Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 292
Filter
1.
Chinese Journal of Experimental Traditional Medical Formulae ; (24): 101-107, 2024.
Article in Chinese | WPRIM | ID: wpr-1013345

ABSTRACT

ObjectiveMyelodysplastic syndromes (MDS) is a group of clonal hematopoietic stem cell disorders,and this study aims to investigate the expression of hypoxia-inducible factor-1α(HIF-1α) in the bone marrow cells of patients with MDS and its correlation with the clinical features of MDS,the therapeutic efficacy of arsenic-containing Chineseherbal compound,and the survival prognosis. MethodAccording to the inclusion and exclusion criteria,27 MDS patients treated with arsenic-containing Chinese herbal compound in the Department of Hematology,Xiyuan Hospital,China Academy of Chinese Medical Sciences from January 2022 to September 2022 were included,and their bone marrow samples were collected by myelotomy. HIF-1α expression level in bone marrow cells was detected by real-time polymerase chain reaction (PCR) to analyze its correlation with clinical features,and logistic and Cox regression was used to analyze the risk factors affecting the efficacy and prognostic survival of MDS patients. ResultThe HIF-1α mRNA expression level was lower in bone marrow cells of MDS patients than in healthy subjects. HIF-1α was positively correlated with the degree of myelodysplasia(r=0.384,P<0.05) and bone marrow granulocytic system%(G%)(r=0.560,P<0.01). Logistic regression showed that HIF-1α was a risk factor for the prognosis in the follow-up of the efficacy of treatment(P<0.05)and Cox regression showed that HIF-1α was an independent factor affecting the survival prognosis of MDS patients [odds ratio(OR)=398.968,95% confidence interval(CI)(1.281,116 858.743),P<0.05]. ConclusionThe level of HIF-1α expression in bone marrow cells of MDS patients was closely related to the degree of clinical myelodysplasia and G%,and HIF-1α was a risk factor for the efficacy for and survival prognosis of MDS patients.

2.
Chinese Journal of Experimental Traditional Medical Formulae ; (24): 10-18, 2024.
Article in Chinese | WPRIM | ID: wpr-1013335

ABSTRACT

ObjectiveThis study aims to investigate the inhibitory effect of Wutoutang on pannus formation in adjuvant-induced arthritis (AIA) rats with wind-cold-dampness Bi syndrome and its potential mechanism. MethodA total of 40 male SD specific pathogen-free (SPF) rats were selected and divided into blank group, wind-cold-dampness Bi syndrome group [Complete Freund's Adjuvant (CFA), 200 μg], Wutoutang group (15 g·kg-1·d-1), and indometacin group (10 mg·kg-1) according to random number table method. Except for the blank group, the other groups were given wind-cold-dampness stimulation before the CFA injection. After the rats were administered for 30 days, the basic conditions, onset time, arthritis index score, and foot swelling volume of AIA rats with wind-cold-dampness Bi syndrome were observed. Finally, peripheral arterial blood, ankle joint, and synovial tissue were taken. Enzyme-linked immunosorbent assay (ELISA) was used to detect serum hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor A (VEGFA) protein content, and rheumatism, including anti-O (ASO), C-reactive protein (CRP), and rheumatoid factor (RF). Hematoxylin-eosin (HE) staining revealed the changes in joint histomorphology. Immunohistochemistry was used to detect the expression of HIF-1α and VEGFA, two important proteins in the ankle pathway. Quantitative real-time polymerase chain reaction (Real-time PCR) was used to reveal mRNA levels of HIF-1α, VEGFA, angiopoietin-1 (Ang-1), and angiopoietin-2 (Ang-2) in rat synovial tissue. ResultThe foot swelling volume and arthritis score of AIA rats with wind-cold-dampness Bi syndrome were substantially higher (P<0.01) compared with the blank group. Serum CRP, RF, and ASO levels were considerably elevated (P<0.01). HE staining showed obvious hyperplasia of ankle synovium and synovial inflammation, angiogenesis and pannus formation, and aggravated bone destruction, indicating successful modeling. After the intervention of Wutoutang, the onset time was delayed (P<0.01). Foot swelling volume and arthritis score were decreased (P<0.01). Serum CRP, RF, and ASO levels were significantly decreased (P<0.01). The inflammatory hyperplasia of synovial tissue, angiogenesis and pannus formation, and bone destruction were alleviated. The mRNA levels of HIF-1α, VEGFA, Ang-1, and Ang-2 in the synovial membrane were significantly decreased (P<0.05, P<0.01). The expressions of HIF-1α and VEGFA in serum and ankle joints were decreased (P<0.01). In the indomethacin group, the onset time of the disease was delayed (P<0.01). Foot swelling volume and arthritis score were decreased (P<0.01). Serum CRP, RF, and ASO levels were significantly decreased (P<0.01). HIF-1α/VEGFA/Ang signaling pathway was activated, and pathological tissue injury was improved. ConclusionWutoutang can delay the onset time of AIA rats with wind-cold-dampness Bi syndrome, reduce foot swelling volume, arthritis score, rheumatic activity, and improve joint histopathology. It can inhibit pannus formation, and its mechanism may be related to down-regulating the expression of the HIF-1α/VEGFA/Ang pathway.

3.
China Pharmacy ; (12): 712-717, 2024.
Article in Chinese | WPRIM | ID: wpr-1013107

ABSTRACT

OBJECTIVE To investigate the effect and mechanism of Astragalus polysaccharide (APS) on peritoneal fibrosis and angiogenesis in rats with peritoneal dialysis (PD). METHODS Rats were randomly divided into normal control group (Control group), model group (PD group), 70 mg/kg APS group (APS-L group), 140 mg/kg APS group (APS-H group), and 140 mg/kg APS+40 mg/kg hypoxia-inducible factor-1α (HIF-1α) agonist DMOG group (APS-H+DMOG group), with 12 rats in each group. PD rat models were constructed in the last four groups of rats. Administration groups were given APS intragastrically and DMOG intraperitoneally. Control group and PD group were given constant volume of normal saline intragastrically, once a day, for 4 consecutive weeks. After the last medication, the peritoneal ultrafiltration (UF), mass transfer of glucose (MTG), the levels of serum creatinine (Scr) and blood urea nitrogen (BUN) were detected in rats; peritoneal histomorphology and peritoneal fibrosis (peritoneal thickness and proportion of collagen fiber deposition) were observed; the microvascular density and the expression levels of α-smooth muscle actin (α-SMA), laminin (LN), HIF-1α and vascular endothelial growth factor (VEGF) proteins were detected in peritoneal tissue of rats. RESULTS Compared with Control group, the mesothelium of rats in the PD group was loosely arranged and shed, inflammatory cells infiltrated, the peritoneal thickness and proportion of collagen fiber deposition were increased significantly (P<0.05). The levels of MTG, Scr and BUN in serum, microvascular density and the expressions of α-SMA, LN, HIF-1α and VEGF proteins were significantly increased, while the level of UF was significantly decreased (P< 0.05); compared with PD group, the levels of above indexes were significantly reversed in APS-L and APS-H groups (P<0.05), and the improvement of APS-H group was better than APS-L group (P<0.05). Compared with APS-H group, the levels of above indexes in APS-H+DMOG group were all reversed (P<0.05). CONCLUSIONS APS inhibits peritoneal fibrosis and angioge-nesis in PD rats by inhibiting HIF-1α/VEGF signaling pathway.

4.
China Pharmacy ; (12): 695-700, 2024.
Article in Chinese | WPRIM | ID: wpr-1013104

ABSTRACT

OBJECTIVE To investigate the effect and mechanism of Panax notoginseng saponins (PNS) on wound healing after anal fistula surgery in rats by regulating the hypoxia-inducible factor-1α (HIF-1α)/vascular endothelial growth factor (VEGF)/ vascular endothelial growth factor receptor-2 (VEGFR2) signaling pathway. METHODS SD rats were selected to establish a postoperative rat model of anal fistula by infecting wound with Escherichia coli. The model rats were randomly grouped into model group, PNS low-dose and high-dose groups (15, 30 mg/cm2), high-dose of PNS+2-methoxyestradiol (2ME2) group (PNS 30 mg/cm2+HIF-1α inhibitor 2ME2 4 mg/kg), with 10 rats in each group. Another 10 normal rats were selected for back hair removal treatment as the control group. Each drug group was injected with the corresponding drug solution intramuscularly or (and) intraperitoneally, once a day, for 3 weeks. After the last administration, the wound healing rate (excluding the control group), microvascular density (MVD), the expression of collagen Ⅰ and fibronectin (FN) in the wound tissue were detected in each group; the levels of angiogenic factors [VEGF, E-mail:842710813@qq.com angiopoietin-Ⅰ (Ang-Ⅰ), Ang-Ⅱ] in serum, the levels of inflammatory factors [interleukin-6 (IL-6) and IL-2] in serum binggui7183@163.com and wound tissue as well as the expressions of the related proteins of HIF-1α/VEGF/VEGFR2 signaling pathway in the wound tissue of rats were also detected in each group. RESULTS The MVD, the expression of collagen Ⅰ and FN in the wound tissue, and the levels of IL-6 and IL-2 in serum and wound tissue of rats increased significantly in the model group, compared to the control group (P<0.05), while the serum levels of VEGF, Ang- Ⅰ and Ang-Ⅱ decreased significantly (P<0.05). The wound healing rate, the MVD in wound tissue, the serum levels of VEGF, Ang-Ⅰ and Ang-Ⅱ, the expressions of collagen Ⅰ and FN in the wound tissue, and protein expressions of HIF-1α, VEGF and VEGFR2 in the PNS low-dose and high-dose groups increased significantly, compared to the model group (P<0.05), while the levels of IL-6 and IL-2 in serum and wound tissue decreased significantly (P<0.05); the high-dose PNS had a stronger effect (P< 0.05). 2ME2 could weaken the effect of PNS on above indicators of rats after anal fistula surgery (P<0.05). CONCLUSIONS PNS can promote the production of angiogenic factors and inhibit the production of pro-inflammatory factors, thereby promoting wound healing in rats after anal fistula surgery. The above effects are related to the activation of HIF-1α/VEGF/VEGFR2 signaling pathway.

5.
Chinese Journal of Experimental Traditional Medical Formulae ; (24): 57-65, 2024.
Article in Chinese | WPRIM | ID: wpr-1012693

ABSTRACT

ObjectiveTo observe the therapeutic effect of Shugan Huazheng prescription on hepatic fibrosis model rats induced by carbon tetrachloride (CCl4) and explore whether it plays its role through hypoxia-induced factor-1α/vascular endothelial growth factor/transforming growth factor-β1 (HIF-1α/VEGF/TGF-β1) pathway. MethodA total of 54 male SPF SD rats were randomly divided into six groups: blank group, model group, colchicine group (0.2 mg·kg-1), and high-, medium-, and low-dose groups (29.52, 14.76, and 7.38 g·kg-1) of Shugan Huazheng prescription, with nine rats in each group. The molding was conducted three times a week for eight weeks. Administration began the day after the first injection, and the drug intervention was once a day for eight weeks. On the day after the last administration, the rats were deprived of food and water, and they were killed the next day, during which the physiological status of each group of rats was dynamically monitored. The pathological changes in the liver were observed by hematoxylin-eosin (HE) staining, and the content of hydroxyproline (HYP) and angiotensin Ⅱ (AngⅡ) in liver tissue were detected by enzyme-related immunosorbent assay (ELISA). Real-time fluorescent quantitative PCR (Real-time PCR) was used to determine the mRNA expression levels of HIF-1α, VEGF, and TGF-β1 in liver tissue, and immunohistochemical method (IHC) and Western blot were used to detect the protein expression levels of HIF-1α, VEGF, and TGF-β1 in liver tissue. ResultCompared with the blank group, the overall condition of rats in the model group decreased significantly. The proliferation of connective tissue and the increase in adipose cells between hepatocytes were obvious. The content of HYP and Ang was increased. The mRNA and protein expressions of HIF-1α, VEGF, and TGF-β1 were increased to varying degrees (P<0.05). Compared with the model group, the proliferation of connective tissue and inflammatory cell infiltration in the liver tissue of colchicine and Shugan Huazheng prescription groups were reduced. The content of HYP and Ang was decreased. The mRNA and protein expression levels of HIF-1α, VEGF, and TGF-β1 were decreased, and the colchicine group and high-dose group of Shugan Huazheng prescription were the most significant (P<0.05). ConclusionShugan Huazheng prescription has an obvious therapeutic effect on CCl4-induced hepatic fibrosis model rats. Its therapeutic mechanism may be related to the regulation of the HIF-1α/VEGF/TGF-β1 signaling pathway and the improvement of hepatic hypoxia, vascular remodeling, and the syndrome of Qi deficiency and blood stasis in hepatic fibrosis.

6.
Chinese Journal of Experimental Traditional Medical Formulae ; (24): 59-65, 2024.
Article in Chinese | WPRIM | ID: wpr-1011443

ABSTRACT

ObjectiveTo reveal the effects of Huanglian Jiedutang (HLJDT) on the learning and memory abilities of APP/PS1 transgenic mice via hypoxia-inducible factor-1α (HIF-1α)/vascular endothelial growth factor (VEGF) signaling pathway. MethodForty 5-month-old β-amyloid precursor protein (APP)/presenilin 1(PS1) mice were randomized into the model, donepezil (0.001 g·kg-1·d-1), and low-, medium-, and high-dose (1.5, 3, 6 g·kg-1·d-1, respectively) HLJDT groups, and 8 C57BL/6 mice were taken as the normal group. After 45 days of continuous administration, Morris water maze test was conducted, and the organ indexes were calculated. The morphological structure of cerebral vascular endothelial cells in mice was observed under a transmission electron microscope. Western blot was employed to measure the protein levels of APP, HIF-1α, VEGF,VEGFA, and brain-derived neurotrophic factor (BDNF) in the hippocampus. The mRNA levels of APP, HIF-1α, and VEGF were determined by real-time fluorescence quantitative polymerase chain reaction (Real-time PCR). ResultCompared with the normal group, the model group showed prolonged escape latency (P<0.05), reduced distance and time around the target platform (P<0.05), decrease brain and spleen indexes (P<0.05), vascular endothelial cells with karyopyknosis and not abundant cytoplasm, up-regulated protein levels of APP, HIF-1α, VEGF, and VEGFA (P<0.05), down-regulated protein level of BDNF (P<0.05), and up-regulated mRNA levels of APP, HIF-1α, and VEGF (P<0.05) in the hippocampus. Compared with the model group, high-dose HLJDT shortened the escape latency (P<0.05), increased the distance and time around the target platform (P<0.05), raised the brain and spleen indexes (P<0.05), repaired the organelles of vascular endothelial cells, down-regulated the protein levels of APP, HIF-1α, VEGF, and VEGFA (P<0.05), up-regulated the protein level of BDNF (P<0.05), and down-regulated the mRNA levels of APP, HIF-1α, and VEGF (P<0.05) in the hippocampus. ConclusionHLJDT can improve the learning and memory abilities of mice by reducing the expression of HIF-1α and VEGF, thus protecting the nerves.

7.
International Eye Science ; (12): 345-350, 2024.
Article in Chinese | WPRIM | ID: wpr-1011380

ABSTRACT

AIM: To investigate the effect of long non-coding RNA-HIF1A-AS1(lncRNA HIF1A-AS1)on the chemotherapy sensitivity of vincristine(VCR)-resistant in retinoblastoma(RB)cells by regulating the expression of hypoxia-inducible factor-1α(HIF-1α).METHODS: The human RB VCR-resistant cell line SO-RB50/VCR was established, expression of lncRNA HIF1A-AS1 in SO-RB50 and SO-RB50/VCR cells were detected by reverse transcription-quantitative real-time PCR(RT-qPCR); inhibition of lncRNA HIF1A-AS1 expression or simultaneous overexpression of HIF-1α in SO-RB50/VCR cells, and then median inhibitory concentration(IC50)of VCR and cell proliferation and apoptosis were detected in SO-RB50/VCR cells; the protein expressions of HIF-1α, multidrug resistance associate protein(MRP)and P-glycoprotein(P-gp)were measured by Western blot.RESULTS: Compared with SO-RB50 cells, the expression levels of lncRNA HIF1A-AS1 and HIF-1α protein in SO-RB50/VCR cells were increased(P&#x003C;0.05); after inhibiting the expression of lncRNA HIF1A-AS1 in SO-RB50/VCR cells, the apoptosis rate was significantly increased(P&#x003C;0.05), optical density(OD450), the IC50 value of VCR on cells and the expression levels of HIF-1α, MRP and P-gp proteins were significantly reduced(P&#x003C;0.05); overexpression of HIF-1α attenuates the inhibitory effect of down-regulated lncRNA HIF1A-AS1 expression on drug resistance in SO-RB50/VCR cells.CONCLUSION: The lncRNA HIF1A-AS1 was highly expressed in SO-RB50/VCR cells, and inhibition of lncRNA HIF1A-AS1 expression reduced VCR resistance in SO-RB50/VCR cells by down-regulating HIF-1α expression.

8.
Acta Pharmaceutica Sinica B ; (6): 273-291, 2024.
Article in English | WPRIM | ID: wpr-1011239

ABSTRACT

Obesity has been known to negatively modulate the life-span and immunosuppressive potential of mesenchymal stromal cells (MSC). However, it remains unclear what drives the compromised potency of obese MSC. In this study, we examined the involvement of adiponectin, an adipose tissue-derived hormone, in obesity-induced impaired therapeutic function of MSC. Diet-induced obesity leads to a decrease in serum adiponectin, accompanied by impairment of survival and immunomodulatory effects of adipose-derived MSC (ADSC). Interestingly, priming with globular adiponectin (gAcrp) improved the immunomodulatory potential of obese ADSC. Similar effects were also observed in lean ADSC. In addition, gAcrp potentiated the therapeutic effectiveness of ADSC in a mouse model of DSS-induced colitis. Mechanistically, while obesity inhibited the glycolytic capacity of MSC, gAcrp treatment induced a metabolic shift toward glycolysis through activation of adiponectin receptor type 1/p38 MAPK/hypoxia inducible factor-1α axis. These findings suggest that activation of adiponectin signaling is a promising strategy for enhancing the therapeutic efficacy of MSC against immune-mediated disorders.

9.
Protein & Cell ; (12): 36-51, 2024.
Article in English | WPRIM | ID: wpr-1010778

ABSTRACT

Hypoxia-inducible factor (HIF-1α), a core transcription factor responding to changes in cellular oxygen levels, is closely associated with a wide range of physiological and pathological conditions. However, its differential impacts on vascular cell types and molecular programs modulating human vascular homeostasis and regeneration remain largely elusive. Here, we applied CRISPR/Cas9-mediated gene editing of human embryonic stem cells and directed differentiation to generate HIF-1α-deficient human vascular cells including vascular endothelial cells, vascular smooth muscle cells, and mesenchymal stem cells (MSCs), as a platform for discovering cell type-specific hypoxia-induced response mechanisms. Through comparative molecular profiling across cell types under normoxic and hypoxic conditions, we provide insight into the indispensable role of HIF-1α in the promotion of ischemic vascular regeneration. We found human MSCs to be the vascular cell type most susceptible to HIF-1α deficiency, and that transcriptional inactivation of ANKZF1, an effector of HIF-1α, impaired pro-angiogenic processes. Altogether, our findings deepen the understanding of HIF-1α in human angiogenesis and support further explorations of novel therapeutic strategies of vascular regeneration against ischemic damage.


Subject(s)
Humans , Vascular Endothelial Growth Factor A/metabolism , Endothelial Cells/metabolism , Transcription Factors/metabolism , Gene Expression Regulation , Hypoxia/metabolism , Cell Hypoxia/physiology
10.
Int. j. morphol ; 41(2): 362-367, abr. 2023. ilus
Article in English | LILACS | ID: biblio-1440327

ABSTRACT

SUMMARY: Liver transplantation is the only available method to treat liver failure induced by chronic liver injury. We sought to determine whether the angiotensin-converting enzyme inhibitor, captopril, can inhibit the development of chronic liver injury induced by the hepatotoxic agent thioacetamide (TAA) in association with the suppression of inflammation (hsCRP, TNF-α, and IL-6) / hypoxia- inducible factor 1-alpha (HIF-1α) / profibrosis (TIMP-1, MMP-9, and α-SMA) axis that mediates liver injury. Therefore, the model group of rats was injected for eight weeks with 200 mg/kg TAA starting at week two. The protective group was pretreated with 150 mg/ kg captopril daily for two weeks prior to TAA injections and continued receiving both capropril and TAA agents until being humanely scrificed at week 10. We observed a substantial damage to liver tissue in the model group as demonstrated by a significant (p<0.0001) increase in blood and hepatic tissue levels of high sensitivity C-reactive protein (hsCRP), tumor necrosis factor-a (TNF-α), interleukin- 6 (L-6), HIF-1α, tissue inhibitor of metalloproteinases-1 (TIMP-1), matrix metalloproteinase-9 (MMP-9), alpha-smooth muscle actin (α-SMA), alanine aminotransferase (ALT), and aspartate aminotransferase (AST). All these parameters were significantly (p<0.0244) protected by captopril. Also, a significant (p<0.0001) positive correlation was observed between a-SMA (profibrosis) and the serum and tissue levels of hsCRP, TNF-α, HIF-1α, TIMP-1, MMP-9, and ALT. Thus, these findings suggest that the induction of chronic liver injury by the hepatotoxic compound, TAA is associated with the upregulation of inflammation/HIF-1α/profibrosis, with captopril exhibiting beneficial hepatic pleotropic effects.


El trasplante de hígado es el único método disponible para tratar la insuficiencia hepática inducida por una lesión hepática crónica. Buscamos determinar si el inhibidor de la enzima convertidora de angiotensina, captopril, puede inhibir el desarrollo de lesión hepática crónica inducida por el agente hepatotóxico tioacetamida (TAA) en asociación con la supresión de la inflamación (hsCRP, TNF-α e IL-6) / factor inducible por hipoxia 1-alfa (HIF-1α) / profibrosis (TIMP-1, MMP-9 y α- SMA) eje que media la lesión hepática. Por lo tanto, al grupo modelo de ratas se le inyectó durante ocho semanas 200 mg/kg de TAA a partir de la semana dos. El grupo protector fue pretratado con 150 mg/kg de captopril al día durante dos semanas antes de las inyecciones de TAA y continuó recibiendo capropril y agentes TAA hasta que fue sacrificado en la semana 10. Observamos un daño sustancial en el tejido hepático en el grupo modelo, como lo demuestra un aumento significativo (p<0,0001) de los niveles en sangre y tejido hepático de proteína C reactiva de alta sensibilidad (hsCRP), factor de necrosis tumoral-α (TNF-a), interleucina-6 (L-6), HIF-1α, inhibidor tisular de metaloproteinasas-1 (TIMP-1), metaloproteinasa de matriz-9 (MMP-9), actina de músculo liso alfa (α-SMA), alanina aminotransferasa (ALT) y aspartato aminotransferasa (AST). Todos estos parámetros estaban significativamente (p<0,0244) protegidos por captopril. Además, se observó una correlación positiva significativa (p<0,0001) entre α-SMA (profibrosis) y los niveles séricos y tisulares de hsCRP, TNF-α, HIF-1α, TIMP- 1, MMP-9 y ALT. Por lo tanto, estos hallazgos sugieren que la inducción de daño hepático crónico por el compuesto hepatotóxico, TAA, está asociada con la regulación al alza de la inflamación/HIF-1α/profibrosis, con captopril exhibiendo efectos pleotrópicos hepáticos beneficiosos.


Subject(s)
Animals , Male , Rats , Thioacetamide/toxicity , Captopril/administration & dosage , Chemical and Drug Induced Liver Injury/drug therapy , Fibrosis , Immunohistochemistry , Blotting, Western , Actins , Tumor Necrosis Factor-alpha , Tissue Inhibitor of Metalloproteinase-1 , Matrix Metalloproteinase 9 , Disease Models, Animal , Hepatocyte Nuclear Factor 1-alpha , Real-Time Polymerase Chain Reaction , Matrix Metalloproteinase Inhibitors , Inflammation , Liver/drug effects
11.
Biol. Res ; 56: 4-4, 2023. ilus, graf
Article in English | LILACS | ID: biblio-1420302

ABSTRACT

BACKGROUND: Spermatogonial stem cells (SSCs) are critical for sustaining spermatogenesis. Even though several regulators of SSC have been identified in rodents, the regulatory mechanism of SSC in humans has yet to be discovered. METHODS: To explore the regulatory mechanisms of human SSCs, we analyzed publicly available human testicular single-cell sequencing data and found that Ankyrin repeat and SOCS box protein 9 (ASB9) is highly expressed in SSCs. We examined the expression localization of ASB9 using immunohistochemistry and overexpressed ASB9 in human SSC lines to explore its role in SSC proliferation and apoptosis. Meanwhile, we used immunoprecipitation to find the target protein of ASB9 and verified its functions. In addition, we examined the changes in the distribution of ASB9 in non-obstructive azoospermia (NOA) patients using Western blot and immunofluorescence. RESULTS: The results of uniform manifold approximation and projection (UMAP) clustering and pseudotime analysis showed that ASB9 was highly expressed in SSCs, and its expression gradually increased during development. The immunohistochemical and dual-color immunofluorescence results displayed that ASB9 was mainly expressed in nonproliferating SSCs. Overexpression of ASB9 in the SSC line revealed significant inhibition of cell proliferation and increased apoptosis. We predicted the target proteins of ASB9 and verified that hypoxia-inducible factor 1-alpha inhibitor (HIF1AN), but not creatine kinase B-type (CKB), has a direct interaction with ASB9 in human SSC line using protein immunoprecipitation experiments. Subsequently, we re-expressed HIF1AN in ASB9 overexpressing cells and found that HIF1AN reversed the proliferative and apoptotic changes induced by ASB9 overexpression. In addition, we found that ABS9 was significantly downregulated in some NOA patients, implying a correlation between ASB9 dysregulation and impaired spermatogenesis. CONCLUSION: ASB9 is predominantly expressed in human SSCs, it affects the proliferation and apoptotic process of the SSC line through HIF1AN, and its abnormal expression may be associated with NOA.


Subject(s)
Humans , Male , Testis/metabolism , Ubiquitin-Protein Ligases/metabolism , Repressor Proteins/metabolism , Spermatogenesis/physiology , Ubiquitins/metabolism , Cell Line , Apoptosis , Cell Proliferation , Suppressor of Cytokine Signaling Proteins/metabolism , Mixed Function Oxygenases/metabolism
12.
Acta Pharmaceutica Sinica ; (12): 3311-3320, 2023.
Article in Chinese | WPRIM | ID: wpr-999069

ABSTRACT

The Tongmai Yangxin pill (TMYX) has potential clinical effects on no-reflow (NR); however, the effective substances and mechanisms by which this occurs remain unclear. This study evaluates the cardioprotective effects and molecular mechanisms of TMYX against NR. We used a myocardial NR rat model (2 h after myocardial ischemia and 2 h after reperfusion) to confirm the effect and mechanism of action of TMYX in alleviating NR. In vitro studies in isolated coronary microvasculature of NR rats and in silico network pharmacology analyses were performed to reveal the underlying mechanisms of TMYX and determine the main components, targets, and pathways of TMYX, respectively. The experiment was approved by the Ethics Committee of Hunan University of Chinese Medicine (LLBH-202212160001). TMYX showed therapeutic effects on NR by improving cardiac structure and function, reducing NR, ischemic areas, and cardiomyocyte injury, and decreasing the content of cardiac troponin I (cTnI). Moreover, the mechanism of TMYX predicted by network pharmacology is related to the hypoxia inducible factor-1 (HIF-1), nuclear factor kappa-B (NF-κB), and tumor necrosis factor (TNF) signaling pathways. TMYX increased the expression of G protein-coupled estrogen receptor (GPER), phospho-extracellular signal-regulated kinase (p-ERK), and HIF-1α. In vitro, TMYX enhanced the diastolic function of coronary microvascular cells; however, this effect was inhibited by GPER inhibitor (G-15), eNOS inhibitor (L-NAME), and sGC inhibitor (ODQ). This study integrates pharmacology and experimental evaluation to reveal that TMYX activates HIF-1α/eNOS signaling pathway by upregulating GPER to relax coronary microvessels, thereby significantly alleviating NR.

13.
International Eye Science ; (12): 1087-1092, 2023.
Article in Chinese | WPRIM | ID: wpr-976475

ABSTRACT

AIM:To clarify the effect of miR-519d-3p on high glucose-induced human retinal microvascular endothelial cells(HRMEC)dysfunction and angiogenesis, and to elucidate the regulatory mechanism of miR-519d-3p on hypoxia inducible factor 1 subunit alpha(HIF-1α).METHODS: The normal glucose(NG)and high glucose(HG)cell models were established by inducing HRMEC with 5 and 30 mmol/L glucose, respectively. Control group: HG cell model was transfected with negative control mimics; mannitol group: the control group was added with 25 mmol/L mannitol; miR-519d-3p overexpression group: HG cell model was transfected with miR-519d-3p mimics; miR-519d-3p combined with HIF-1α overexpression group: HG cell model was co-transfected with miR-519d-3p mimics and HIF-1α overexpression vector. The expression of miR-519d-3p in each group was tested by real-time fluorescence quantitative PCR. The expression of HIF-1α protein in each group was tested by Western blotting. The binding sites between miR-519d-3p and HIF-1α were detected by luciferase reporter gene assay. The cell proliferation of each group was detected by CCK-8. The cell apoptosis of each group was tested by Hoechst 33342 staining. The protein expression of extracellular fluid inflammatory factors tumor necrosis factor-α(TNF-α), interleukin(IL)-1β and IL-6 in each group was tested by ELISA. The formation of new capillary lumen-like structures was detected by tubule formation assay.RESULTS: Compared with the NG, miR-519d-3p expression was significantly reduced in the HG cell model, while HIF-1α protein expression was significantly increased in the HG(all P&#x003C;0.01). Compared with the control group, HIF-1α protein expression was significantly reduced in the miR-519d-3p overexpression group(P&#x003C;0.01). The “CGUGAAA” sequence of miR-519d-3p could specifically bind to the “GCACUUU” sequence of HIF-1α 3'-untranslated region(3'-UTR). Compared with the control group, the miR-519d-3p overexpression group showed a significant increase in 24, 48 and 72h absorbance values, a significant decrease in cell apoptotic rate, a significant decrease in the concentrations of TNF-α, IL-1β and IL-6, and a significant decrease in the number of new capillary lumen-like structures(all P&#x003C;0.01). Compared with the miR-519d-3p overexpression group, the miR-519d-3p combined with HIF-1α overexpression group showed a significant decrease in 24, 48 and 72h absorbance values, a significant increase in cell apoptotic rate, a significant increase in the concentrations of TNF-α, IL-1β and IL-6, and a significant increase in the number of new capillary lumen-like structures(all P&#x003C;0.01). There was no difference between the control group and mannitol group in the comparison of the above indicators(all P&#x003E;0.05).CONCLUSION: miR-519d-3p expression is down-regulated while HIF-1α protein expression is up-regulated in high glucose induced HRMEC model. HIF-1α is a target gene of miR-519d-3p. The miR-519d-3p targets HIF-1α to increase cell proliferation and reduce cell apoptosis and inflammation, thereby alleviating high glucose-induced HRMEC dysfunction and inhibiting angiogenesis.

14.
Journal of Zhejiang University. Science. B ; (12): 221-231, 2023.
Article in English | WPRIM | ID: wpr-971482

ABSTRACT

Metabolic reprogramming is a common phenomenon in cancer, with aerobic glycolysis being one of its important characteristics. Hypoxia-inducible factor-1α (HIF1Α) is thought to play an important role in aerobic glycolysis. Meanwhile, naringin is a natural flavanone glycoside derived from grapefruits and many other citrus fruits. In this work, we identified glycolytic genes related to HIF1Α by analyzing the colon cancer database. The analysis of extracellular acidification rate and cell function verified the regulatory effects of HIF1Α overexpression on glycolysis, and the proliferation and migration of colon cancer cells. Moreover, naringin was used as an inhibitor of colon cancer cells to illustrate its effect on HIF1Α function. The results showed that the HIF1Α and enolase 2 (ENO2) levels in colon cancer tissues were highly correlated, and their high expression indicated a poor prognosis for colon cancer patients. Mechanistically, HIF1Α directly binds to the DNA promoter region and upregulates the transcription of ENO2; ectopic expression of ENO2 increased aerobic glycolysis in colon cancer cells. Most importantly, we found that the appropriate concentration of naringin inhibited the transcriptional activity of HIF1Α, which in turn decreased aerobic glycolysis in colon cancer cells. Generally, naringin reduces glycolysis in colon cancer cells by reducing the transcriptional activity of HIF1Α and the proliferation and invasion of colon cancer cells. This study helps to elucidate the relationship between colon cancer progression and glucose metabolism, and demonstrates the efficacy of naringin in the treatment of colon cancer.


Subject(s)
Humans , Glycolysis , Colonic Neoplasms/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Phosphopyruvate Hydratase/metabolism , Flavanones/pharmacology , Cell Line, Tumor , Databases, Genetic , Cell Proliferation/drug effects , Transfection , Warburg Effect, Oncologic
15.
Journal of Zhejiang University. Science. B ; (12): 50-63, 2023.
Article in English | WPRIM | ID: wpr-971468

ABSTRACT

Accumulating evidence has confirmed the links between transfer RNA (tRNA) modifications and tumor progression. The present study is the first to explore the role of tRNA methyltransferase 5 (TRMT5), which catalyzes the m1G37 modification of mitochondrial tRNAs in hepatocellular carcinoma (HCC) progression. Here, based on bioinformatics and clinical analyses, we identified that TRMT5 expression was upregulated in HCC, which correlated with poor prognosis. Silencing TRMT5 attenuated HCC proliferation and metastasis both in vivo and in vitro, which may be partially explained by declined extracellular acidification rate (ECAR) and oxygen consumption rate (OCR). Mechanistically, we discovered that knockdown of TRMT5 inactivated the hypoxia-inducible factor-1 (HIF-1) signaling pathway by preventing HIF-1α stability through the enhancement of cellular oxygen content. Moreover, our data indicated that inhibition of TRMT5 sensitized HCC to doxorubicin by adjusting HIF-‍1α. In conclusion, our study revealed that targeting TRMT5 could inhibit HCC progression and increase the susceptibility of tumor cells to chemotherapy drugs. Thus, TRMT5 might be a carcinogenesis candidate gene that could serve as a potential target for HCC therapy.


Subject(s)
Humans , Carcinoma, Hepatocellular/pathology , Cell Hypoxia , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Neoplasms/pathology , Signal Transduction/genetics , tRNA Methyltransferases/metabolism
16.
Journal of Zhejiang University. Science. B ; (12): 32-49, 2023.
Article in English | WPRIM | ID: wpr-971467

ABSTRACT

Hypoxia, as an important hallmark of the tumor microenvironment, is a major cause of oxidative stress and plays a central role in various malignant tumors, including glioblastoma. Elevated reactive oxygen species (ROS) in a hypoxic microenvironment promote glioblastoma progression; however, the underlying mechanism has not been clarified. Herein, we found that hypoxia promoted ROS production, and the proliferation, migration, and invasion of glioblastoma cells, while this promotion was restrained by ROS scavengers N-acetyl-L-cysteine (NAC) and diphenyleneiodonium chloride (DPI). Hypoxia-induced ROS activated hypoxia-inducible factor-1α (HIF-1α) signaling, which enhanced cell migration and invasion by epithelial-mesenchymal transition (EMT). Furthermore, the induction of serine protease inhibitor family E member 1 (SERPINE1) was ROS-dependent under hypoxia, and HIF-1α mediated SERPINE1 increase induced by ROS via binding to the SERPINE1 promoter region, thereby facilitating glioblastoma migration and invasion. Taken together, our data revealed that hypoxia-induced ROS reinforce the hypoxic adaptation of glioblastoma by driving the HIF-1α-SERPINE1 signaling pathway, and that targeting ROS may be a promising therapeutic strategy for glioblastoma.


Subject(s)
Humans , Cell Hypoxia , Cell Line, Tumor , Glioblastoma/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Microenvironment , Brain Neoplasms/pathology
17.
Asian Journal of Andrology ; (6): 152-157, 2023.
Article in English | WPRIM | ID: wpr-971026

ABSTRACT

Chromodomain-helicase-DNA-binding protein 1 (CHD1) deletion is among the most common mutations in prostate cancer (PCa), but its role remains unclear. In this study, RNA sequencing was conducted in PCa cells after clustered regularly interspaced palindromic repeat (CRISPR)/CRISPR-associated protein 9 (Cas9)-based CHD1 knockout. Gene set enrichment analysis (GSEA) indicated upregulation of hypoxia-related pathways. A subsequent study confirmed that CHD1 deletion significantly upregulated hypoxia-inducible factor 1α (HIF1α) expression. Mechanistic investigation revealed that CHD1 deletion upregulated HIF1α by transcriptionally downregulating prolyl hydroxylase domain protein 2 (PHD2), a prolyl hydroxylase catalyzing the hydroxylation of HIF1α and thus promoting its degradation by the E3 ligase von Hippel-Lindau tumor suppressor (VHL). Functional analysis showed that CHD1 deletion promoted angiogenesis and glycolysis, possibly through HIF1α target genes. Taken together, these findings indicate that CHD1 deletion enhances HIF1α expression through PHD2 downregulation and therefore promotes angiogenesis and metabolic reprogramming in PCa.


Subject(s)
Male , Humans , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , DNA-Binding Proteins/metabolism , Prolyl Hydroxylases/metabolism , Hypoxia , Prostatic Neoplasms/pathology , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Cell Line, Tumor , DNA Helicases/metabolism
18.
China Journal of Chinese Materia Medica ; (24): 2352-2359, 2023.
Article in Chinese | WPRIM | ID: wpr-981311

ABSTRACT

This study aims to explore the mechanism of Yanghe Decoction(YHD) against subcutaneous tumor in pulmonary metastasis from breast cancer, which is expected to lay a basis for the treatment of breast carcinoma with YHD. The chemical components of medicinals in YHD, and the targets of the components were retrieved from Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform(TCMSP) and SwissTargetPrediction. The disease-related targets were searched from GeneCards and Online Mendelian Inheritance in Man(OMIM). Excel was employed to screen the common targets and plot the Venn diagram. The protein-protein interaction network was constructed. R language was used for Gene Ontology(GO) term enrichment and Kyoto Encyclopedia of Genes and Genomes(KEGG) pathway enrichment. A total of 53 female SPF Bablc/6 mice were randomized into normal group(same volume of normal saline, ig), model group(same volume of normal saline, ig), and low-dose and high-dose YHD groups(YHD, ig, 30 days), with 8 mice in normal group and 15 mice in each of the other groups. Body weight and tumor size was measured every day. Curves for body weight variation and growth of tumor in situ were plotted. In the end, the subcutaneous tumor sample was collected and observed based on hematoxylin and eosin(HE) staining. The mRNA and protein levels of hypoxia inducible factor-1α(HIF-1α), pyruvate kinase M2(PKM2), lactate dehydrogenase A(LDHA), and glucose transporter type 1(GLUT1) were detected by PCR and Western blot. A total of 213 active components of YHD and 185 targets against the disease were screened out. The hypothesis that YHD may regulate glycolysis through HIF-1α signaling pathway to intervene in breast cancer was proposed. Animal experiment confirmed that the mRNA and protein levels of HIF-1α, PKM2, LDHA, and GLUT1 in the high-and low-dose YHD groups were lower than those in the model group. YHD has certain inhibitory effect on subcutaneous tumor in pulmonary metastasis from breast cancer in the early stage, which may intervene pulmonary metastasis from breast cancer by regulating glycolysis through HIF-1α signaling pathway.


Subject(s)
Female , Mice , Animals , Glucose Transporter Type 1/genetics , Network Pharmacology , Animal Experimentation , Saline Solution , Drugs, Chinese Herbal/therapeutic use , Medicine, Chinese Traditional , Signal Transduction , Glycolysis , RNA, Messenger , Neoplasms/drug therapy , Molecular Docking Simulation
19.
Chinese Journal of Experimental Traditional Medical Formulae ; (24): 9-17, 2023.
Article in Chinese | WPRIM | ID: wpr-979446

ABSTRACT

ObjectiveTo investigate the mechanism of Xumingtang in Gu Jin Lu Yan (《古今录验》) in regulating cell pyroptosis through the hypoxia-inducible factor-1α (HIF-1α)/NOD-like receptor pyrin domain-containing protein 3 (NLRP3) pathway in ischemic stroke (IS). MethodSD rats were randomly divided into a sham operation group, a model group, low- and high-dose Xumingtang groups, and a metformin group, with 20 rats in each group. Oral administration was performed for 3 days, and tissue samples were collected. Differential messenger RNA (mRNA) was screened using high-throughput sequencing, and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed on key differentially expressed genes. The modified neurological severity score (mNSS) and 2,3,5-triphenyltetrazolium chloride (TTC) staining were used to evaluate the effect of brain infarction. Hematoxylin-eosin (HE) staining was used for pathological morphological observation of brain tissue. Enzyme-linked immunosorbent assay (ELISA) was used to compare the levels of interleukin-1β (IL-1β) and interleukin-18 (IL-18) in the ischemic cortical region. Double staining immunohistochemistry was used to detect the co-localization of HIF-1α and NLRP3. Real-time quantitative polymerase chain reaction (PCR) was performed to detect the mRNA expression of NLRP3, HIF-1α, Caspase-1 (CASP-1), and gasdermin D (GSDMD). Western blot was used to detect the protein expression of HIF-1α, NLRP3, CASP-1, and GSDMD. ResultA total of 5 705 differentially expressed genes (2 733 downregulated and 2 972 upregulated) were obtained by mRNA sequencing. After conversion to homologous genes and intersection with the pyroptosis gene set, 95 key differentially expressed pyroptosis genes were obtained. Compared with the sham operation group, the model group showed significantly increased mNSS scores, larger brain infarction areas (P<0.01), diverse neuronal morphology, disordered arrangement, widened cell gaps, significantly increased levels of IL-1β and IL-18 in the ischemic cortical region (P<0.01), enhanced co-localization fluorescence intensity, and significantly increased mRNA and protein expression levels of HIF-1α, NLRP3, CASP-1, and GSDMD (P<0.01). Compared with the model group, the high-dose Xumingtang group showed the most significant improvement in neurological function scores and brain infarction areas (P<0.01). The neuronal integrity and arrangement were more complete, and the cell gaps were narrower in all groups with drug treatment, with significantly reduced co-localization fluorescence intensity. Xumingtang could reduce the levels of IL-1β, IL-18, and the mRNA and protein expression of HIF-1α, NLRP3, CASP-1, and GSDMD (P<0.05, P<0.01), with the high-dose Xumingtang group showing the most significant effect (P<0.01). ConclusionXumingtang in Gu Jin Lu Yan can inhibit cell pyroptosis and promote neurological function recovery after IS, which may be related to the inhibition of the HIF-1α/NLRP3 pathway.

20.
Acta Pharmaceutica Sinica ; (12): 1401-1411, 2023.
Article in Chinese | WPRIM | ID: wpr-978737

ABSTRACT

Coronary heart disease (CHD) and stroke are the most well-known cardiovascular diseases, which share many common pathological basis. Yindan Xinnaotong soft capsule (YDXNT) is a commonly used Chinese patent medicine in the treatment of stroke and CHD. However, its action of mechanism of co-treatment for stroke and CHD is still unclear. The aim of this study was to explore the common mechanism of YDXNT in co-treatment of CHD and stroke using network pharmacology, experimental verification and molecular docking. An integrated literature mining and databases of IPA, ETCM, HERB, Swiss Target Prediction, OMIM and GeneCards were used to screen and predict active ingredients and potential targets of YDXNT in co-treatment of CHD and stroke. The protein-protein interaction network, GO analysis and pathway analysis were analyzed by IPA software. The effect of YDXNT on core targets was verified by immunofluorescence. UPLC-QTOF/MS and molecular docking were used to screen and predict the main active constituents of YDXNT and their interactions with core targets. A total of 151 potential targets are predicted for YDXNT in co-treatment of CHD and stroke. Hypoxia-inducible factor-1α (HIF1α)-matrix metalloproteinase-9 (MMP9)-mediated HIF1α signaling pathway serves as one of the common mechanisms. YDXNT could reduce the increase of mitochondrial fluorescence intensity and the protein expression of HIF1α and MMP9 in HL-1 and HA induced by oxygen and glucose deprivation/reperfusion (OGD/R) in a dose-dependent manner. Baicalin may be the material basis for treating stroke and CHD with YDXNT. In conclusion, the HIF1α signaling pathway is one of the common key mechanisms of YDXNT in the co-treatment of stroke and CHD. The study provides support and basis for the in-depth scientific connotation of the traditional Chinese medicine theory of "same treatment to different diseases".

SELECTION OF CITATIONS
SEARCH DETAIL