1.Read-through circular RNA rt-circ-HS promotes hypoxia inducible factor 1α expression and renal carcinoma cell proliferation, migration and invasiveness.
Yun Yi XU ; Zheng Zheng SU ; Lin Mao ZHENG ; Meng Ni ZHANG ; Jun Ya TAN ; Ya Lan YANG ; Meng Xin ZHANG ; Miao XU ; Ni CHEN ; Xue Qin CHEN ; Qiao ZHOU
Journal of Peking University(Health Sciences) 2023;55(2):217-227
OBJECTIVE:
To identify and characterize read-through RNAs and read-through circular RNAs (rt-circ-HS) derived from transcriptional read-through hypoxia inducible factor 1α (HIF1α) and small nuclear RNA activating complex polypeptide 1 (SNAPC1) the two adjacent genes located on chromosome 14q23, in renal carcinoma cells and renal carcinoma tissues, and to study the effects of rt-circ-HS on biological behavior of renal carcinoma cells and on regulation of HIF1α.
METHODS:
Reverse transcription-polymerase chain reaction (RT-PCR) and Sanger sequencing were used to examine expression of read-through RNAs HIF1α-SNAPC1 and rt-circ-HS in different tumor cells. Tissue microarrays of 437 different types of renal cell carcinoma (RCC) were constructed, and chromogenic in situ hybridization (ISH) was used to investigate expression of rt-circ-HS in different RCC types. Small interference RNA (siRNA) and artificial overexpression plasmids were designed to examine the effects of rt-circ-HS on 786-O and A498 renal carcinoma cell proliferation, migration and invasiveness by cell counting kit 8 (CCK8), EdU incorporation and Transwell cell migration and invasion assays. RT-PCR and Western blot were used to exa-mine expression of HIF1α and SNAPC1 RNA and proteins after interference of rt-circ-HS with siRNA, respectively. The binding of rt-circ-HS with microRNA 539 (miR-539), and miR-539 with HIF1α 3' untranslated region (3' UTR), and the effects of these interactions were investigated by dual luciferase reporter gene assays.
RESULTS:
We discovered a novel 1 144 nt rt-circ-HS, which was derived from read-through RNA HIF1α-SNAPC1 and consisted of HIF1α exon 2-6 and SNAPC1 exon 2-4. Expression of rt-circ-HS was significantly upregulated in 786-O renal carcinoma cells. ISH showed that the overall positive expression rate of rt-circ-HS in RCC tissue samples was 67.5% (295/437), and the expression was different in different types of RCCs. Mechanistically, rt-circ-HS promoted renal carcinoma cell proliferation, migration and invasiveness by functioning as a competitive endogenous inhibitor of miR-539, which we found to be a potent post-transcriptional suppressor of HIF1α, thus promoting expression of HIF1α.
CONCLUSION
The novel rt-circ-HS is highly expressed in different types of RCCs and acts as a competitive endogenous inhibitor of miR-539 to promote expression of its parental gene HIF1α and thus the proliferation, migration and invasion of renal cancer cells.
Humans
;
Carcinoma, Renal Cell/pathology*
;
Cell Proliferation
;
Hypoxia
;
Kidney Neoplasms
;
MicroRNAs/genetics*
;
Neoplasm Invasiveness/genetics*
;
RNA, Circular/metabolism*
;
RNA, Small Interfering
;
Hypoxia-Inducible Factor 1, alpha Subunit/genetics*
2.Targeting TRMT5 suppresses hepatocellular carcinoma progression via inhibiting the HIF-1α pathways.
Qiong ZHAO ; Luwen ZHANG ; Qiufen HE ; Hui CHANG ; Zhiqiang WANG ; Hongcui CAO ; Ying ZHOU ; Ruolang PAN ; Ye CHEN
Journal of Zhejiang University. Science. B 2023;24(1):50-63
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.
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*
3.Effect of hypoxia on HIF -1 α/MDR1/VEGF expression in gastric cancer cells treated with 5 -fluorouracil.
Lu WANG ; Wei XING ; Jin QI ; Yongyan LU ; Linbiao XIANG ; Yali ZHOU
Journal of Central South University(Medical Sciences) 2022;47(12):1629-1636
OBJECTIVES:
Fluorouracil chemotherapeutic drugs are the classic treatment drugs of gastric cancer. But the problem of drug resistance severely limits their clinical application. This study aims to investigate whether hypoxia microenvironment affects gastric cancer resistance to 5-fluorouracil (5-FU) and discuss the changes of gene and proteins directly related to drug resistance under hypoxia condition.
METHODS:
Gastric cancer cells were treated with 5-FU in hypoxia/normoxic environment, and were divided into a Normoxic+5-FU group and a Hypoxia+5-FU group. The apoptosis assay was conducted by flow cytometry Annexin V/PI double staining. The real-time reverse transcription-polymerase chain reaction (RT-PCR) and Western blotting were used to detect the expression level of hypoxia inducible factor-1α (HIF-1α), multidrug resistance (MDR1) gene, P-glycoprotein (P-gp), and vascular endothelial growth factor (VEGF) which were related to 5-FU drug-resistance. We analyzed the effect of hypoxia on the treatment of gastric cancer with 5-FU.
RESULTS:
Compared with the Normoxic+5-FU group, the apoptosis of gastric cancer cells treated with 5-FU in the Hypoxia+5-FU group was significantly reduced (P<0.05), and the expression of apoptosis promoter protein caspase 8 was also decreased. Compared with the the Normoxic+5-FU group, HIF-1α mRNA expression in the Hypoxia+5-FU group was significantly increased (P<0.05), and the mRNA and protein expression levels of MDR1, P-gp and VEGF were also significantly increased (all P<0.05). The increased expression of MDR1, P-gp and VEGF had the same trend with the expression of HIF-1α.
CONCLUSIONS
Hypoxia is a direct influencing factor in gastric cancer resistance to 5-FU chemotherapy. Improvement of the local hypoxia microenvironment of gastric cancer may be a new idea for overcoming the resistance to 5-FU in gastric cancer.
Humans
;
Fluorouracil/therapeutic use*
;
Vascular Endothelial Growth Factor A/metabolism*
;
Stomach Neoplasms/drug therapy*
;
Drug Resistance, Multiple
;
Vascular Endothelial Growth Factors/metabolism*
;
Hypoxia
;
ATP Binding Cassette Transporter, Subfamily B/genetics*
;
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics*
;
Cell Line, Tumor
;
Cell Hypoxia
;
RNA, Messenger/metabolism*
;
Hypoxia-Inducible Factor 1, alpha Subunit/genetics*
;
Tumor Microenvironment
4.Danlou Tablet Improves Chronic Intermittent Hypoxia-Induced Dyslipidemia and Arteriosclerosis by HIF-1α-Angptl4 mRNA Signaling Pathway.
Jing-Jing TANG ; Guang-Xi LI ; Zhi-Guo LIU ; Rong YI ; Dong YU ; Yue-Bo ZHANG ; Shuang-Qiao ZHAO ; Shi-Han WANG
Chinese journal of integrative medicine 2022;28(6):509-517
OBJECTIVE:
To detect whether Danlou Tablet (DLT) regulates the hypoxia-induced factor (HIF)-1α-angiopoietin-like 4 (Angptl4) mRNA signaling pathway and explore the role of DLT in treating chronic intermittent hypoxia (CIH)-induced dyslipidemia and arteriosclerosis.
METHODS:
The mature adipocytes were obtained from 3T3-L1 cell culturation and allocated into 8 groups including control groups (Groups 1 and 5, 0.1 mL of cell culture grade water); DLT groups (Groups 2 and 6, 0.1 mL of 1,000 µg/mL DLT submicron powder solution); dimethyloxalylglycine (DMOG) groups (Groups 3 and 7, DMOG and 0.1 mL of cell culture grade water); DMOG plus DLT groups (Groups 4 and 8, DMOG and 0.1 mL of 1,000 µg/mL DLT submicron powder solution). Groups 1-4 used mature adipocytes and groups 5-8 used HIF-1 α-siRNA lentivirus-transfected mature adipocytes. After 24-h treatment, real-time polymerase chain reaction and Western blot were employed to determine the mRNA and protein expression levels of HIF-1 α and Angptl4. In animal experiments, the CIH model in ApoE-/- mice was established. Sixteen mice were complete randomly divided into 4 groups including sham group, CIH model group [intermittent hypoxia and normal saline (2 mL/time) gavage once a day]; Angptl4 Ab group [intermittent hypoxia and Angptl4 antibody (30 mg/kg) intraperitoneally injected every week]; DLT group [intermittent hypoxia and DLT (250 mg/kg) once a day], 4 mice in each group. After 4-week treatment, enzyme linked immunosorbent assay was used to detect the expression levels of serum total cholesterol (TC) and triglyceride (TG). Hematoxylin-eosin and CD68 staining were used to observe the morphological properties of arterial plaques.
RESULTS:
Angptl4 expression was dependent on HIF-1 α, with a reduction in mRNA expression and no response in protein level to DMOG or DLT treatment in relation to siHIF-1 α -transfected cells. DLT inhibited HIF-1 α and Angptl4 mRNA expression (P<0.05 or P<0.01) and reduced HIF-1 α and Angptl4 protein expressions with DMOG in mature adipocytes (all P<0.01), as the effect on HIF-1 α protein also existed in the presence of siHIF-1 α (P<0.01). ApoE-/- mice treated with CIH had increased TG and TC levels (all P<0.01) and atherosclerotic plaque. Angptl4 antibody and DLT both reduce TG and TC levels (all P<0.01), as well as reducing atherosclerotic plaque areas, narrowing arterial wall thickness and alleviating atherosclerotic lesion symptoms to some extent.
CONCLUSION
DLT had positive effects in improving dyslipidemia and arteriosclerosis by inhibiting Angptl4 protein level through HIF-1 α-Angptl4 mRNA signaling pathway.
Angiopoietin-Like Protein 4/genetics*
;
Animals
;
Apolipoproteins E
;
Atherosclerosis/metabolism*
;
Drugs, Chinese Herbal
;
Dyslipidemias/genetics*
;
Hypoxia/metabolism*
;
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism*
;
Mice
;
Plaque, Atherosclerotic
;
Powders
;
RNA, Messenger/genetics*
;
Signal Transduction
;
Triglycerides
;
Water
5.Effects of intermittent hypoxia stimulation with different frequencies on HT22 cell viability and expression of Hif-1α and p-NF-κB.
Sheng-Chang YANG ; Yang ZHAO ; Yu-Ying ZHENG ; Wen-Ya LI ; Ming ZHAO ; En-Sheng JI
Acta Physiologica Sinica 2021;73(1):26-34
Intermittent hypoxia (IH) could induce cognitive impairment through oxidative stress and inflammation. However, the degree of cell damage is closely related to the IH stimulus frequency. IH stimulation with different frequencies also induces opposite results on neuronal cell lines. Therefore, this study was aimed to compare the effects of IH stimulation with three different frequencies on murine hippocampal neuronal HT22 cell activity, and to explore the molecular mechanism of the IH stimulus frequency-related neuron injury. HT22 cells were cultured and divided into control group and three IH stimulation groups with different frequencies. Oxygen concentration in the chamber was circulated between 21% and 1% (IH1 group, 6 cycles/h; IH2 group, 2 cycles/h; IH3 group, 0.6 cycle/h). Cell morphology was observed at 6, 12, 24 and 48 h of IH treatment. Cell viability was determined by the CCK-8 kit, lactate dehydrogenase (LDH) content in cell supernatant was determined by LDH kit, oxidative stress level was detected by the reactive oxygen species (ROS) probe, and protein expression levels of hypoxia inducible factor-1α (Hif-1α) and phosphorylated nuclear factor κB (p-NF-κB) were detected by Western blot. The results showed that, compared with control group, cell number and activity in the three IH groups were decreased, LDH content and ROS levels were increased with the prolongation of IH stimulation time, and the changes were most obvious in the IH1 group among those of the three IH groups. Hif-1α expression and the p-NF-κB/NF-κB ratio were also up-regulated with the prolongation of IH stimulation time, and the changes of IH1 group were the most significant. These results suggest that IH stimulation induces oxidative stress injury in HT22 cells, which is related to increased Hif-1α expression and NF-κB phosphorylation. Moreover, the higher frequency of IH stimulation induces more serious cell injury.
Animals
;
Cell Hypoxia
;
Cell Survival
;
Hypoxia
;
Hypoxia-Inducible Factor 1, alpha Subunit/genetics*
;
Mice
;
NF-kappa B/metabolism*
;
Oxidative Stress
;
Reactive Oxygen Species
6.Salt-inducible kinase 2 regulates energy metabolism in rats with cerebral ischemia-reperfusion.
Ran ZHANG ; Yun LIU ; Cui ZHANG ; Mengyao MA ; Shu LI ; Yun HONG
Journal of Zhejiang University. Medical sciences 2021;50(3):352-360
To investigate the effects of salt-inducible kinase 2 (SIK2) on energy metabolism in rats with cerebral ischemia-reperfusion. Adult SD male rats were divided into 5 groups: sham group, ischemia group, reperfusion group, adenovirus no-load group, and SIK2 overexpression group with 5 animals in each group. The middle cerebral artery occlusion (MCAO) was induced with the modified Zea-Longa line thrombus method to establish the cerebral ischemia reperfusion model. Eight days before the MCAO, SIK2 overexpression was induced by injecting 7 μL adenovirus in the right ventricle, then MCAO was performed for followed by reperfusion HE staining was used to observe the pathological changes of cerebral tissue in rats; TTC staining was used to observe the volume of cerebral infarct. The levels of adenosine triphosphate (ATP) and adenosine diphosphate (ADP) in rat brain tissue were detected by ELISA; the levels of SIK2 and hypoxia-inducible factor 1α (HIF-1α) in the rat brain tissues were detected by RT-qPCR and Western blotting. Compared with the sham group, SIK2 level was decreased in the ischemia group, and it was further declined in the reperfusion group (<0.05). Compared with the sham group and ischemic group, the pathological injury in reperfusion group were more severe, and the infarct size was larger; compared with the reperfusion group and adenovirus no-load group, the pathological injury of the SIK2 overexpression group was milder, and the infarct size is less. Compared with the sharn group, HIF-1α was increased in both ischemia group and reperfusion group, especially in ischemia group (all <0.05); HIF-1α level in the SIK2 overexpression group was higher than that in the reperfusion group and adenovirus no-load group (all <0.05). ATP level in ischemia group and reperfusion group was lower than that in the sham group, and the reperfusion group decreased more significantly than the ischemia group (<0.05); ADP content was increased in the ischemia and reperfusion group, and the ADP content in reperfusion group was significantly higher than that in the ischemia group (<0.05). ATP level in the SIK2 overexpression group was higher than that in the reperfusion group and adenovirus no-load group (all <0.05), and ADP was decreased in the SIK2 overexpression group (all <0.05). SIK2 can up-regulate the ATP level and down-regulate the ADP level in rat brain tissue and alleviate cerebral ischemia-reperfusion injury by increase the level of HIF-1α.
Animals
;
Brain Ischemia
;
Energy Metabolism
;
Hypoxia-Inducible Factor 1, alpha Subunit/genetics*
;
Infarction, Middle Cerebral Artery
;
Male
;
Protein-Serine-Threonine Kinases
;
Rats
;
Rats, Sprague-Dawley
;
Reperfusion
;
Reperfusion Injury
7.Effects of PGC1
Jungang NIE ; Na TA ; Lijuan LIU ; Guoxiang SHI ; Ting KANG ; Zeqi ZHENG
Journal of Central South University(Medical Sciences) 2020;45(10):1155-1163
OBJECTIVES:
Peroxisome proliferator-activated receptor gamma coactivator 1α (PGC1α) controls mitochondrial biogenesis, but its role in cardiovascular diseases is unclear. The purpose of this study is to explore the effect of PGC1α on myocardial ischemia-reperfusion injury and the underlying mechanisms.
METHODS:
The transverse coronary artery of SD rat was ligated for 30 minutes followed by 2 hours of reperfusion. Triphenyltetrazolium chloride (TTC) staining was performed to measure the area of myocardial infarction. Immunohistochemistry and Western blotting were used to detect the PGC1α expression in myocardium. The rat cardiomyocyte H9C2 was subjected to hypoxia/reoxygenation (H/R) with the knockdown of PGC1α or hypoxia- inducible factor 1α (HIF-1α), or with treatment of metformin. Western blotting was used to detect the expression of PGC1α, HIF-1α, p21, BAX, and caspase-3. CCK-8 was performed to detect cell viability, and flow cytometry was used to detect apoptosis and mitochondrial superoxide (mitoSOX) release. RT-qPCR was used to detect the mRNA expression of PGC1α and HIF-1α. Besides, chromatin immunoprecipitation (ChIP)-qPCR and luciferase reporter gene assay were applied to detect the transcriptional regulation effect of HIF-1α on PGC1α.
RESULTS:
After I/R, the PGC1α expression was increased in infarcted myocardium. H/R induced H9C2 cell apoptosis (
CONCLUSIONS
After I/R, HIF-1α up-regulates the expression of PGC1α, leading to an increase in ROS production and aggravation of injury. Metformin can inhibit the accumulation of HIF-1α during hypoxia and effectively protect myocardium from ischemia/reperfusion injury.
Animals
;
Apoptosis
;
Hypoxia-Inducible Factor 1, alpha Subunit/genetics*
;
Myocardial Reperfusion Injury/genetics*
;
Myocytes, Cardiac/metabolism*
;
Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism*
;
Rats
;
Rats, Sprague-Dawley
;
Reperfusion Injury
8.Expression and significance of hypoxia-inducible factor-1α and vascular endothelial growth factor and receptor 2 in stage 3 pressure injury of rats.
Xiao-Hui WANG ; Xiao-Ping CHEN ; Hong-Ping WANG ; Ying-Ying PAN ; Li-Ping JIANG
Chinese Journal of Applied Physiology 2019;35(3):199-203
OBJECTIVE:
To analyze the expression and relationship of hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF) and kinase insert domain receptor (KDR) in local skin tissues of pressure injury and investigate the possible mechanism of stage 3 pressure injury refractory wound.
METHODS:
Forty male SD rats were randomly divided into normal control group, compressed 3 d, 5 d, 7 d, and 9 d groups. Stage 3 pressure injury animal model were established by magnet compression. The morphology of skin was observed by HE staining. The expression of VEGF was detected by immunohistochemistry. The expression levels of HIF-1α, VEGF and KDR protein in skin tissue were detected by Western blot. One-way analysis of variance and LSD test were performed on the data.
RESULTS:
①The HE results showed that compared with the normal control group, the epidermis of the compressed group was gradually thickened, the number of blood vessels was decreased, the collagen arrangement disordered and inflammatory cells infiltration were increased. ②Immunohistochemical results showed that the expression of VEGF protein in the 3 d group was significantly higher than that in the normal control group (P<0.01). The expression of VEGF protein in the skin tissue of 5 d, 7 d and 9 d groups was lower than that in normal control group (P<0.05). WB results were consistent with immunohistochemistry results. ③WB results showed that the expression of HIF-1α in the skin tissues of the rats in 3 d, 5 d and 7 d groups was higher than that in the normal control group (P<0.01 or P<0.05). The expression of KDR protein was lower than that of the normal control group (P<0.05 or P<0.01).
CONCLUSION
HIF-1α mediated reduction of VEGF and KDR protein expression and decreased tissue angiogenesis may be one of the important causes of chronic dysfunction of stage 3 pressure injury.
Animals
;
Hypoxia-Inducible Factor 1, alpha Subunit
;
genetics
;
metabolism
;
Male
;
Pressure
;
adverse effects
;
Random Allocation
;
Rats
;
Rats, Sprague-Dawley
;
Skin
;
injuries
;
Vascular Endothelial Growth Factor A
;
genetics
;
metabolism
;
Vascular Endothelial Growth Factor Receptor-2
;
genetics
;
metabolism
9.Role of cytoplasmic lncRNAs in regulating cancer signaling pathways.
Pei-Fen FU ; Xin ZHENG ; Xiao FAN ; Ai-Fu LIN
Journal of Zhejiang University. Science. B 2019;20(1):1-8
Cancer remains a serious healthcare problem despite significant improvements in early detection and treatment approaches in the past few decades. Novel biomarkers for diagnosis and therapeutic strategies are urgently needed. In recent years, long noncoding RNAs (lncRNAs) have been reported to be aberrantly expressed in tumors and show crosstalk with key cancer-related signaling pathways. In this review, we summarized the current progress of research on cytoplasmic lncRNAs and their roles in regulating cancer signaling and tumor progression, further characterization of which may lead to effective approaches for cancer prevention and therapy.
Animals
;
Biomarkers, Tumor/metabolism*
;
Cytoplasm/metabolism*
;
Hippo Signaling Pathway
;
Humans
;
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism*
;
Neoplasms/metabolism*
;
Phosphatidylinositol 3-Kinases/metabolism*
;
Protein Serine-Threonine Kinases/metabolism*
;
Proto-Oncogene Proteins c-akt/metabolism*
;
RNA, Long Noncoding/metabolism*
;
Signal Transduction/genetics*
10.Expression of hypoxia-inducible factor 1α is associated with lymph node metastasis in oral squamous cell carcinoma.
Jian Nan LI ; Zhi En FENG ; Lin WANG ; Yi Xiang WANG ; Chuan Bin GUO
Journal of Peking University(Health Sciences) 2018;50(1):26-32
OBJECTIVE:
To explore the association between hypoxia-inducible factor 1α (HIF-1α) expression and lymph node metastasis in oral squamous cell carcinoma (OSCC).
METHODS:
Tumor specimens from 125 patients with histologically-proven, surgically-treated OSCC were examined by immunohistochemical staining for expression of HIF-1α. The patients were divided into two groups by the expression of HIF-1α, high expression of HIF -1α group (H-group) and low expression of HIF-1α group (L-group). The main assessment parameters were lymph node metastasis rate and disease-specific survival (DSS). The lymph node metastasis rate and clinicopathologic features were compared using Mann-Whitney test. The Kaplan-Meier curve was generated for each group and compared using the log-rank test. Cox proportional hazard models were utilized for multivariate analyses of HIF-1α expression and other baseline factors with DSS. All calculations and analyses were performed using the SPSS 17.0 software package.
RESULTS:
The protein expression levels of HIF-1α were up-regulated in OSCC and two patients were unable to evaluate. There were 48 patients in L-group and 75 patients in H-group. Lymph node metastasis rate was 37.5% (18/48) for L-group and 58.7% (44/75) for H-group (P=0.027). Expression of HIF-1α was significantly correlated with lymph node metastasis. The patients of L-group had a significantly better DSS than the patients of H-group (70.8% vs. 46.7%, P=0.005), while the patients of L-group had a significantly better disease-free survival (DFS) than the patients of H-group (60.4% vs. 36.0%, P=0.009) by Kaplan-Meier method. A multivariate survival analysis also showed that HIF-1α expression (HR=2.164, 95%CI: 1.150-4.074, P=0.017) and T-stage (HR=1.387, 95%CI: 1.066-1.804, P=0.015) both were the independent factors associated with prognosis.
CONCLUSION
HIF-1α expression is significantly correlated with lymph node metastasis in OSCC. HIF-1α expression is an independent predictive factor for prognosis of OSCC patients, and may serve as a potential biomarker for molecular diagnosis and targeted therapy in future.
Biomarkers/metabolism*
;
Carcinoma, Squamous Cell/pathology*
;
Humans
;
Hypoxia
;
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism*
;
Immunohistochemistry
;
Lymph Nodes
;
Lymphatic Metastasis/genetics*
;
Mouth Neoplasms/pathology*
;
Prognosis

Result Analysis
Print
Save
E-mail