEDLINE | ID: mdl-35187131

ABSTRACT

Coronary heart disease (CHD) is one of the main causes of death worldwide. In the past few decades, several in-depth research on the pathological mechanisms and effective treatment methods for CHD have been conducted. At present, the intervention of a variety of therapeutic drugs and treatment technologies have greatly reduced the burden on global public health. However, severe arrhythmia and myocardial fibrosis accompanying CHD in the later stages need to be addressed urgently. Mitochondria are important structural components for energy production and the main sites for aerobic respiration in cells. Mitochondria are involved in arrhythmia, myocardial fibrosis, and acute CHD and play a crucial role in regulating myocardial ischemia/hypoxia. Mitochondrial dysfunction or mitophagy disorders (including receptor-dependent mitophagy and receptor-independent mitophagy) play an important role in the pathogenesis of CHD, especially mitophagy. Mitophagy acts as a "mediator" in the inflammatory damage of cardiomyocytes or vascular endothelial cells and can clear mitochondria or organelles damaged by inflammation under normal conditions. We reviewed experimental advances providing evidence that mitochondrial homeostasis or mitochondrial quality control are important in the pathological mechanism of CHD. Further, we reviewed and summarized relevant regulatory drugs that target mitochondrial function and quality control.

7.
Biomed Pharmacother ; 147: 112674, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35093758

ABSTRACT

To investigate the protection of cinnamic aldehyde (CA) against myocardial ischemia/hypoxia (I/H) injury and its potential mechanisms in vivo and in vitro. Mice were pretreated with CA for 7 days, and then isoproterenol (85 mg/kg) was administered for 2 consecutive days to assess its cardioprotection. Furthermore, an in vitro myocardial I/H model was established by administering CoCl2 (600 µM) to H9c2 cells for 24 h. H9c2 cells were pretreated with CA for 12 h to assess its protection. We observed that CA improved electrocardiogram and histopathological changes and decreased creatine kinase and lactate dehydrogenase activities and oxidative stress levels. The TUNEL results showed that CA reduced the degree of apoptosis. Furthermore, CA could lead to a down-regulation of the Caspase-3 and Bax protein expressions, but an up-regulation of the Bcl-2 protein expressions. Importantly, CA increased p-PI3K and p-AKT protein expressions, indicating the activation of the PI3K/AKT signaling pathway. Moreover, treatment with CA improved the cell viability rate and mitochondrial membrane potential while markedly decreasing apoptosis and oxidative stress levels in vitro. Our results suggested that CA exerts cardioprotection on myocardial I/H injury, which possibly occurred in connection with inhibition of oxidative stress and apoptosis via activation of the PI3K/AKT signaling pathway.


Subject(s)
Acrolein/analogs & derivatives , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/pathology , Phosphatidylinositol 3-Kinases/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Acrolein/pharmacology , Animals , Animals, Outbred Strains , Apoptosis/drug effects , Cell Line , Creatine Kinase/drug effects , Dose-Response Relationship, Drug , L-Lactate Dehydrogenase/drug effects , Male , Mice , Oxidative Stress/drug effects , Random Allocation
8.
Biomedicines ; 11(1)2022 Dec 29.
Article in English | MEDLINE | ID: mdl-36672601

ABSTRACT

The central nervous system (CNS) injury, which occurs because of mechanical trauma or ischemia/hypoxia, is one of the main causes of mortality and morbidity in the modern society. Until know, despite the fact that numerous preclinical and clinical studies have been undertaken, no significant neuroprotective strategies have been discovered that could be used in the brain trauma or ischemia treatment. Although there are many potential explanations for the failure of those studies, it is clear that there are questions regarding the use of experimental models, both in vivo and in vitro, when studying CNS injury and searching new therapeutics. Due to some ethical issues with the use of live animals in biomedical research, implementation of experimental strategies that prioritize the use of cells and tissues in the in vitro environment has been encouraged. In this review, we examined some of the most commonly used in vitro models and the most frequently utilized cellular platforms in the research of traumatic brain injury and cerebral ischemia. We also proposed some future strategies that could improve the usefulness of these studies for better bench-to-bedside translational outcomes.

9.
Am J Transl Res ; 14(12): 8611-8620, 2022.
Article in English | MEDLINE | ID: mdl-36628240

ABSTRACT

OBJECTIVES: Angiotensin receptor-neprilysin inhibitor (ARNI) improves cardiac function and protects from an ischemic myocardium. However, the role and mechanism of ARNI on autophagy in cardiac ischemic injury are poorly understood. Here, we investigated the protective effect and underlying mechanisms of ARNI on autophagy in H9c2 cardiomyocytes induced through ischemia and hypoxia (IH) treatment. METHODS: The cytotoxicity of IH injury on H9C2 cells with and without ARNI were evaluated using cell counting kit-8 (CCK-8) and lactate dehydrogenase (LDH) release assays. The effect of ARNI on apoptosis was detected using flow cytometry. The expression of autophagic proteins (LC3-II, Beclin 1, and p62) was detected using western blot. RESULTS: The viability of H9c2 cells was significantly decreased at different IH-treated time points; ARNI pretreatment increased cell viability and inhibited IH injury in a dose-dependent manner. H9c2 cells treated with IH (6 h) significantly increased LDH release, while ARNI dose-dependently improved LDH release, with 20 µmol/L ARNI having the most significant effect. ARNI also ameliorated IH-induced apoptosis. IH treatment increased the protein expression of LC3-II and Beclin 1 and decreased the expression of p62, which were reversed by ARNI pretreatment. Furthermore, autophagy was further increased after pretreatment with rapamycin in IH-induced H9c2 cells, which abrogated the protective effect of ARNI. CONCLUSIONS: Our study shows that ARNI has a protective effect on IH-induced cardiomyocyte injury, which may be related to the inhibition of autophagy.

10.
J Am Heart Assoc ; 10(22): e022011, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34726071

ABSTRACT

Background Acute myocardial infarction (AMI) is one of the leading causes of cardiovascular morbidity and mortality worldwide. Pyroptosis is a form of inflammatory cell death that plays a major role in the development and progression of cardiac injury in AMI. However, the underlying mechanisms for the activation of pyroptosis during AMI are not fully elucidated. Methods and Results Here we show that RBP4 (retinol-binding protein 4), a previous identified proinflammatory adipokine, was increased both in the myocardium of left anterior descending artery ligation-induced AMI mouse model and in ischemia-hypoxia‒induced cardiomyocyte injury model. The upregulated RBP4 may contribute to the activation of cardiomyocyte pyroptosis in AMI because overexpression of RBP4 activated NLRP3 (nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3) inflammasome, promoted the precursor cleavage of Caspase-1, and subsequently induced GSDMD (gasdermin-D)-dependent pyroptosis. In contrast, knockdown of RBP4 alleviated ischemia-hypoxia‒induced activation of NLRP3 inflammasome signaling and pyroptosis in cardiomyocytes. Mechanistically, coimmunoprecipitation assay showed that RBP4 interacted directly with NLRP3 in cardiomyocyte, while genetic knockdown or pharmacological inhibition of NLRP3 attenuated RBP4-induced pyroptosis in cardiomyocytes. Finally, knockdown of RBP4 in heart decreased infarct size and protected against AMI-induced pyroptosis and cardiac dysfunction in mice. Conclusions Taken together, these findings reveal RBP4 as a novel modulator promoting cardiomyocyte pyroptosis via interaction with NLRP3 in AMI. Therefore, targeting cardiac RBP4 might represent a viable strategy for the prevention of cardiac injury in patients with AMI.


Subject(s)
Heart Injuries , Myocardial Infarction , Animals , Hypoxia , Inflammasomes/metabolism , Mice , Myocardial Infarction/genetics , Myocytes, Cardiac/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis , Retinol-Binding Proteins , Retinol-Binding Proteins, Plasma
11.
Zhonghua Gan Zang Bing Za Zhi ; 29(7): 705-710, 2021 Jul 20.
Article in Chinese | MEDLINE | ID: mdl-34371544

ABSTRACT

Ischemic hepatitis is inflammation caused by necrosis of liver cells due to ischemia and hypoxia caused by low cardiac output or septic shock. It is often complicated by heart failure or severe septic shock. One of the pathogenesis of ischemic hepatitis is hepatocyte injury caused by ischemia and hypoxia, which results in damage-associated molecular patterns (DAMPs) release and binding to membrane receptors such as toll like receptors (TLRs) to cause inflammatory reactions.The other is when the ischemic liver is reperfused, hepatocyte mitochondrias will produce a large amount of ROS causing ischemia reperfusion injury. These two mechanisms and related molecular pathways are elaborated in this paper.


Subject(s)
Hepatitis , Reperfusion Injury , Hepatitis/etiology , Hepatocytes , Humans , Ischemia , Liver
12.
Bioengineered ; 12(1): 4385-4396, 2021 12.
Article in English | MEDLINE | ID: mdl-34304702

ABSTRACT

Myocardial ischemia/hypoxia-reperfusion injury mediates the progression of multiple cardiovascular diseases. It has been reported that knockdown of adaptor protein containing a PH domain, PTB domain and leucine zipper motif 1 (APPL1) is a significant factor for the progression of myocardial injury. However, the role of APPL1 in myocardial ischemia remains unclear. Hence, the aim of the present study was to investigate the specific mechanism underlying the role of APPL1 in myocardial ischemia.In our study, the mRNA level of APPL1 was detected by quantitative real-time PCR (RT-qPCR). The expressions of APPL1, Apoptotic protease activating factor-1 (APAF-1), cleaved caspase9 and other inflammation- and apoptosis-related proteins were determined by western blotting. The secretion of inflammatory cytokines and lactate dehydrogenase (LDH) levels were measured by commercial assay kits. The H9C2 cell viability was analyzed by cell counting kit-8 (CCK-8) assay. The apoptosis rate of H9C2 cells was analyzed by TUNEL assay. The interaction between APPL1 and APAF-1/caspase9 was determined by Immunoprecipitation (IP).Our findings demonstrated that APPL1 was low expressed in myocardial ischemia tissues and cells. APPL1 knockdown suppressed the viability of myocardial ischemia cells and aggravated hypoxia/reperfusion-induced LDH hypersecretion, inflammation and apoptosis. In addition, the overexpression of APPL1 induced inactivation of APAF-1/Caspase9 signaling pathway. Significantly, APAF1 inhibitor reversed the effect of APPL1 knockdown on viability, LDH secretion, inflammation and apoptosis.We conclude that APPL1 inhibits myocardial ischemia/hypoxia-reperfusion injury via inactivation of APAF-1/Caspase9 signaling pathway. Hence, APPL1 may be a novel and effective target for the treatment of myocardial ischemia.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptotic Protease-Activating Factor 1/metabolism , Caspase 9/metabolism , Myocardial Reperfusion Injury/metabolism , Nerve Tissue Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptotic Protease-Activating Factor 1/genetics , Caspase 9/genetics , Cell Line , Heart , Male , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , Nerve Tissue Proteins/genetics , Rats , Signal Transduction/genetics
13.
Front Cardiovasc Med ; 8: 796254, 2021.
Article in English | MEDLINE | ID: mdl-35004909

ABSTRACT

Lysosomal dysfunction has been found in many pathological conditions, and methods to improve lysosomal function have been reported to be protective against infarcted hearts. However, the mechanisms underlying lysosomal dysfunction caused by ischemic injury are far less well-established. The retromer complex is implicated in the trafficking of cation-independent mannose 6-phosphate receptor (CI-MPR), which is an important protein tag for the proper transport of lysosomal contents and therefore is important for the maintenance of lysosomal function. In this study, we found that the function of retrograde transport in cardiomyocytes was impaired with ischemia/hypoxia (I/H) treatment, which resulted in a decrease in CI-MPR and an abnormal distribution of lysosomal cathepsins. I/H treatment caused a reduction in TBC1D5 and a blockade of the Rab7 membrane cycle, which impeded retromer binding to microtubules and motor proteins, resulting in an impairment of retrograde transport and a decrease in CI-MPR. We also established that TBC1D5 was an important regulator of the distribution of lysosomal cathepsins. Our findings shed light on the regulatory role of retromer in ischemic injury and uncover the regulatory mechanism of TBC1D5 over retromer.

14.
Biomed Pharmacother ; 131: 110787, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33152945

ABSTRACT

AIM: Acute myocardial infarction (AMI) is one of the deadliest diseases worldwide. The search for countermeasures to reduce cardiomyocytes death in the infarcted area has always been the focus of research. Ubiquitin (UB) is a small polypeptide mainly involved in proteasome-mediated protein degradation in cells, whereas extracellular UB in body fluids can also function through its receptor CXC chemokine receptor type 4 (CXCR4). This study aimed to explore the functional roles of extracellular UB in cardiomyocytes during ischemia/hypoxia (I/H). METHODS: H9C2 cells were subjected to I/H treatment and cell injury was evaluated by cell viability, morphology changes and apoptosis rate. UB expression and levels of ubiquitinated proteins after I/H injury were measured. The effects of extracellular UB on I/H-induced cardiomyocytes apoptosis and the possible underlying mechanisms were studied. RESULTS: I/H injury induced the decrease of cell viability as well as enhanced impaired cell morphology and apoptosis rate in H9C2 cells. Levels of UB mRNA and ubiquitinated proteins were significantly up-regulated after I/H treatment, whereas the concentration of extracellular UB in the conditioned media did not show significant change and the intracellular mono-UB levels in cells were down-regulated. Extracellular UB treatment protected cardiomyocytes from I/H injury by inhibiting the overactivation of mitochondria-dependent apoptosis pathway and up-regulating autophagy level. Inhibition of CXCR4 receptor using AMD3100 abolished cardioprotective effects of extracellular UB. CONCLUSION: The up-regulation of UB was suggested to be an adaptive response to resist I/H-induced cardiomyocytes apoptosis, and additional extracellular UB treatment might serve as a new potential therapeutic drug for AMI.


Subject(s)
Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Receptors, CXCR4/metabolism , Ubiquitin/metabolism , Animals , Apoptosis/physiology , Autophagy , Benzylamines , Cell Survival/physiology , Cyclams , Down-Regulation , Extracellular Space/metabolism , Heterocyclic Compounds/pharmacology , Hypoxia/complications , Ischemia/complications , Mitochondria/pathology , Rats , Ubiquitin/genetics , Up-Regulation
15.
J Geriatr Cardiol ; 17(5): 284-293, 2020 May.
Article in English | MEDLINE | ID: mdl-32547612

ABSTRACT

BACKGROUND: Myocardial injury caused by microvascular obstruction (MVO) is characterized by persistent ischemia/hypoxia (IH) of cardiomyocytes after microembolization. Autophagy and Egr-1 were closely associated with various cardiovascular diseases, including MVO. Bim and Beclin-1 are the important genes for autophagy and apoptosis. We aimed to explore whether the Egr-1/Bim/Beclin-1 pathway is involved in regulating autophagy and apoptosis in IH-exposed cardiomyocytes. METHODS: Neonatal rat cardiomyocytes exposed to the IH environment in vitro were transfected with lentivirus expressing Egr-1 or Egr-1 shRNA, or further treated with 3-methyladenine (3-MA). The expressions of autophagy and apoptosis-associated genes were evaluated using RT-qPCR and Western blots assays. Autophagic vacuoles and autophagic flux were detected by transmission electron microscopy (TEM) and confocal microscope, respectively. Cell injury was assessed by lactate dehydrogenase (LDH) leakage, and apoptosis was determined by flow cytometry. RESULTS: IH exposure elevated Egr-1 and Bim expressions, and decreased Beclin-1 expression in rat cardiomyocytes. Egr-1 overexpression in IH-exposed cardiomyocytes significantly up-regulated the levels of Egr-1 and Bim, and down-regulated the level of Beclin-1. Egr-1 knockdown resulted in down-regulated expressions of Egr-1 and Bim, as well as up-regulated expression of Beclin-1. In addition, Egr-1 knockdown induced autophagy was suppressed by 3-MA treatments. TEM and autophagic flux experiments also confirmed that Egr-1 inhibited autophagy progression in IH-exposed cardiomyocytes. Egr-1 suppression protected cardiomyocytes from IH-induced injury, as evidenced by the positive correlations between Egr-1 expression and LDH leakage or apoptosis index in IH-exposed cardiomyocytes. CONCLUSIONS: IH-induced cardiomyocyte autophagy and apoptosis are regulated by the Egr-1/Bim/Beclin-1 pathway, which is a potential target for treating cardiomyocyte injury caused by MVO in the IH environment.

16.
In Vitro Cell Dev Biol Anim ; 56(4): 307-312, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32406012

ABSTRACT

Apigenin is a traditional Chinese medicine found in many plants that plays critical roles in several diseases, including cardiovascular diseases. We investigated the protective effect of apigenin against ischemia/hypoxia (I/H)-induced myocardium injury in vitro and explored the potential molecular mechanisms. Together with the flow cytometry results, our results indicated that apigenin attenuated the pyroptosis and apoptosis of myoblastic H9c2 cells that were induced by I/H injury. Furthermore, the pro-inflammatory cytokines (interleukin 18, IL-18, and IL-1ß) were investigated for the roles of apigenin in I/H-induced myocardium injury. It was observed that increases in IL-1ß and IL-18 levels were significantly reduced by apigenin treatment of I/H-induced H9C2 cells. The results demonstrated that apigenin protected H9c2 cells against I/H-induced myocardium injury. The protective effects were most likely related to the reduction of pyroptosis, apoptosis, and pro-inflammatory cytokines.


Subject(s)
Apigenin/pharmacology , Apoptosis/drug effects , Cardiotonic Agents/pharmacology , Myocardial Reperfusion Injury/pathology , Pyroptosis/drug effects , Animals , Cell Hypoxia/drug effects , Cell Line , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Rats
17.
Clin Neurol Neurosurg ; 193: 105770, 2020 06.
Article in English | MEDLINE | ID: mdl-32197144

ABSTRACT

OBJECTIVE: White matter lesions (WMLs) are more common in migraine patients than in the normal population. Ischemia/hypoxia and oxidative stress are considered to play a role in WMLs formation. This study aimed to investigate ischemia-modified albumin (IMA), ferroxidase and thiol/disulfide homeostasis in migraineurs with and without WMLs. PATIENTS AND METHODS: Sixty-two migraineurs with WML, 59 migraineurs without WML and 61 controls were included in the study. All participants underwent brain MRI. WMLs was evaluated according to the Fazekas scale. IMA, ferroxidase, total thiol, native thiol and disulfide measurements were carried out in all participants. RESULTS: The IMA levels were higher in the migraine groups compared to the control group (p < 0.001) and in the WML group compared to non-WML (p < 0.001). The total and native thiol levels were higher in the non-WML group compared to the control and WML groups (p < 0.001 for both). The disulfide levels were similar between the control and non-WML groups, but they were significantly lower in the WML group compared to the control and non-WML groups. There was no significant difference between the groups in terms of the ferroxidase levels (p = 0.092). The thiol/disulfide, IMA and ferroxidase levels were not significantly correlated with the frequency and duration of attacks, severity of pain and disability due to migraine. CONCLUSION: Increased serum IMA levels in migraineurs point to the role of ischemia/hypoxia, and increased total thiol and decreased disulfide levels indicate an oxidant/antioxidant imbalance in migraine. Ischemia/hypoxia may play a role in WMLs formation in migraine.


Subject(s)
Brain Ischemia/pathology , Migraine Disorders/pathology , White Matter/pathology , Adolescent , Adult , Biomarkers/metabolism , Ceruloplasmin/metabolism , Disulfides/metabolism , Female , Humans , Hypoxia, Brain/metabolism , Hypoxia, Brain/pathology , Magnetic Resonance Imaging , Male , Middle Aged , Oxidative Stress , Pain Measurement , Serum Albumin, Human/metabolism , Sulfhydryl Compounds/metabolism , Young Adult
18.
Hum Cell ; 33(3): 545-558, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32146707

ABSTRACT

This study aimed to investigate the usefulness of the thyroid-related hormones as markers of acute systemic hypoxia/ischemia to identify deaths caused by asphyxiation due to neck compression in human autopsy cases. The following deaths from pathophysiological conditions were examined: mechanical asphyxia and acute/subacute blunt head injury; acute/subacute non-head blunt injury; sharp instrument injury as the hemorrhagic shock condition; drowning as alveolar injury; burn; and death due to cardiac dysfunction. Blood samples were collected from the left and right cardiac chambers and iliac veins, and serum triiodothyronine (T3), thyroxine (T4), thyroglobulin (Tg), and thyroid-stimulating hormone (TSH) levels were measured using electrochemiluminescence immunoassays. Two types of thyroid cell lines were used to confirm independent thyroid function under the condition of hypoxia (3% O2). The human thyroid carcinoma cell line (HOTHC) cell line derived from human anaplastic thyroid carcinoma and the UD-PTC (sample of the second resection papillary thyroid carcinoma) cell line derived from human thyroid papillary adenoma, which forms Tg retention follicles, were used to examine the secretion levels of T3, T4, and Tg hormones. The results showed a strong correlation between T3 and T4 levels in all blood sampling sites, while the TSH and Tg levels were not correlated with the other markers. Serum T3 and T4 levels were higher in cases of mechanical asphyxia and acute/subacute blunt head injury, representing hypoxic and ischemic conditions of the brain as compared to those in other causes of death. In the thyroid gland cell line, T4, T3, and Tg levels were stimulated after exposure to hypoxia for 10-30 min. These findings suggest that systemic advanced hypoxia/ischemia may cause a rapid and TSH-independent release of T3 and T4 thyroid hormones in autopsy cases. These findings demonstrate that increased thyroid-related hormone (T3 and T4) levels in the pathophysiological field may indicate systemic hypoxia/ischemia.


Subject(s)
Asphyxia/diagnosis , Hypoxia/diagnosis , Ischemia/diagnosis , Thyroglobulin/blood , Thyrotropin/blood , Triiodothyronine/blood , Adult , Aged , Aged, 80 and over , Autopsy , Biomarkers/blood , Female , Head Injuries, Closed , Humans , Male , Middle Aged , Thyroxine
19.
J Physiol Sci ; 70(1): 10, 2020 Feb 17.
Article in English | MEDLINE | ID: mdl-32066368

ABSTRACT

Exercise has been reported to induce autophagy. We hypothesized that exercise preconditioning (EP)-related autophagy in cardiomyocytes could be attributed to intermittent ischemia-hypoxia, allowing the heart to be protected for subsequent high-intensity exercise (HE). We applied approaches, chromotrope-2R brilliant green (C-2R BG) staining and plasma cTnI levels measuring, to characterize two periods of cardioprotection after EP: early EP (EEP) and late EP (LEP). Further addressing the relationship between ischemia-hypoxia and autophagy, key proteins, Beclin1, LC3, Cathepsin D, and p62, were determined by immunohistochemical staining, western blotting, and by their adjacent slices with C-2R BG. Results indicated that exercise-induced ischemia-hypoxia is a key factor in Beclin1-dependent autophagy. High-intensity exercise was associated with the impairment of autophagy due to high levels of LC3II and unchanged levels of p62, intermittent ischemia-hypoxia by EP itself plays a key role in autophagy, which resulted in more favorable cellular effects during EEP-cardioprotection compared to LEP.


Subject(s)
Autophagy , Heart Injuries/metabolism , Ischemic Preconditioning, Myocardial/methods , Physical Conditioning, Animal/adverse effects , Reperfusion Injury , Troponin I/blood , Animals , Gene Expression Regulation/physiology , Humans , Myocardial Ischemia/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Troponin I/genetics , Troponin I/metabolism
20.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-847756

ABSTRACT

BACKGROUND: Traditional Chinese medicine has certain value and significance in the treatment of ischemic cardiovascular and cerebrovascular diseases by regulating the ischemia-hypoxia microenvironment and improving the survival rate and differentiation rate of stem cells. OBJECTIVE: To sort out and analyze the research progress of traditional Chinese medicine on regulating ischemia-hypoxia microenvironment intervention on proliferation, differentiation, aging and autophagy of bone marrow mesenchymal stem cells in recent years. METHODS: The full-text database of Chinese journals, PubMed and Wanfang were retrieved with the keywords of “bone mesenchymal stem cells, ischemia-hypoxia microenvironment, proliferation, differentiation, aging” in English or “bone marrow mesenchymal stem cells, ischemia and hypoxia, proliferation, differentiation, aging” in Chinese for articles regarding effects of ischemia and hypoxia microenvironment on survival rate and differentiation rate of bone marrow mesenchymal stem cells published from 2002 to 2019. Fifty-five articles were selected for review, including 22 Chinese articles and 33 English articles. RESULTS AND CONCLUSION: The ischemia-hypoxia microenvironment is the important reason for the low survival rate and differentiation rate of bone marrow mesenchymal stem cells. There are many adverse reactions in the intervention of bone marrow mesenchymal stem cells with gene modification or cell molecules and drugs, which have become difficult problems to be solved in modern medicine. Exploring the internal relationship between microenvironment and stem cells using single or active components of traditional Chinese medicine combined with RNA transcriptomics is a new way to improve the viability of stem cells.

SELECTION OF CITATIONS
SEARCH DETAIL
...