1.Mechanism of L-perilla alcohol in intervening hypoxic pulmonary hypertension based on network pharmacology and experimental verification.
Yu-Rong WANG ; Yang YU ; Zhuo-Sen LIANG ; Li TONG ; Dian-Xiang LU ; Xing-Mei NAN
China Journal of Chinese Materia Medica 2025;50(1):209-217
The mechanism of L-perilla alcohol(L-POH) in intervening hypoxic pulmonary hypertension(HPAH) was discussed based on network pharmacology, and experimental verification. The active components and potential targets of the volatile oil of Rhodiola tangutica(VORA) in the intervention of HPAH were screened by network pharmacology. The biological process of Gene Ontology(GO) and the signaling pathway enrichment of Kyoto Encyclopedia of Genes and Genomes(KEGG) were analyzed for the core targets, and a "component-common target-disease" network was constructed. Four active components were screened from VORA: L-POH, linalool, geraniol, and(-)-myrtenol. The core targets for treating HPAH were HSP90AA1, AKT1, ESR1, PIK3CA, EP300, EGFR, and JAK2. GO enrichment analysis mainly involved biological processes such as reaction to hypoxia, heme binding, and steroid binding. KEGG enrichment analysis mainly involved hypoxia-inducing factor 1(HIF-1) signaling pathway, phosphatidylinositol 3-kinase/protein kinase B(PI3K/AKT) signaling pathway, and Janus kinase/activator of signal transduction and transcription(JAK/STAT) signaling pathway. The vasodilation effects of the four active components were screened by perfusion experiment of extracorporeal vascular rings, and the mechanism of the main active component L-POH was studied by channel blockers. The inhibitory effects of the four active components on the proliferation of pulmonary artery smooth muscle cells(PASMCs) induced by hypoxia were screened by cell proliferation experiment, and the mechanism of the main active component L-POH was studied by flow cytometry, cell cycle experiment, and Western blot. The results showed that L-POH could directly act on vascular smooth muscle to relax pulmonary arterioles, induce ATP-sensitive potassium channels to open, and inhibit extracellular Ca~(2+) influx through voltage-gated calcium channels to relax blood vessels. In addition, L-POH could inhibit the abnormal proliferation of PASMCs induced by hypoxia and promote its apoptosis, and its mechanism may be related to the increase in Bax protein expression and the decrease in p-JAK2, p-STAT3, Bcl-2, and cyclinA2 protein expression. In summary, L-POH can interfere with HPAH by relaxing pulmonary arterioles and inhibiting the proliferation of smooth muscle cells.
Network Pharmacology
;
Animals
;
Hypertension, Pulmonary/physiopathology*
;
Drugs, Chinese Herbal/administration & dosage*
;
Rats
;
Hypoxia/metabolism*
;
Rhodiola/chemistry*
;
Signal Transduction/drug effects*
;
Humans
;
Monoterpenes/chemistry*
;
Male
;
Cell Proliferation/drug effects*
;
Rats, Sprague-Dawley
2.Effect of total secondary ginsenosides on apoptosis and energy metabolism of H9c2 cells under hypoxia based on mitochondrial biogenesis.
Zhong-Jie YUAN ; Yue XIAO ; Zhen LIU ; Ai-Qun ZHANG ; Bin LI ; Shang-Xian GAO
China Journal of Chinese Materia Medica 2025;50(5):1255-1266
This study explores the effect of total secondary ginsenosides(TSG) on apoptosis and energy metabolism in H9c2 cells under hypoxia and its potential mechanisms. H9c2 cell viability was observed and the apoptosis rate was calculated to determine suitable intervention concentrations of TSG, antimycin A complex(AMA), and coenzyme Q10(CoQ10), along with the duration of hypoxia. H9c2 cells at the logarithmic phase were divided into a normal group, a model group, a TSG group, an AMA group, a TSG+AMA group, and a CoQ10 group. All groups, except the normal group, were treated with their respective intervention drugs and cultured under hypoxic conditions. Adenosine triphosphate(ATP) content and creatine kinase(CK) activity were measured using an ATP chemiluminescence assay kit and a CK colorimetric assay kit. Flow cytometry was used to assess apoptosis rates, and Western blot evaluated the expression levels of apoptosis-related proteins, including B-cell lymphoma 2(Bcl-2), Bcl-2-associated X protein(Bax), cysteinyl aspartate-specific protease(caspase)-3, caspase-8, and caspase-9, as well as mitochondrial biogenesis-related proteins peroxisome proliferator-activated receptor-γ coactivator 1α(PGC-1α), estrogen-related receptor-α(ERRα), nuclear respiratory factor(NRF)-1, NRF-2, peroxisome proliferator activated receptor-α(PPARα), and Na~+-K~+-ATPase. RT-PCR was employed to analyze the mRNA expression of mitochondrial biogenesis factors, including PGC-1α, ERRα, NRF-1, NRF-2, PPARα, mitochondrial transcription factor A(TFAM), mitochondrial cytochrome C oxidase 1(COX1), and mitochondrial NADH dehydrogenase subunit 1(ND1), ND2. The selected intervention concentrations were 7.5 μg·mL~(-1) for TSG, 10 μmol·L~(-1) for AMA, and 1×10~(-4) mol·L~(-1) for CoQ10, with a hypoxia duration of 6 h. Compared with the normal group, the model group showed decreased ATP content and CK activity, increased apoptosis rates, decreased Bcl-2 expression, and increased Bax, caspase-3, caspase-8, and caspase-9 expression in H9c2 cells. Additionally, the protein and mRNA expression levels of mitochondrial biogenesis-related factors(PGC-1α, ERRα, NRF-1, NRF-2, PPARα), mRNA expression of TFAM, COX1, and ND1, ND2, and protein expression of Na~+-K~+-ATPase in mitochondrial DNA, were also reduced. In the TSG and CoQ10 groups, ATP content and CK activity increased, and apoptosis rates decreased compared with those in the model group. The TSG group showed decreased protein expression of apoptosis-related proteins Bax, caspase-3, caspase-8, and caspase-9, increased protein and mRNA expression of mitochondrial biogenesis factors PGC-1α, ERRα, NRF-1, and PPARα, and increased NRF-2 protein expression and TFAM mRNA expression in mitochondrial DNA. Conversely, in the AMA group, ATP content and CK activity decreased, the apoptosis rate increased, Bcl-2 expression decreased, and Bax, caspase-3, caspase-8, and caspase-9 expression increased, alongside reductions in PGC-1α, ERRα, NRF-1, NRF-2, PPARα protein and mRNA expression, as well as TFAM, COX1, ND1, ND2 mRNA expression and Na~+-K~+-ATPase protein expression. Compared with the TSG group, the TSG+AMA group exhibited decreased ATP content and CK activity, increased apoptosis rates, decreased Bcl-2 expression, and increased Bax, caspase-3, caspase-8, and caspase-9 expression, along with decreased PGC-1α, ERRα, NRF-1, NRF-2, and PPARα protein and mRNA expression and TFAM, COX1, and ND1, ND2 mRNA expression. Compared with the AMA group, the TSG+AMA group showed increased CK activity, decreased apoptosis rate, increased Bcl-2 expression, and decreased Bax, caspase-8, and caspase-9 expression. Additionally, the protein and mRNA expression of PGC-1α, ERRα, NRF-1, PPARα, mRNA expression of TFAM, COX1, ND1, ND2, and Na~+-K~+-ATPase protein expression increased. In conclusion, TSG enhance ATP content and CK activity and inhibit apoptosis in H9c2 cells under hypoxia, and the mechanisms may be related to the regulation of PGC-1α, ERRα, NRF-1, NRF-2, PPARα, and TFAM expression, thus promoting mitochondrial biogenesis.
Apoptosis/drug effects*
;
Ginsenosides/pharmacology*
;
Energy Metabolism/drug effects*
;
Mitochondria/metabolism*
;
Animals
;
Rats
;
Cell Line
;
Cell Hypoxia/drug effects*
;
Organelle Biogenesis
;
Adenosine Triphosphate/metabolism*
;
Humans
;
Cell Survival/drug effects*
3.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
4.Astragali Radix-Curcumae Rhizoma drug pair inhibits growth of osteosarcoma by affecting cell adhesion and angiogenesis via PI3K/Akt/HIF-1α pathway.
Dao-Tong YUAN ; Zhi-Meng ZHANG ; Rui GONG ; Xi-Min JIN ; Can-Ran WANG ; Jie ZHAO
China Journal of Chinese Materia Medica 2025;50(8):2217-2228
This study aims to investigate the optimal ratio of Astragali Radix-Curcumae Rhizoma(AC) for inhibiting the proliferation of 143B osteosarcoma cells, and to investigate the mechanism by which AC inhibits osteosarcoma growth and metastasis through angiogenesis and cell adhesion mediated by the phosphatidylinositol 3-kinase(PI3K)/protein kinase B(Akt)/hypoxia inducible factor-1α(HIF-1α) pathway. A subcutaneous 143B tumor-bearing nude mouse model was successfully established and randomly divided into the model group, and the AC 1∶1, 2∶1, and 4∶1 groups. Body weight, tumor volume, and tumor weight were recorded. Real-time quantitative polymerase chain reaction(RT-qPCR) and Western blot were used to detect the mRNA and protein expression levels of PI3K, Akt, phosphorylated Akt(p-Akt), HIF-1α, vascular endothelial growth factor A(VEGFA), transforming growth factor-β1(TGF-β1), epithelial cadherin(E-cadherin), neural cadherin(N-cadherin), vimentin, matrix metalloproteinase 2(MMP2), matrix metalloproteinase 9(MMP9), B-cell lymphoma-2(Bcl-2), Bcl-2-associated X protein(Bax), and caspase-3 in the hypoxic core region of the tumor tissue. A cell hypoxia model was established, and the effects of AC-medicated serum(model group, AC 1∶1, 2∶1, and 4∶1 groups) on angiogenesis, proliferation, adhesion, invasion, and migration of 143B osteosarcoma cells were examined through CCK-8, flow cytometry, Transwell assay, cell adhesion assay, and HUVEC tube formation assay. The results showed that compared with the model group, the tumor weight and volume were smallest in the 2∶1 group. The expression levels of PI3K, Akt, p-Akt, HIF-1α, VEGFA, and TGF-β1 were significantly decreased, and the protein expression of E-cadherin was significantly increased, while the protein expression of N-cadherin, vimentin, MMP2, and MMP9 was significantly decreased. Additionally, the protein expression of Bax and caspase-3 was significantly increased, and Bcl-2 protein expression was significantly decreased. In vitro experiments showed that after intervention with AC-medicated serum at a 2∶1 ratio, the cell activity, adhesion, invasion, and migration of 143B cells were significantly reduced, apoptosis was significantly increased, and HUVEC tube formation was significantly decreased. In conclusion, the 2∶1 ratio of AC showed the most effective inhibition of 143B cell growth. AC can inhibit the growth and metastasis of osteosarcoma 143B cells by regulating the PI3K/Akt/HIF-1α signaling pathway, inhibiting angiogenesis and reducing cell adhesion, invasion, and migration.
Osteosarcoma/pathology*
;
Animals
;
Proto-Oncogene Proteins c-akt/genetics*
;
Hypoxia-Inducible Factor 1, alpha Subunit/genetics*
;
Humans
;
Mice
;
Cell Adhesion/drug effects*
;
Cell Proliferation/drug effects*
;
Neovascularization, Pathologic/metabolism*
;
Drugs, Chinese Herbal/administration & dosage*
;
Phosphatidylinositol 3-Kinases/genetics*
;
Cell Line, Tumor
;
Mice, Nude
;
Signal Transduction/drug effects*
;
Astragalus Plant/chemistry*
;
Bone Neoplasms/physiopathology*
;
Male
;
Rhizome/chemistry*
;
Mice, Inbred BALB C
;
Angiogenesis
5.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
6.Buyang Huanwu Decoction promotes angiogenesis after oxygen-glucose deprivation/reoxygenation injury of bEnd.3 cells by regulating YAP1/HIF-1α signaling pathway via caveolin-1.
Bo-Wei CHEN ; Yin OUYANG ; Fan-Zuo ZENG ; Ying-Fei LIU ; Feng-Ming TIAN ; Ya-Qian XU ; Jian YI ; Bai-Yan LIU
China Journal of Chinese Materia Medica 2025;50(14):3847-3856
This study aims to explore the mechanism of Buyang Huanwu Decoction(BHD) in promoting angiogenesis after oxygen-glucose deprivation/reoxygenation(OGD/R) of mouse brain microvascular endothelial cell line(brain-derived Endothelial cells.3, bEnd.3) based on the caveolin-1(Cav1)/Yes-associated protein 1(YAP1)/hypoxia-inducible factor-1α(HIF-1α) signaling pathway. Ultra-high performance liquid chromatography-quadrupole-time-of-flight mass spectrometry(UPLC-Q-TOF-MS) was used to analyze the blood components of BHD. The cell counting kit-8(CCK-8) method was used to detect the optimal intervention concentration of drug-containing serum of BHD after OGD/R injury of bEnd.3. The lentiviral transfection method was used to construct a Cav1 silent stable strain, and Western blot and polymerase chain reaction(PCR) methods were used to verify the silencing efficiency. The control bEnd.3 cells were divided into a normal group(sh-NC control group), an OGD/R model + blank serum group(sh-NC OGD/R group), and an OGD/R model + drug-containing serum group(sh-NC BHD group). Cav1 silent cells were divided into an OGD/R model + blank serum group(sh-Cav1 OGD/R group) and an OGD/R model + drug-containing serum group(sh-Cav1 BHD group). The cell survival rate was detected by the CCK-8 method. The cell migration ability was detected by a cell migration assay. The lumen formation ability was detected by an angiogenesis assay. The apoptosis rate was detected by flow cytometry, and the expression of YAP1/HIF-1α signaling pathway-related proteins in each group was detected by Western blot. Finally, co-immunoprecipitation was used to verify the interaction between YAP1 and HIF-1α. The results showed astragaloside Ⅳ, formononetin, ferulic acid, and albiflorin in BHD can all enter the blood. The drug-containing serum of BHD at a mass fraction of 10% may be the optimal intervention concentration for OGD/R-induced injury of bEnd.3 cells. Compared with the sh-NC control group, the sh-NC OGD/R group showed significantly decreased cell survival rate, cell migration rate, mesh number, node number, and lumen length, significantly increased cell apoptotic rate, significantly lowered phosphorylation level of YAP1 at S127 site, and significantly elevated nuclear displacement level of YAP1 and protein expression of HIF-1α, vascular endothelial growth factor(VEGF), and vascular endothelial growth factor receptor 2(VEGFR2). Compared with the same type of OGD/R group, the sh-NC BHD group and sh-Cav1 BHD group had significantly increased cell survival rate, cell migration rate, mesh number, node number, and lumen length, a significantly decreased cell apoptotic rate, a further decreased phosphorylation level of YAP1 at S127 site, and significantly increased nuclear displacement level of YAP1 and protein expression of HIF-1α, VEGF, and VEGFR2. Compared with the sh-NC OGD/R group, the sh-Cav1 OGD/R group exhibited significantly decreased cell survival rate, cell migration rate, mesh number, node number, and lumen length, a significantly increased cell apoptotic rate, a significantly increased phosphorylation level of YAP1 at S127 site, and significantly decreased nuclear displacement level of YAP1 and protein expression of HIF-1α, VEGF, and VEGFR2. Compared with the sh-NC BHD group, the sh-Cav1 BHD group showed significantly decreased cell survival rate, cell migration rate, mesh number, node number, and lumen length, a significantly increased cell apoptotic rate, a significantly increased phosphorylation level of YAP1 at the S127 site, and significantly decreased nuclear displacement level of YAP1 and protein expression of HIF-1α, VEGF, and VEGFR2. YAP1 protein was present in the protein complex precipitated by the HIF-1α antibody, and HIF-1α protein was also present in the protein complex precipitated by the YAP1 antibody. The results confirmed that the drug-containing serum of BHD can increase the activity of YAP1/HIF-1α pathway in bEnd.3 cells damaged by OGD/R through Cav1 and promote angiogenesis in vitro.
Drugs, Chinese Herbal/pharmacology*
;
Animals
;
Mice
;
Signal Transduction/drug effects*
;
Glucose/metabolism*
;
Caveolin 1/genetics*
;
Hypoxia-Inducible Factor 1, alpha Subunit/genetics*
;
YAP-Signaling Proteins
;
Oxygen/metabolism*
;
Endothelial Cells/metabolism*
;
Cell Line
;
Adaptor Proteins, Signal Transducing/genetics*
;
Neovascularization, Physiologic/drug effects*
;
Cell Hypoxia/drug effects*
;
Angiogenesis
7.Kazinol B alleviates hypoxia/reoxygenation-induced hepatocyte injury by inhibiting the JNK signaling pathway.
Yi ZHU ; Junhui LI ; Min YANG ; Pengpeng ZHANG ; Cai LI ; Hong LIU
Journal of Central South University(Medical Sciences) 2025;50(2):181-189
OBJECTIVES:
Hypoxia/reoxygenation (H/R) injury is a critical pathological process during liver transplantation. Kazinol B has known anti-inflammatory, anti-apoptotic, and metabolic regulatory properties, but its protective mechanism in H/R-induced liver injury remains unclear. This study aims to investigate the protective effects and underlying mechanisms of Kazinol B in H/R-induced hepatocyte injury.
METHODS:
An ischemia-reperfusion model was established in healthy adult male Sprague-Dawley rats, and an in vitro H/R model was created using cultured hepatocytes. Hepatocytes were treated with Kazinol B (0-100 μmol/L) to assess cytotoxicity and protective effects. Cell viability was evaluated using the cell counting kit-8 (CCK-8) and lactate dehydrogenase (LDH) release assays. Expression of apoptosis-related proteins, B-cell lymphoma 2 (Bcl-2), Bcl-2-associated death promoter (Bad), and cleaved caspase-3, was detected by Western blotting. Reactive oxygen species (ROS) levels were assessed via fluorescence probes, and inflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) were measured using enzyme-linked immunosorbent assay (ELISA). TdT-mediated nick end labeling (TUNEL) staining was performed to assess DNA damage and apoptosis.
RESULTS:
Kazinol B had no significant effect on hepatocyte viability at 0-50 μmol/L, but showed cytotoxicity at 100 μmol/L (P<0.05). At 0.1-20 μmol/L, Kazinol B significantly improved cell survival, reduced LDH release, decreased apoptosis, and attenuated DNA damage (all P<0.001). At 10 μmol/L, Kazinol B markedly down-regulated Bad and cleaved caspase-3 (both P<0.05), and up-regulated Bcl-2 (P<0.01). It also dose-dependently reduced ROS levels and inflammatory cytokines TNF-α and IL-1β (all P<0.01). Both in vitro and in vivo, Kazinol B inhibited activation of the c-Jun N-terminal kinase (JNK) pathway without affecting extracellular regulated protein kinase (ERK) signaling (P>0.05). TUNEL staining showed that the protective effect of Kazinol B against apoptosis was partially reversed by the JNK agonist anisomycin (P<0.01).
CONCLUSIONS
Kazinol B mitigates hepatocyte injury induced by H/R by inhibiting the JNK signaling pathway. Its protective effect is associated with suppression of oxidative stress and inflammation, indicating its potential as a hepatoprotective agent.
Animals
;
Hepatocytes/pathology*
;
Rats, Sprague-Dawley
;
Male
;
Rats
;
Reperfusion Injury/prevention & control*
;
Apoptosis/drug effects*
;
Reactive Oxygen Species/metabolism*
;
MAP Kinase Signaling System/drug effects*
;
Cell Survival/drug effects*
;
Cell Hypoxia
;
Cells, Cultured
8.Li Qi Huo Xue Di Wan alleviates hypoxia-induced injury in human cardiac microvascular endothelial cells by inhibiting apoptosis and necroptosis pathways.
Can TANG ; Yiyue ZHANG ; Xiuju LUO ; Jun PENG
Journal of Central South University(Medical Sciences) 2025;50(4):631-640
OBJECTIVES:
Injury to human cardiac microvascular endothelial cells (HCMECs) compromises myocardial microcirculation and may contribute to major cardiovascular events such as coronary heart disease, posing a serious health threat. Understanding the mechanisms of hypoxia-induced HCMEC damage is thus of great clinical relevance. This study aims to investigate the protective effects and underlying mechanisms of Li Qi Huo Xue Di Wan against hypoxia-induced HCMEC injury.
METHODS:
HCMECs were cultured under hypoxic conditions for 24 hours to establish a cellular model of hypoxic injury. Cells were divided into six groups: normal control, hypoxia, hypoxia + low-dose Li Qi Huo Xue Di Wan, hypoxia + medium-dose, hypoxia + high-dose, and hypoxia + salvianolic acid B (positive control). Cell viability was assessed using the MTS assay. Lactate dehydrogenase (LDH) release and malondialdehyde (MDA) content were measured to evaluate cytotoxicity and oxidative stress. Activities of superoxide dismutase (SOD), catalase (CAT), caspase-3, and caspase-8 were determined with corresponding assay kits. Apoptosis was analyzed by flow cytometry, and expression of necroptosis-related proteins, receptor-interacting protein kinase 1 (RIPK1) and its phosphorylated form (p-RIPK1), receptor-interacting protein kinase 3 (RIPK3) and its phosphorylated form (p-RIPK3), mixed lineage kinase domain-like protein (MLKL) and its phosphorylated form (p-MLKL), was examined via Western blotting.
RESULTS:
Compared with the control group, hypoxia significantly decreased cell viability (P<0.01), increased MDA levels (P<0.05), and reduced CAT and SOD activity (P<0.05), accompanied by elevated apoptosis (P<0.01) and increased levels of p-RIPK1, p-RIPK3, and p-MLKL (P<0.05). High-dose Li Qi Huo Xue Di Wan significantly improved cell viability (P<0.01), reduced MDA content (P<0.05), increased CAT activity (P<0.05), and suppressed necroptosis-related protein expression (P<0.05) compared with the hypoxia group.
CONCLUSIONS
Li Qi Huo Xue Di Wan exerts a protective effect against hypoxia-induced injury in HCMECs. This effect is mediated by attenuation of oxidative stress, thereby reducing both apoptosis and necroptosis.
Humans
;
Apoptosis/drug effects*
;
Necroptosis/drug effects*
;
Drugs, Chinese Herbal/pharmacology*
;
Cell Hypoxia/drug effects*
;
Endothelial Cells/pathology*
;
Oxidative Stress/drug effects*
;
Cells, Cultured
;
Cell Survival/drug effects*
;
Receptor-Interacting Protein Serine-Threonine Kinases/metabolism*
9.Alamandine inhibits pathological retinal neovascularization by targeting the MrgD-mediated HIF-1α/VEGF pathway.
Kun ZHAO ; Yaping JIANG ; Wen HUANG ; Yukang MAO ; Yihui CHEN ; Peng LI ; Chuanxi YANG
Journal of Zhejiang University. Science. B 2025;26(10):1015-1036
Retinopathy of prematurity (ROP) is a vision-threatening disorder that leads to pathological growth of the retinal vasculature due to hypoxia. Here, we investigated the potential effects of alamandine, a novel heptapeptide in the renin-angiotensin system (RAS), on hypoxia-induced retinal neovascularization and its underlying mechanisms. In vivo, the C57BL/6J mice with oxygen-induced retinopathy (OIR) were injected intravitreally with alamandine (1.0 μmol/kg per eye). In vitro, human retinal microvascular endothelial cells (HRMECs) were utilized to investigate the effects of alamandine (10 μg/mL) on proliferation, apoptosis, migration, and tubular formation under vascular endothelial growth factor (VEGF) stimulation. Single-cell RNA sequencing (scRNA-seq) matrix data from the Gene Expression Omnibus (GEO) database and RAS-related genes from the Molecular Signatures Database (MSigDB) were sourced for subsequent analyses. By integrating scRNA-seq data across multiple species, we identified that RAS-associated endothelial cell populations were highly related to retinal neovascularization. The liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis revealed a significant decrease in alamandine levels in both the serum and retina of OIR mice compared to those in the control group. Next, alamandine ameliorated hypoxia-induced retinal pathological neovascularization and physiologic revascularization in OIR mice. In vitro, alamandine effectively mitigated VEGF-induced proliferation, scratch wound healing, and tube formation of HRMECs primarily by inhibiting the hypoxia-inducible factor-1α (HIF-1α)/VEGF pathway. Further, coincubation with D-Pro7 (Mas-related G protein-coupled receptor D (MrgD) antagonist) hindered the beneficial impacts of alamandine on hypoxia-induced pathological angiogenesis both in vivo and in vitro. Our findings suggested that alamandine could mitigate retinal neovascularization by targeting the MrgD-mediated HIF-1α/VEGF pathway, providing a potential therapeutic agent for OIR prevention and treatment.
Animals
;
Retinal Neovascularization/prevention & control*
;
Mice, Inbred C57BL
;
Vascular Endothelial Growth Factor A/metabolism*
;
Humans
;
Mice
;
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism*
;
Oligopeptides/therapeutic use*
;
Signal Transduction/drug effects*
;
Cell Proliferation/drug effects*
;
Endothelial Cells/drug effects*
;
Retinopathy of Prematurity/drug therapy*
;
Apoptosis/drug effects*
;
Cell Movement/drug effects*
;
Renin-Angiotensin System/drug effects*
;
Cells, Cultured
10.Buyang Huanwu Decoction reduces mitochondrial autophagy in rheumatoid arthritis synovial fibroblasts in hypoxic culture by inhibiting the BNIP3-PI3K/Akt pathway.
Junping ZHAN ; Shuo HUANG ; Qingliang MENG ; Wei FAN ; Huimin GU ; Jiakang CUI ; Huilian WANG
Journal of Southern Medical University 2025;45(1):35-42
OBJECTIVES:
To investigate the role of the BNIP3-PI3K/Akt signaling pathway in mediating the inhibitory effect of Buyang Huanwu Decoction (BYHWT) on mitochondrial autophagy in human synovial fibroblasts from rheumatoid arthritis patients (FLS-RA) cultured under a hypoxic condition.
METHODS:
Forty normal Wistar rats were randomized into two groups (n=20) for daily gavage of BYHWT or distilled water for 7 days to prepare BYHWT-medicated or control sera. FLS-RA were cultured in routine condition or exposed to hypoxia (10% O2) for 24 h wigh subsequent treatment with IL-1β, followed by treatment with diluted BYHWT-medicated serum (5%, 10% and 20%) or control serum. AnnexinV-APC/7-AAD double staining and T-AOC kit were used for detecting apoptosis and total antioxidant capacity of the cells, and the changes in ROS, ATP level, mitochondrial membrane potential and Ca2+ homeostasis were analyzed. The changes in mRNA and protein expressions of BNIP3, PI3K and AKT and mRNA expressions of LC3, Beclin-1 and P62 were detected using RT-qPCR and Western blotting.
RESULTS:
Treatment with BYHWT-medicated serum dose-dependently lowered apoptosis rate of IL-1β-induced FLS-RA with hypoxic exposure. The treatment significantly decreased T-AOC concentration, increased ROS production, autophagosome formation and ATPase levels, and lowered mitochondrial membrane potential and Ca2+ level in the cells. In IL-1β-induced FLS-RA with hypoxic exposure, treatment with BYHWT-medicated serum significantly increased BNIP3 protein expression, decreased the protein expressions of PI3K and AKT, increased the mRNA expressions of BNIP3 and P62, and lowered the mRNA expressions of PI3K, AKT, LC3 and Beclin-1 without significantly affecting Beclin-1 protein expression. The cells treated with 5% and 10% BYHWT-medicated serum showed no significant changes in LC3 expression.
CONCLUSIONS
BYHWT inhibits mitochondrial autophagy in IL-1β-induced FLS-RA with hypoxic exposure possibly by inhibiting BNIP3-mediated PI3K/AKT signaling pathway.
Drugs, Chinese Herbal/pharmacology*
;
Arthritis, Rheumatoid/pathology*
;
Animals
;
Signal Transduction/drug effects*
;
Proto-Oncogene Proteins c-akt/metabolism*
;
Autophagy/drug effects*
;
Humans
;
Fibroblasts/cytology*
;
Rats, Wistar
;
Membrane Proteins/metabolism*
;
Rats
;
Phosphatidylinositol 3-Kinases/metabolism*
;
Mitochondria/metabolism*
;
Cells, Cultured
;
Proto-Oncogene Proteins/metabolism*
;
Apoptosis/drug effects*
;
Cell Hypoxia
;
Synovial Membrane/cytology*
;
Male
;
Mitochondrial Proteins

Result Analysis
Print
Save
E-mail