1.Clematichinenoside AR protects bone marrow mesenchymal stem cells from hypoxia-induced apoptosis by maintaining mitochondrial homeostasis.
Zi-Tong ZHAO ; Peng-Cheng TU ; Xiao-Xian SUN ; Ya-Lan PAN ; Yang GUO ; Li-Ning WANG ; Yong MA
China Journal of Chinese Materia Medica 2025;50(5):1331-1339
This study aims to elucidate the role and mechanism of clematichinenoside AR(CAR) in protecting bone marrow mesenchymal stem cells(BMSCs) from hypoxia-induced apoptosis. BMSCs were isolated by the bone fragment method and identified by flow cytometry. Cells were cultured under normal conditions(37℃, 5% CO_2) and hypoxic conditions(37℃, 90% N_2, 5% CO_2) and treated with CAR. The BMSCs were classified into eight groups: control(normal conditions), CAR(normal conditions + CAR), hypoxia 24 h, hypoxia 24 h + CAR, hypoxia 48 h, hypoxia 48 h + CAR, hypoxia 72 h, and hypoxia 72 h + CAR. The cell counting kit-8(CCK-8) assay and terminal-deoxynucleoitidyl transferase mediated nick end labeling(TUNEL) were employed to measure cell proliferation and apoptosis, respectively. The number of mitochondria and mitochondrial membrane potential were measured by MitoTracker®Red CM-H2XRo staining and JC-1 staining, respectively. The level of reactive oxygen species(ROS) was measured with the DCFH-DA fluorescence probe. The protein levels of B-cell lymphoma-2 associated X protein(BAX), caspase-3, and optic atrophy 1(OPA1) were determined by Western blot. The results demonstrated that CAR significantly increased cell proliferation. Compared with the control group, the hypoxia groups showed increased apoptosis rates, reduced mitochondria, elevated ROS levels, decreased mitochondrial membrane potential, upregulated expression of BAX and caspase-3, and downregulated expression of OPA1. In comparison to the corresponding hypoxia groups, CAR intervention significantly decreased the apoptosis rate, increased mitochondria, reduced ROS levels, elevated mitochondrial membrane potential, downregulated the expression of BAX and caspase-3, and upregulated the expression of OPA1. Therefore, it can be concluded that CAR may exert an anti-apoptotic effect on BMSCs under hypoxic conditions by regulating OPA1 to maintain mitochondrial homeostasis.
Mesenchymal Stem Cells/metabolism*
;
Apoptosis/drug effects*
;
Mitochondria/metabolism*
;
Animals
;
Rats
;
Cell Hypoxia/drug effects*
;
Homeostasis/drug effects*
;
Reactive Oxygen Species/metabolism*
;
Rats, Sprague-Dawley
;
Membrane Potential, Mitochondrial/drug effects*
;
Saponins/pharmacology*
;
Caspase 3/genetics*
;
Male
;
bcl-2-Associated X Protein/genetics*
;
Bone Marrow Cells/metabolism*
;
Cell Proliferation/drug effects*
;
Protective Agents/pharmacology*
;
Cells, Cultured
2.Alleviation of hypoxia/reoxygenation injury in HL-1 cells by ginsenoside Rg_1 via regulating mitochondrial fusion based on Notch1 signaling pathway.
Hui-Yu ZHANG ; Xiao-Shan CUI ; Yuan-Yuan CHEN ; Gao-Jie XIN ; Ce CAO ; Zi-Xin LIU ; Shu-Juan XU ; Jia-Ming GAO ; Hao GUO ; Jian-Hua FU
China Journal of Chinese Materia Medica 2025;50(10):2711-2718
This paper explored the specific mechanism of ginsenoside Rg_1 in regulating mitochondrial fusion through the neurogenic gene Notch homologous protein 1(Notch1) pathway to alleviate hypoxia/reoxygenation(H/R) injury in HL-1 cells. The relative viability of HL-1 cells after six hours of hypoxia and two hours of reoxygenation was detected by cell counting kit-8(CCK-8). The lactate dehydrogenase(LDH) activity in the cell supernatant was detected by the lactate substrate method. The content of adenosine triphosphate(ATP) was detected by the luciferin method. Fluorescence probes were used to detect intracellular reactive oxygen species(Cyto-ROS) levels and mitochondrial membrane potential(ΔΨ_m). Mito-Tracker and Actin were co-imaged to detect the number of mitochondria in cells. Fluorescence quantitative polymerase chain reaction and Western blot were used to detect the mRNA and protein expression levels of Notch1, mitochondrial fusion protein 2(Mfn2), and mitochondrial fusion protein 1(Mfn1). The results showed that compared with that of the control group, the cell activity of the model group decreased, and the LDH released into the cell culture supernatant increased. The level of Cyto-ROS increased, and the content of ATP decreased. Compared with that of the model group, the cell activity of the ginsenoside Rg_1 group increased, and the LDH released into the cell culture supernatant decreased. The level of Cyto-ROS decreased, and the ATP content increased. Ginsenoside Rg_1 elevated ΔΨ_m and increased mitochondrial quantity in HL-1 cells with H/R injury and had good protection for mitochondria. After H/R injury, the mRNA and protein expression levels of Notch1 and Mfn1 decreased, while the mRNA and protein expression levels of Mfn2 increased. Ginsenoside Rg_1 increased the mRNA and protein levels of Notch1 and Mfn1, and decreased the mRNA and protein levels of Mfn2. Silencing Notch1 inhibited the action of ginsenoside Rg_1, decreased the mRNA and protein levels of Notch1 and Mfn1, and increased the mRNA and protein levels of Mfn2. In summary, ginsenoside Rg_1 regulated mitochondrial fusion through the Notch1 pathway to alleviate H/R injury in HL-1 cells.
Ginsenosides/pharmacology*
;
Receptor, Notch1/genetics*
;
Signal Transduction/drug effects*
;
Mice
;
Animals
;
Mitochondrial Dynamics/drug effects*
;
Mitochondria/metabolism*
;
Cell Line
;
Reactive Oxygen Species/metabolism*
;
Oxygen/metabolism*
;
Cell Hypoxia/drug effects*
;
Cell Survival/drug effects*
;
Membrane Potential, Mitochondrial/drug effects*
;
Humans
3.Protective mechanism of Chaihu Shugan San against CORT-induced damage in PC12 cells based on mitochondrial dynamics.
Ling-Yuan ZHANG ; Qi-Qi ZHENG ; Jia-Li SHI ; Pei-Fang WANG ; Jia-Li LU ; Jian-Ying SHEN
China Journal of Chinese Materia Medica 2025;50(16):4546-4554
In this report, the protective effect and molecular mechanism of Chaihu Shugan San-containing serum on corticosterone(CORT)-induced mitochondrial damage in pheochromocytoma(PC12) cells was studied based on CORT-induced rat PC12 cell model. The cultured cells were divided into five groups: blank control group, CORT group(400 μmol·L~(-1) CORT), Chaihu Shugan San-containing serum group(400 μmol·L~(-1) CORT + 10% Chaihu Shugan San-containing serum), control serum group(400 μmol·L~(-1) CORT + 10% control serum), and fluoxetine group(400 μmol·L~(-1) CORT + 10% fluoxetine-containing serum). The study was carried out by cell activity detection, mitochondrial morphology observation, membrane potential measurement, energy metabolism analysis, and mitochondrial dynamics-related protein detection. The results showed that CORT treatment significantly reduced the survival rate of PC12 cells, altered mitochondrial morphology, and decreased mitochondrial membrane potential and adenosine triphosphate(ATP) synthetic rate. Both Chaihu Shugan San-and fluoxetine-containing serum significantly increased the survival rate of CORT-treated PC12 cells and the ATP synthetic rate in the mitochondria. Unlike fluoxetine, Chaihu Shugan San-containing serum significantly inhibited the decrease in mitochondrial membrane potential caused by CORT and increased the oxygen consumption rate(OCR) values of both mitochondrial maximum respiration and reserve respiration capacity. Western blot analysis showed that CORT induced upregulated protein expressions of dynamin-related protein 1(Drp1) and peroxisome proliferator-activated receptor gamma co-activator 1α(PGC-1α) in PC12 cells and specific protein expression of optic atrophy protein 1(OPA1), yet it repressed the protein expressions of silent information regulator 1(SIRT1) and mitochondrial fusion protein 1(Mfn1) in PC12 cells. Both Chaihu Shugan San-and fluoxetine-containing serum significantly inhibited the protein expression of Drp1. However, only Chaihu Shugan San-containing serum could significantly inhibit the CORT-induced upregulation protein of PGC-1α. RESULTS:: herein suggest that Chaihu Shugan San-containing serum can alleviate CORT-induced damage in PC12 cells, which may be related to the mitochondrial fragmentation/lipid peroxidation protection by Drp1 inhibition, as well as mitochondrial dynamics and energy metabolism mediated by PGC-1α/SIRT1 signaling pathway.
Animals
;
PC12 Cells
;
Rats
;
Mitochondrial Dynamics/drug effects*
;
Mitochondria/metabolism*
;
Corticosterone/adverse effects*
;
Membrane Potential, Mitochondrial/drug effects*
;
Drugs, Chinese Herbal/pharmacology*
;
Protective Agents/pharmacology*
;
Cell Survival/drug effects*
4.In vitro effects of antidepressants on human sperm function.
Rita António SANTOS ; Ana Paula SOUSA ; Teresa ALMEIDA-SANTOS ; João RAMALHO-SANTOS ; Renata Santos TAVARES
Asian Journal of Andrology 2025;27(1):30-36
Depression currently affects about 280 million people worldwide and its prevalence has been increasing dramatically, especially among the young and people of reproductive age, which consequently leads to an increase in antidepressant consumption. Antidepressants are associated with sexual dysfunction in both men and women; however, their role in male fertility has been scarcely studied. Fluoxetine and sertraline, two serotonin reuptake inhibitors (SSRIs), are among the most prescribed antidepressants worldwide. To determine their possible effects, human sperm cells were exposed to either sertraline or fluoxetine at concentrations previously found in blood and seminal fluid of patients undergoing treatment. Spermatozoa were incubated for up to 24 h at 37°C and 5% CO 2 , and important functional parameters such as sperm motility, viability, mitochondrial membrane potential, cellular reactive oxygen species (ROS) production, chromatin/DNA integrity, acrosome status, and tyrosine phosphorylation were assessed. At low levels, fluoxetine consistently decreased progressive motility throughout time while promoting fluctuations in ROS levels and sperm capacitation. Nevertheless, it did not affect viability, mitochondrial membrane potential, acrosome reaction nor chromatin/DNA integrity. Sertraline, on the other hand, had little to nonsignificant impact at low doses, but affected almost all tested parameters at supratherapeutic concentrations. Altogether, our results suggest that both antidepressants may impair sperm function, possibly through different mechanisms of action, but fluoxetine is the only exhibiting mild negative effects at doses found in vivo .
Humans
;
Male
;
Spermatozoa/drug effects*
;
Fluoxetine/pharmacology*
;
Sperm Motility/drug effects*
;
Sertraline/pharmacology*
;
Reactive Oxygen Species/metabolism*
;
Antidepressive Agents/pharmacology*
;
Membrane Potential, Mitochondrial/drug effects*
;
Sperm Capacitation/drug effects*
;
Selective Serotonin Reuptake Inhibitors/pharmacology*
;
Cell Survival/drug effects*
;
Acrosome Reaction/drug effects*
5.Triclocarban impacts human sperm motility by inhibiting glycolysis and oxidative phosphorylation.
Long-Long FU ; Wei-Zhou WANG ; Yan FENG ; Fu CHEN ; Bin LIU ; Liang HUANG ; Lin-Yuan ZHANG ; Lei CHEN
Asian Journal of Andrology 2025;27(6):707-713
Triclocarban (TCC) is a broad-spectrum antimicrobial widely used in various personal care products, textiles, and children's toys. TCC has potential reproductive and developmental toxicity in animals. However, little is known regarding the effect of TCC on human sperm function. In this study, an in vitro assay was used to investigate the effects of TCC on normal human spermatozoa and the possible underlying mechanisms involved. Semen from healthy male donors was collected and cultured in complete Biggers, Whitten and Whittingham (BWW) and low-sugar BWW media, followed by treatment with TCC at concentrations of 0, 0.1 µmol l -1 , 1 µmol l -1 , 10 µmol l -1 , and 100 µmol l -1 for 4 h. TCC was found to reduce the sperm total motility and progressive motility. Moreover, the sperm kinematic parameters, straight-line velocity (VSL), average path velocity (VAP), and curvilinear velocity (VCL) were affected in a dose-dependent manner. After treatment with TCC at the lowest effective concentration of 10 µmol l -1 , TCC caused a significant decrease in mitochondrial adenosine triphosphate (ATP) production and mitochondrial membrane potential (MMP) and a significant increase in reactive oxygen species (ROS), similar to the observations with the positive control carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone (FCCP), suggesting that TCC may decrease sperm motility by affecting the oxidative phosphorylation (OXPHOS) pathway. In a sugar-free and low-sugar BWW culture environment, TCC enhanced the damaging effect on sperm motility and ATP, MMP, and lactate decreased significantly, suggesting that TCC may also affect the glycolytic pathway that supplies energy to spermatozoa. This study demonstrates a possible mechanism of TCC toxicity in spermatozoa involving both the OXPHOS and glycolysis pathways.
Male
;
Sperm Motility/drug effects*
;
Humans
;
Carbanilides/pharmacology*
;
Oxidative Phosphorylation/drug effects*
;
Glycolysis/drug effects*
;
Membrane Potential, Mitochondrial/drug effects*
;
Adenosine Triphosphate/metabolism*
;
Spermatozoa/metabolism*
;
Reactive Oxygen Species/metabolism*
;
Mitochondria/metabolism*
6.Hydroxysafflor Yellow A Ameliorates the Replicative Senescence of Human Umbilical Cord Mesenchymal Stem Cells by Suppressing Oxidative Stress.
Si-Yun WANG ; Qi ZHU ; Chun-Xia TAN ; Fang LU ; Tao LU
Journal of Experimental Hematology 2025;33(5):1507-1515
OBJECTIVE:
To investigate the effects and mechanisms of hydroxysafflor yellow A (HSYA) on replicative senescence in human umbilical cord mesenchymal stem cells (hUC-MSCs).
METHODS:
hUC-MSCs were cultured to construct a replicative senescence model through continuous amplification in vitro. Cells at passage 2 served as the control group, while cells at passage 10 were designated as the senescence group. The senescent cells were cultured in a culture medium containing HSYA. Cell viability was detected by the CCK-8 assay, and cell confluence was analyzed using the Incucyte S3 live-cell analysis system. The optimal concentration and time point were determined and utilized for subsequent experiments. Senescent cells were pretreated with 0.01 mg/ml HSYA, and the proportion of senescence-associated β-galactosidase (SA-β-gal) positive cells was detected to assess the senescence state. The relative telomere length was detected by qPCR. Reactive oxygen species (ROS) levels were measured using the fluorescent probe DCFH-DA. Mitochondrial membrane potential was assessed by JC-1 staining. The expression of p53, p16, p21, OCT4, and SOX2 genes was detected by qPCR. The expression of p16, p53, OCT4, and SOX2 proteins was analyzed by Western blot.
RESULTS:
HSYA significantly decreased the SA-β-gal positive staining rate, inhibited telomere attrition, reduced the ROS accumulation, increased mitochondrial membrane potential in senescent cells. Additionally, HSYA downregulated the expression of p53 and p16, and upregulated the expression of OCT4. HSYA decreased p16 protein level and increased OCT4 and SOX2 protein levels.
CONCLUSION
HSYA may ameliorate replicative senescence in hUC-MSCs by modulating the p53 and p16 signaling pathways and suppressing oxidative stress.
Humans
;
Mesenchymal Stem Cells/drug effects*
;
Cellular Senescence/drug effects*
;
Chalcone/pharmacology*
;
Oxidative Stress/drug effects*
;
Quinones/pharmacology*
;
Umbilical Cord/cytology*
;
Reactive Oxygen Species/metabolism*
;
Cells, Cultured
;
Cyclin-Dependent Kinase Inhibitor p16/metabolism*
;
Tumor Suppressor Protein p53/metabolism*
;
Membrane Potential, Mitochondrial
;
Cell Proliferation
7.Chrysophanol Induces Cell Death and Inhibits Invasiveness through Alteration of Calcium Levels in HepG2 Human Liver Cancer Cells.
Shu-Chao CHEN ; Qiao-Wen CHEN ; Chih-Yuan KO
Chinese journal of integrative medicine 2025;31(5):434-440
OBJECTIVE:
To investigate the effect of chrysophanol, a phytochemical derived from Radix et Rhizoma Rhei on HepG2 liver cancer cells.
METHODS:
HepG2 cell line was treated with different concentrations chrysophanol (0-100 μmol/L) for 24 h. The cell counting kit 8 assay was employed to assess cell viability. Intracellular calcium levels were examined using Fluo-4 AM and Mag-fluo-4 AM staining, followed by flow cytometry analysis. Mitochondrial membrane potential was measured with JC-1 assay kit. Additionally, the expressions of key proteins such as p-JNK, Bax, cytochrome c (Cyt C), cleaved caspase-3 (cCaspase-3), and caspase-8 were analyzed by Western blot. The inhibitory effects of chrysophanol on the invasion of cells were determined using a Transwell assay. Analysis of invasiveness was conducted by wound healing assay.
RESULTS:
Chrysophanol significantly reduced the proliferation of HepG2 liver cancer cells by affecting intracellular calcium distribution, diminishing mitochondrial membrane potential, and enhancing the expressions of p-JNK, Bax, Cyt C, cCaspase-3, and caspase-8 in the groups treated with 75 or 100 μmol/L chrysophanol compared to the control group (P<0.05). Additionally, 75 and 100 μmol/L chrysophanol exhibited inhibitory effects on cell migration and wound healing.
CONCLUSION
Chrysophanol demonstrates potential against HepG2 liver cancer cells, suggesting its potential use as a therapeutic agent for liver cancer treatment.
Humans
;
Calcium/metabolism*
;
Hep G2 Cells
;
Liver Neoplasms/metabolism*
;
Neoplasm Invasiveness
;
Membrane Potential, Mitochondrial/drug effects*
;
Anthraquinones/pharmacology*
;
Cell Proliferation/drug effects*
;
Cell Death/drug effects*
;
Apoptosis/drug effects*
;
Cell Movement/drug effects*
;
Cell Survival/drug effects*
8.Dexmedetomidine attenuates heat stress-induced oncosis in human skeletal muscle cells by activating the Nrf2/Ho-1 pathway.
Yang LIU ; Yiqing JIA ; Chengcheng LI ; Handing MAO ; Shuyuan LIU ; Yi SHAN
Journal of Southern Medical University 2025;45(3):603-613
OBJECTIVES:
To investigate the protective effects of dexmedetomidine (DEX) against heat stress (HS)-induced oncosis in human skeletal muscle cells (HSKMCs) and its underlying mechanisms.
METHODS:
A HSKMC model of HS-induced oncosis were established by 43 ℃ water bath for 4 h, and the effects of treatments with 30 μmol/L DEX, ML385 (a Nrf2 inhibitor) +DEX, si-Nrf2+HS, and si-Nrf2+DEX prior to modeling on cell viability was assessed using CCK-8 assay. Oncosis characteristics were evaluated using transmission electron microscopy and Annexin V-FITC/PI flow cytometry. The oxidative stress markers (GSH, GSH-Px, MDA, SOD and ROS), mitochondrial membrane potential, energy metabolism, and inflammatory cytokines (TNF-α, IL-6 and IL-1β) in the cells were quantified using standard kits, and the expressions of porimin, caspase-3 and Nrf2 pathway proteins were analyzed using Western blotting and qRT-PCR.
RESULTS:
HS induced typical oncotic features in HSKMCs including organelle swelling and cytoplasmic vacuolization. DEX pretreatment significantly attenuated these changes, reduced Annexin V+/PI+ cell ratio and cellular porimin expression, and lowered the levels of ROS and MDA while restoring GSH and SOD levels. DEX pretreatment also significantly increased the mitochondrial membrane potential and ATP level, upregulated the expressions of Nrf2, p-Nrf2, HO-1 and NQO1, and suppressed the expressions of TNF-α, IL-6 and IL-1β. The protective effects of DEX were obviously attenuated by interventions with ML385 or si-Nrf2.
CONCLUSIONS
DEX mitigates HS-induced HSKMC oncosis by activating the Nrf2/HO-1 pathway to relieve oxidative stress, mitochondrial dysfunction, and inflammatory responses.
Humans
;
Dexmedetomidine/pharmacology*
;
NF-E2-Related Factor 2/metabolism*
;
Oxidative Stress/drug effects*
;
Heat-Shock Response/drug effects*
;
Signal Transduction/drug effects*
;
Membrane Potential, Mitochondrial
;
Muscle, Skeletal/cytology*
;
Heme Oxygenase-1/metabolism*
;
Apoptosis/drug effects*
9.Astragaloside IV alleviates D-GAL-induced endothelial cell senescence by promoting mitochondrial autophagy via inhibiting the PINK1/Parkin pathway.
Ming YI ; Ye LUO ; Lu WU ; Zeheng WU ; Cuiping JIANG ; Shiyu CHEN ; Xiao KE
Journal of Southern Medical University 2025;45(11):2427-2437
OBJECTIVES:
To explore the mechanism by which astragaloside IV (AS-IV) alleviates D-galactose (D-GAL)-induced senescence in human umbilical vein endothelial cells (HUVECs).
METHODS:
Cultured HUVECs were treated with D-GAL (40 g/L), AS-IV (200 μmol/L), D-GAL+AS-IV, or D-GAL+AS-IV+MTK458 (a mitochondrial autophagy agonist, 25 μmol/L) for 48 h, and the changes in cell proliferation, migration, and angiogenesis capacity were evaluated. Cell apoptosis, reactive oxygen species (ROS) levels, mitochondrial membrane potential, and expressions of autophagy-related proteins (LC3-II/LC3-I) and PINK1/Parkin pathway proteins in the treated cells were detected.
RESULTS:
AS-IV treatment significantly reduced the inhibitory effect of D-GAL on HUVEC viability, effectively alleviated D-GAL-induced impairment of tube-forming ability, and promoted angiogenesis and migration ability of the cells. AS-IV also significantly reduced the rate of D-GAL-induced HUVECs positive for senescence-associated β-galactosidase (SA-β-Gal) staining and inhibited the expression of senescence-related genes P21 and P53. AS-IV restored mitochondrial membrane potential and reduced intracellular ROS levels in D-GAL-induced HUVECs, and inhibited the fusion of autophagosomes and lysosomes to prevent the completion of autophagic flux. In HUVECs treated with both D-GAL and AS-IV, the application MTK458 significantly increased the number of yellow spots and enhanced the expressions of P21, P53, PINK1, Parkin, LC3, and Beclin proteins.
CONCLUSIONS
AS-IV alleviates D-GAL-induced endothelial cell senescence by inhibiting the PINK1/Parkin pathway to regulate mitochondrial autophagy.
Humans
;
Human Umbilical Vein Endothelial Cells/drug effects*
;
Cellular Senescence/drug effects*
;
Autophagy/drug effects*
;
Saponins/pharmacology*
;
Ubiquitin-Protein Ligases/metabolism*
;
Mitochondria/drug effects*
;
Triterpenes/pharmacology*
;
Protein Kinases/metabolism*
;
Galactose/pharmacology*
;
Reactive Oxygen Species/metabolism*
;
Signal Transduction/drug effects*
;
Cells, Cultured
;
Apoptosis/drug effects*
;
Membrane Potential, Mitochondrial
;
Cell Proliferation/drug effects*
10.Inhibition of miR-30d-5p promotes mitochondrial autophagy and alleviates high glucose-induced injury in podocytes.
Ying CAI ; Sheng CHEN ; Xiaoli JIANG ; Qiyuan WU ; Bei GUO ; Fang WANG
Journal of Zhejiang University. Medical sciences 2024;53(6):756-764
OBJECTIVES:
To study the role of microRNA (miR)-30d-5p in high glucose-induced podocyte injury.
METHODS:
Podocytes were hyperglycated with 30 mmol/L glucose, transfected with miR-30d-5p inhibitor and mimic, and then treated with 1 mg/mL 3-methyladenine (3-MA). The transfection efficiency of miR-30d-5p was quantified by reverse transcription PCR. Apoptosis was detected by flow cytometry. The expressions of nephrin, microtubule-associated protein light chain (LC) 3Ⅱ/LC3Ⅰ, P62, autophagy-related gene (ATG) 5, PTEN induced putative kinase (PINK) 1 and Parkin gene (PARK2) were detected by Western blotting. The mito-chondrial membrane potential was detected by JC-1 fluorescent probe, and adenosine triphosphate (ATP) content in cells was detected by relevant kits.
RESULTS:
Under high glucose induction, podocyte apoptosis increased, miR-30d-5p and P62 expressions were upregulated, while nephrin, ATG5, PINK1, PARK2 and LC3Ⅱ/LC3Ⅰ expressions decreased (all P<0.01). MiR-30d-5p inhibitor reversed the effect of high glucose on apoptosis, and the expression of ATG5, PINK1, PARK2, nephrin, LC3Ⅱ/LC3Ⅰ and P62 (all P<0.01). High glucose induced loss of mitochondrial membrane potential and ATP content in podocytes, while inhibition of miR-30d-5p increased them. Autophagy inhibitors 3-MA and miR-30d-5p mimics reversed the effects of miR-30d-5p inhibition on apoptosis, autophagy and mitochondrial function of podocytes induced by high glucose (all P<0.05).
CONCLUSIONS
Inhibition of miR-30d-5p may promote mitochondrial autophagy (mitophagy) by promoting the expression of ATG5, PINK1, PARK2 and alleviating high glucose-induced podocyte damage.
Podocytes/drug effects*
;
MicroRNAs/metabolism*
;
Glucose/adverse effects*
;
Autophagy/drug effects*
;
Animals
;
Mice
;
Autophagy-Related Protein 5/genetics*
;
Apoptosis/drug effects*
;
Mitochondria/metabolism*
;
Ubiquitin-Protein Ligases/genetics*
;
Membrane Proteins/genetics*
;
Membrane Potential, Mitochondrial/drug effects*
;
Microtubule-Associated Proteins/genetics*
;
Protein Kinases

Result Analysis
Print
Save
E-mail