1.MiR -18a -5p aggravates homocysteine -induced myocardial injury via autophagy.
Juan YIN ; Longlong HU ; Xueling HAN ; Lu CHEN ; Lingling YU ; Yinhui LU
Journal of Central South University(Medical Sciences) 2023;48(1):24-33
OBJECTIVES:
Hyperhomocysteinaemia (Hcy) is an independent risk factor for cardiovascular and cerebrovascular diseases. MicroRNA (miR)-18a-5p is closely related to cardiovascular diseases. This study aims to investigate the effects of miR-18a-5p on homocysteine (Hcy)-induced myocardial cells injury.
METHODS:
H9c2 cells were transfected with miR-18a-5p mimic/miR-18a-5p mimic negative control (NC) or combined with Hcy for intervention, and untreated cells were set as a control group. The transfection efficiency was verified by real-time RT-PCR, and cell counting kit-8 (CCK-8) assay was used to determine cell viability. Flow cytometry was used to detect apoptosis and reactive oxygen species (ROS) levels. Western blotting was performed to measure the protein levels of microtubule-associated protein 1 light chain 3 (LC3)-I, LC3-II, Beclin1, p62, Bax, Bcl-2, and Notch2. Dual luciferase reporter assay was used to detect the interaction of miR-18a-5p with Notch2.
RESULTS:
Compared with the control, treatment with Hcy or transfection with miR-18a-5p mimic alone, or combined treatment with Hcy and miR-18a-5p mimic/miR-18a-5p mimic NC significantly reduced the H9c2 cell viability, promoted apoptosis and ROS production, up-regulated the expressions of Bax and Beclin, down-regulated the expressions of Bcl-2, p62, and Notch2, and increased the ratio of LC3-II/LC3-I (all P<0.05). Compared with the combined intervention of miR-18a-5p mimic NC and Hcy group, the above indexes were more significantly changed in the combined intervention of miR-18a-5p mimic and Hcy group, and the difference between the 2 groups was statistically significant (all P<0.05). There is a targeted binding between Notch2 and miR-18a-5p.
CONCLUSIONS
MiR-18a-5p could induce autophagy and apoptosis via increasing ROS production in cardiomyocytes, and aggravate Hcy-induced myocardial injury. Notch2 is a target of miR-18a-5p.
Apoptosis/genetics*
;
Autophagy/genetics*
;
bcl-2-Associated X Protein
;
MicroRNAs/metabolism*
;
Proto-Oncogene Proteins c-bcl-2/genetics*
;
Reactive Oxygen Species
;
Rats
;
Animals
;
Myocytes, Cardiac/drug effects*
;
Homocysteine/adverse effects*
;
Hyperhomocysteinemia
2.Inhibition of chemotherapy-related breast tumor EMT by application of redox-sensitive siRNA delivery system CSO-ss-SA/siRNA along with doxorubicin treatment.
Xuan LIU ; Xue-Qing ZHOU ; Xu-Wei SHANG ; Li WANG ; Yi LI ; Hong YUAN ; Fu-Qiang HU
Journal of Zhejiang University. Science. B 2020;21(3):218-233
Metastasis is one of the main reasons causing death in cancer patients. It was reported that chemotherapy might induce metastasis. In order to uncover the mechanism of chemotherapy-induced metastasis and find solutions to inhibit treatment-induced metastasis, the relationship between epithelial-mesenchymal transition (EMT) and doxorubicin (DOX) treatment was investigated and a redox-sensitive small interfering RNA (siRNA) delivery system was designed. DOX-related reactive oxygen species (ROS) were found to be responsible for the invasiveness of tumor cells in vitro, causing enhanced EMT and cytoskeleton reconstruction regulated by Ras-related C3 botulinum toxin substrate 1 (RAC1). In order to decrease RAC1, a redox-sensitive glycolipid drug delivery system (chitosan-ss-stearylamine conjugate (CSO-ss-SA)) was designed to carry siRNA, forming a gene delivery system (CSO-ss-SA/siRNA) downregulating RAC1. CSO-ss-SA/siRNA exhibited an enhanced redox sensitivity compared to nonresponsive complexes in 10 mmol/L glutathione (GSH) and showed a significant safety. CSO-ss-SA/siRNA could effectively transmit siRNA into tumor cells, reducing the expression of RAC1 protein by 38.2% and decreasing the number of tumor-induced invasion cells by 42.5%. When combined with DOX, CSO-ss-SA/siRNA remarkably inhibited the chemotherapy-induced EMT in vivo and enhanced therapeutic efficiency. The present study indicates that RAC1 protein is a key regulator of chemotherapy-induced EMT and CSO-ss-SA/siRNA silencing RAC1 could efficiently decrease the tumor metastasis risk after chemotherapy.
Amines/chemistry*
;
Antineoplastic Agents/adverse effects*
;
Breast Neoplasms/pathology*
;
Chitosan/chemistry*
;
Doxorubicin/adverse effects*
;
Drug Delivery Systems
;
Epithelial-Mesenchymal Transition/drug effects*
;
Female
;
Humans
;
MCF-7 Cells
;
Neoplasm Metastasis/prevention & control*
;
Oxidation-Reduction
;
RNA, Small Interfering/administration & dosage*
;
Reactive Oxygen Species/metabolism*
;
rac1 GTP-Binding Protein/physiology*
3.Effects of high glucose induced primary cardiomyocytes injury on necroptosis and the related mechanism.
Ting Ting FANG ; Rui Ping CAO ; Hong Wei YE ; Shan Feng MA ; Qin GAO
Chinese Journal of Applied Physiology 2019;35(2):160-164
OBJECTIVE:
To observe whether necroptosis was happened in high glucose (HG) - induced primary cardiomyocytes injury and to investigate the likely mechanism.
METHODS:
The primary cultured cardiomyocytes were divided into 4 groups (n=9): control group (the cardiomyocytes were incubated with 5.5 mmol/L glucose for 48 h), HG group (the cardiomyocytes were incubated with 30 mmol/L glucose for 48 h), HG + necrostatin-1 (Nec-1) group (the cardiomyocytes was co-incubated with necroptosis inhibitor Nec-1 at 100 μmol/L and HG for 48 h) and hypertonic pressure group (HPG, the cardiomyocytes was co-incubated with 5.5 mmol/L glucose and 24.5 mmol/L mannitol for 48 h). Cell viability was measured by MTT method, reactive oxygen species (ROS) generation was measured by DHE staining. The levels of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and interleukin-1β (IL-1β) were tested by ELISA method. The mRNA and protein expressions of necroptosis related genes receptor interacting serine/threonine protein kinase 1 (RIP1), RIP3, mixed lineage kinase domain-like protein (MLKL) were tested by quantitative real-time PCR and Western blot.
RESULTS:
The results showed HG intervention decreased cardiomyocytes viability, increased ROS generation, up-regulated the levels of TNF-α, IL-6 and IL-1β, increased RIP1, RIP3, MLKL expressions at mRNA and protein levels. Nec-1 treatment attenuated HG-induced increased cardiomyocytes viability, reduced ROS generation, down-regulated the levels of TNF-α, IL-6 and IL-1β, decreased RIP1, RIP3, MLKL expressions at mRNA and protein levels.
CONCLUSION
Necroptosis was happened in high glucose-induced primary cardiomyocytes injury. Inhibition of necroptosis can reduce high glucose-induced cardiomyocytes damage, may be related to inhibition of oxidative stress and depression of inflammative factors releasing.
Apoptosis
;
Cells, Cultured
;
Cytokines
;
metabolism
;
Glucose
;
adverse effects
;
Humans
;
Myocytes, Cardiac
;
cytology
;
pathology
;
Necrosis
;
Oxidative Stress
;
Reactive Oxygen Species
;
metabolism
4.New advances in renal mechanisms of high fructose-induced salt-sensitive hypertension.
Acta Physiologica Sinica 2018;70(6):581-590
Fructose intake has increased dramatically over the past century and the upward trend has continued until recently. Increasing evidence suggests that the excessive intake of fructose induces salt-sensitive hypertension. While the underlying mechanism is complex, the kidney likely plays a major role. This review will highlight recent advances in the renal mechanisms of fructose-induced salt-sensitive hypertension, including (pro)renin receptor-dependent activation of intrarenal renin-angiotensin system, increased nephron Na transport activity via sodium/hydrogen exchanger 3 and Na/K/2Cl cotransporter, increased renal uric acid production, decreased renal nitric oxide production, and increased renal reactive oxygen species production, and suggest actions based on these mechanisms that have therapeutic implications.
Blood Pressure
;
Fructose
;
adverse effects
;
Humans
;
Hypertension
;
chemically induced
;
physiopathology
;
Kidney
;
physiopathology
;
Nitric Oxide
;
metabolism
;
Reactive Oxygen Species
;
metabolism
;
Renin-Angiotensin System
;
Sodium Chloride, Dietary
;
adverse effects
;
Sodium-Hydrogen Exchanger 3
;
metabolism
;
Uric Acid
;
metabolism
5.Protective effect of dexmedetomidine against glutamate-induced cytotoxicity in PC12 cells and its mechanism.
Wei-Dong ZHANG ; Hao ZHANG ; Hai WANG ; Na ZHANG ; Chun-Yan DU ; Jun YU ; Ze-Guo FENG
Journal of Southern Medical University 2016;37(2):150-156
OBJECTIVETo investigate the protective effects of dexmedetomidine (Dex) against glutamate-induced cytotoxicity in PC12 cells and its mechanism.
METHODSPC12 cells were treated with varying concentrations of dexmedetomidine 1 h before exposure to a high concentration of glutamate. The cell viability was measured by MTT assay, and LDH release, MDA content and SOD activity were measured. The level of ROS was tested by DCFH-DA staining and flow cytometry. The level of intracellular Cawas detected by Fluo-8 staining and flow cytometry, and the mitochondrial membrane potential (MMP) was determined with JC-1 staining and flow cytometry.
RESULTSWithin the concentration range of 0.01 to 100 µmol/L, Dex dose-dependently protected PC12 cells against glutamate-induced cytotoxicity. Treatment with 100 µmol/L Dex significantly increased the cell viability to (86.6∓2.2)% of that of the control cells (P<0.01) and decreased LDH release to 1.4∓0.1 folds of the control level (P<0.01). In PC12 cells exposed to glutamate, Dex pretreatment significantly reduced MDA content (P<0.01), enhanced SOD activity (P<0.01), inhibited ROS overproduction (P<0.01), reduced intracellular Calevel (P<0.01) and maintained a stable MMP (P<0.01).
CONCLUSIONDexmedetomidine can protect PC12 cells against glutamate-induced injury possibly in relation with its anti-oxidative activity, inhibitory effect on intracellular calcium overload and protective effect of the mitochondria.
Animals ; Apoptosis ; Calcium ; metabolism ; Cell Survival ; drug effects ; Dexmedetomidine ; pharmacology ; Glutamic Acid ; adverse effects ; Membrane Potential, Mitochondrial ; Mitochondria ; drug effects ; metabolism ; PC12 Cells ; Rats ; Reactive Oxygen Species ; metabolism
6.Aqueous extracts of Tribulus terrestris protects against oxidized low-density lipoprotein-induced endothelial dysfunction.
Yue-hua JIANG ; Chuan-hua YANG ; Wei LI ; Sai WU ; Xian-qing MENG ; Dong-na LI
Chinese journal of integrative medicine 2016;22(3):193-200
OBJECTIVETo investigate the role of aqueous extracts of Tribulus terrestris (TT) against oxidized low-density lipoprotein (ox-LDL)-induced human umbilical vein endothelial cells (HUVECs) dysfunction in vitro.
METHODSHUVECs were pre-incubated for 60 min with TT (30 and 3 μg/mL respectively) or 10(-5) mol/L valsartan (as positive controls) and then the injured endothelium model was established by applying 100 μg/mL ox-LDL for 24 h. Cell viability of HUVECs was observed by real-time cell electronic sensing assay and apoptosis rate by Annexin V/PI staining. The cell migration assay was performed with a transwell insert system. Cytoskeleton remodeling was observed by immunofluorescence assay. The content of endothelial nitric oxide synthase (eNOS) was measured by enzyme-linked immunosorbent assay. Intracellular reactive oxygen species (ROS) generation was assessed by immunofluorescence and flow cytometer. Key genes associated with the metabolism of ox-LDL were chosen for quantitative real-time polymerase chain reaction to explore the possible mechanism of TT against oxidized LDL-induced endothelial dysfunction.
RESULTSTT suppressed ox-LDL-induced HUVEC proliferation and apoptosis rates significantly (41.1% and 43.5% after treatment for 3 and 38 h, respectively; P<0.05). It also prolonged the HUVEC survival time and postponed the cell's decaying stage (from the 69th h to over 100 h). According to the immunofluorescence and transwell insert system assay, TT improved the endothelial cytoskeletal network, and vinculin expression and increased cell migration. Additionally, TT regulated of the synthesis of endothelial nitric oxide synthase and generation of intracellular reactive oxygen species (P<0.05). Both 30 and 3 μg/mL TT demonstrated similar efficacy to valsartan. TT normalized the increased mRNA expression of PI3Kα and Socs3. It also decreased mRNA expression of Akt1, AMPKα1, JAK2, LepR and STAT3 induced by ox-LDL. The most notable changes were JAK2, LepR, PI3Kα, Socs3 and STAT3.
CONCLUSIONSTT demonstrated potential lowering lipid benefits, anti-hypertension and endothelial protective effects. It also suggested that the JAK2/STAT3 and/or PI3K/AKT pathway might be a very important pathway which was involved in the pharmacological mechanism of TT as the vascular protective agent.
Apoptosis ; drug effects ; Cell Movement ; drug effects ; Cell Survival ; drug effects ; Cytoskeleton ; drug effects ; metabolism ; Endothelium, Vascular ; drug effects ; pathology ; physiopathology ; Enzyme-Linked Immunosorbent Assay ; Fluorescent Antibody Technique ; Gene Expression Regulation ; drug effects ; Human Umbilical Vein Endothelial Cells ; drug effects ; Humans ; Lipoproteins, LDL ; adverse effects ; Nitric Oxide Synthase Type III ; metabolism ; Plant Extracts ; pharmacology ; Protective Agents ; pharmacology ; Reactive Oxygen Species ; metabolism ; Tribulus ; chemistry ; Vinculin ; metabolism ; Water ; chemistry
7.Exogenous hydrogen sulfide inhibits high-glucose-induced injuries via regulating leptin/leptin receptor signaling pathway in human umbilical vein endothelial cells.
Dong-Bo WU ; ; Jing-Fu CHEN ; Qing XU ; Jia-Qiong LIN ; Jing-Qiu LIAO ; Wen WU
Journal of Southern Medical University 2016;36(8):1055-1061
OBJECTIVETo investigate whether exogenous hydrogen sulfide (H2S) inhibits the high-glucose (HG)-induced injury by modulating leptin/leptin receptor (LEPR) signal pathway in human umbilical vein endothelial cells (HUVECs).
METHODSHUVECs were treated with 40 mmol/L glucose for 3-24 h, and the cell viability was examined by CCK-8 assay. The changes of cell morphology and the number of apoptotic cells were assessed by Hoechst 33258 nuclear staining followed by photofluorography. The intracellular levels of reactive oxygen species (ROS) was detected by DCFH-DA staining followed by photofluorography. Mitochondrial membrane potential (MMP) was determined by Rhodamine 123 (Rh123) staining and photofluorography. The expression levels of leptin and LEPR protein were measured by Western blotting.
RESULTSs The expression of leptin and LERP in HUVECs began to significantly increase at 3 h after HG exposure and reached the peak levels at 9 h (P<0.01). Pretreatment of HUVECs with 400 µmol/L sodium hydrosulfide (H2S donor) for 30 min inhibited HG-induced increase in leptin and leptin receptor expressions in HUVECs (P<0.01). Pretreatment of HUVECs with 400 µmol/L NaHS for 30 min or 50 ng/mL leptin antagonists (LA) for 1 h obviously alleviated HG-induced injury by increasing cell viability, decreasing cell apoptosis and lowering accumulation of intracellular ROS and MMP loss (P<0.01).
CONCLUSIONExogenous H2S protects against HG-induced injury by inhibiting leptin/LEPR pathway in HUVECs.
Apoptosis ; Cell Survival ; Cells, Cultured ; Glucose ; adverse effects ; Human Umbilical Vein Endothelial Cells ; drug effects ; metabolism ; Humans ; Hydrogen Sulfide ; pharmacology ; Leptin ; metabolism ; Membrane Potential, Mitochondrial ; Reactive Oxygen Species ; metabolism ; Receptors, Leptin ; metabolism ; Signal Transduction
8.Arctiin ameliorates advanced oxidation protein product-induced epithelial-to- mesenchymal transition in HK-2 cells by inhibiting endoplasmic reticulum stress.
Jun ZHANG ; Li-Li HUANG ; Xiu-Jie LIANG ; Yue WANG ; Na DUAN ; Xiao-Hong XIANG ; Shuang-Shuang SHU ; Ting-Ting GUO ; Lei YANG ; Xun TANG
Journal of Southern Medical University 2016;36(6):833-837
OBJECTIVETo investigate the effect of arctiin on advanced oxidation protein product (AOPP)-induced epithelial-to-mesenchymal transition (EMT) in tubular cells and explore the mechanisms underlying this effect.
METHODSHuman proximal tubular cells (HK-2 cells) were treated with bovine serum albumin (BSA) or AOPPs in the presence or absence of arctiin. The expressions of E-cadherin, vimentin, and GRP78 at the protein and mRNA levels in the cells were examined using Western blotting and quantitative real-time PCR. The level of reactive oxygen species (ROS) was measured by flow cytometry with DCFH-DA as the fluorescent probe.
RESULTSCompared with BSA-treated cells, the cells treated with AOPPs showed decreased expression of epithelial cell marker E-cadherin and overexpression of mesenchymal marker vimentin and endoplasmic reticulum stress marker GRP78 with an increased ROS level. These changes induced by AOPPs were partly inhibited by arctiin.
CONCLUSIONArctiin can ameliorate AOPP-induced EMT in tubular cells by inhibiting endoplasmic reticulum stress, and oxidative stress response may participate in this process.
Advanced Oxidation Protein Products ; adverse effects ; Cadherins ; metabolism ; Cell Line ; Endoplasmic Reticulum Stress ; Epithelial Cells ; cytology ; drug effects ; Epithelial-Mesenchymal Transition ; Furans ; pharmacology ; Glucosides ; pharmacology ; Heat-Shock Proteins ; metabolism ; Humans ; Kidney Tubules ; cytology ; drug effects ; Oxidative Stress ; Reactive Oxygen Species ; metabolism ; Vimentin ; metabolism
9.Food-advanced glycation end products aggravate the diabetic vascular complications via modulating the AGEs/RAGE pathway.
Xing LV ; Gao-Hong LV ; Guo-Ying DAI ; Hong-Mei SUN ; Hui-Qin XU
Chinese Journal of Natural Medicines (English Ed.) 2016;14(11):844-855
The aim of this study was to investigate the effects of high-advanced glycation end products (AGEs) diet on diabetic vascular complications. The Streptozocin (STZ)-induced diabetic mice were fed with high-AGEs diet. Diabetic characteristics, indicators of renal and cardiovascular functions, and pathohistology of pancreas, heart and renal were evaluated. AGEs/RAGE/ROS pathway parameters were determined. During the experiments, the diabetic mice exhibited typical characteristics including weight loss, polydipsia, polyphagia, polyuria, high-blood glucose, and low-serum insulin levels. However, high-AGEs diet effectively aggravated these diabetic characteristics. It also increased the 24-h urine protein levels, serum levels of urea nitrogen, creatinine, c-reactive protein (CRP), low density lipoprotein (LDL), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) in the diabetic mice. High-AGEs diet deteriorated the histology of pancreas, heart, and kidneys, and caused structural alterations of endothelial cells, mesangial cells and podocytes in renal cortex. Eventually, high-AGEs diet contributed to the high-AGE levels in serum and kidneys, high-levels of reactive oxygen species (ROS) and low-levels of superoxide dismutase (SOD) in serum, heart, and kidneys. It also upregulated RAGE mRNA and protein expression in heart and kidneys. Our results showed that high-AGEs diet deteriorated vascular complications in the diabetic mice. The activation of AGEs/RAGE/ROS pathway may be involved in the pathogenesis of vascular complications in diabetes.
Animals
;
Diabetes Mellitus, Experimental
;
complications
;
metabolism
;
Diabetic Angiopathies
;
genetics
;
metabolism
;
Diet
;
adverse effects
;
Glycation End Products, Advanced
;
metabolism
;
Humans
;
Interleukin-6
;
metabolism
;
Kidney
;
metabolism
;
Male
;
Mice
;
Mice, Inbred C57BL
;
Oxidative Stress
;
Pancreas
;
metabolism
;
Reactive Oxygen Species
;
metabolism
;
Receptor for Advanced Glycation End Products
;
genetics
;
metabolism
;
Superoxide Dismutase
;
metabolism
;
Tumor Necrosis Factor-alpha
;
metabolism
10.Evidence for beneficial effects of vitamin E.
The Korean Journal of Internal Medicine 2015;30(5):571-579
Oxidative stress and lipid peroxidation have been implicated in the pathogenesis of various diseases, including atherosclerosis and fatty liver diseases, and consequently the role of antioxidants in the prevention and treatment of such diseases has received much attention. In particular, the effects of vitamin E, the most important lipophilic radical-scavenging antioxidant, have been investigated extensively. Many in vitro, animal, and epidemiological studies have reported positive results, but large-scale randomized controlled intervention studies and meta-analyses have produced inconsistent and often disappointing results. In the present review article, the role and action of vitamin E are discussed, with consideration of the factors that determine the outcome of vitamin E treatment. Vitamin E should benefit subjects experiencing oxidative stress due to free radicals when administered at the correct time and for an appropriate duration.
Animals
;
Antioxidants/adverse effects/*therapeutic use
;
Humans
;
Lipid Peroxidation/*drug effects
;
Oxidative Stress/*drug effects
;
Reactive Oxygen Species/*metabolism
;
Vitamin E/adverse effects/*therapeutic use

Result Analysis
Print
Save
E-mail