Hechao Zhou's research while affiliated with First Affiliated Hospital of China Medical University and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (7)


Development and internal validation of a nomogram based on peripheral blood inflammatory markers for predicting prognosis in nasopharyngeal carcinoma
  • Article
  • Full-text available

March 2024

·

3 Reads

Cancer Medicine

Cancer Medicine

Jing Lai

·

Peixin Lin

·

Jiafeng Zhuang

·

[...]

·

Jing Huang

Background Inflammatory markers, including the product of neutrophil count, platelet count, and monocyte count divided by lymphocyte count (PIV) and the platelet‐to‐white blood cell ratio (PWR), have not been previously reported as prognostic factors in nasopharyngeal carcinoma (NPC) patients. In order to predict overall survival (OS) in NPC patients, our goal was to create and internally evaluate a nomogram based on inflammatory markers (PIV, PWR). Methods A retrospective study was done on patients who received an NPC diagnosis between January 2015 and December 2018. After identifying independent prognostic indicators linked to OS using Cox proportional hazards regression analysis, we created a nomogram with the factors we had chosen. Results A total of 630 NPC patients in all were split into training ( n = 441) and validation sets ( n = 189) after being enrolled in a population‐based study in 2015–2018 and monitored for a median of 5.9 years. In the training set, the age, PIV, and PWR, selected as independent predictors for OS via multivariate Cox's regression model, were chosen to develop a nomogram. Both training and validation cohorts had C‐indices of 0.850 (95% confidence interval [CI]: 0.768–0.849) and 0.851 (95% CI: 0.765–0.877). Furthermore, compared with traditional TNM staging, our nomogram demonstrated greater accuracy in predicting patient outcomes. The risk stratification model derived from our prediction model may facilitate personalized treatment strategies for NPC patients. Conclusion Our findings confirmed the prognostic significance of the PWR and PIV in NPC. High PIV levels (>363.47) and low PWR (≤36.42) values are associated with worse OS in NPC patients.

Download
Share

The flowchart of the research. GBM glioblastoma, LGG low grade glioma, TCGA The Cancer Genome Atlas, DEGs differentially expressed genes, GO Gene Ontology, KEGG Kyoto Encyclopedia of Genes and Genomes, lncRNAs long noncoding RNAs, ORLs oxidative stress-related lncRNAs, ROC receiver operating characteristic, GSEA gene set enrichment analysis, CGGA Chinese Glioma Genome Atlas.
Volcano plot, LASSO regression analysis and gene association network of 6-ORLs. (A) Volcano plot of 120 OR-DEGs in TCGA glioma cohort. (B) The partial likelihood deviance with changing of log(λ). (C) LASSO coefficient profiles of oxidative stress-associated lncRNAs. (D) Sankey diagram of 6-ORLs, OR-DEGs and risk type. (E) Relationship network of 6-ORLs and OR-DEGs.
Validation of 6-ORLs prognostic signature in TCGA cohort. (A) Distribution of risk scores. (B) Survival status of patients with different risk scores. (C) Kaplan–Meier survival curve for different risk subgroups. (D) Time-dependent ROC curves and (E) Clinicopathological variables ROC curves for 6-ORLs prognostic signature. (F–G) Hazard ratio distributions of risk scores and clinicopathological variables in (F) univariate and (G) multivariate Cox regressions.
Nomogram and internal validation of TCGA cohorts for 6-ORLs prognostic signature. (A) Nomogram of risk scores and clinicopathological variables. (B) Online version of Nomogram (https://dnszy.shinyapps.io/Glioma_OS_lncRNA/). (C) Decision curve analysis (DCA) of Nomogram. (D–F) Calibration curves of Nomogram for 1, 3 and 5 years. (G–N) The risk score, patient survival status distribution, and Kaplan–Meier survival curve as well as ROC curves of 6-ORLs prognostic signature in the TCGA training cohort (G–J) and validation cohort (K–N).
Differences in ssGSEA immune infiltration analysis and immune checkpoint-associated gene expression in high- and low-risk subgroups. (A) Immune cell infiltration analysis. (B) Immune-related functional analysis. (C) Differential expression of immune checkpoint-associated genes. *P < 0.05; **P < 0.01; ***P < 0.001; ns non-significant.

+6

Comprehensive analysis of oxidative stress-related lncRNA signatures in glioma reveals the discrepancy of prognostic and immune infiltration

May 2023

·

15 Reads

·

1 Citation

Scientific Reports

Oxidative stress refers to the process of reactive oxide species (ROS) increase in human body due to various factors, which leads to oxidative damage in human tissues. Current studies have confirmed that sustained oxidative stress is one of the distinctive features throughout the development of tumors. Numerous reports have shown that lncRNAs can regulate the process of oxidative stress through multiple pathways. However, the relationship between glioma-associated oxidative stress and lncRNAs is not clearly investigated. RNA sequencing data of GBM (glioblastoma) and LGG (low grade glioma) and corresponding clinical data were retrieved from the TCGA database. Oxidative stress related lncRNAs (ORLs) were identified by Pearson correlation analysis. Prognostic models for 6-ORLs were structured in the training cohort by univariate Cox regression analysis, multivariate Cox regression analysis and LASSO regression analysis. We constructed the nomogram and verified its predictive efficacy by Calibration curves and DCA decision curves. The biological functions and pathways of 6-ORLs-related mRNAs were inferred by Gene Set Enrichment Analysis. Immune cell abundance and immune function associated with risk score (RS) were estimated by ssGSEA, CIBERSORT and MCPcounter synthetically. External validation of the signature was completed using the CGGA-325 and CGGA-693 datasets. 6-ORLs signature—AC083864.2, AC107294.1, AL035446.1, CRNDE, LINC02600, and SNAI3-AS1—were identified through our analysis as being predictive of glioma prognosis. Kaplan–Meier and ROC curves indicated that the signature has a dependable predictive efficacy in the TCGA training cohort, validation cohort and CGGA-325/CGGA-693 test cohort. The 6-ORLs signature were verified to be independent prognostic predictors by multivariate cox regression and stratified survival analysis. Nomogram built with risk scores had strong predictive efficacy for patients' overall survival (OS). The outcomes of the functional enrichment analysis revealing potential molecular regulatory mechanisms for the 6-ORLs. Patients in the high-risk subgroup presented a significant immune microenvironment of macrophage M0 and cancer-associated fibroblast infiltration which was associated with a poorer prognosis. Finally, the expression levels of 6-ORLs in U87/U251/T98/U138 and HA1800 cell lines were verified by RT-qPCR. The nomogram in this study has been made available as a web version for clinicians. This 6-ORLs risk signature has the capabilities to predict the prognosis of glioma patients, assist in evaluating immune infiltration, and assess the efficacy of various anti-tumor systemic therapy regimens.


Erastin decreases radioresistance of NSCLC cells partially by inducing GPX4‑mediated ferroptosis

January 2019

·

52 Reads

·

94 Citations

Oncology Letters

The aim of the present study was to examine whether erastin influences radioresistance in non-small cell lung cancer (NSCLC) cells and produce a preliminary investigation into its mechanism of action. The radioresistant subtype of NSCLC cells, A549-R and H460-R, were induced by high-dose hypofractionated irradiation. Erastin was used to treat the radioresistant cells and radiosensitivity was examined by colony formation assays. Cell death was determined after the cells were treated with erastin, irradiation (IR) or erastin together with IR. The expression of glutathione peroxidase 4 (GPX4) expression in the parental cells and radioresistance cells was detected by western blotting. GPX4 expression in the radioresistance cells was subsequently inhibited, radiosensitivity and cell death was measured, and erastin enhanced radiosensitivity in A549-R and H460-R cells. Erastin and IR exhibited a combined effect on killing cells, as co-treatment with erastin and IR demonstrated a higher effect on killing cells compared with erastin or IR alone. GPX4 expression was inhibited by erastin in the radioresistant cells. Inhibiting GPX4 expression also radiosensitized NSCLC cells to radiation in the radioresistant cell lines. Erastin-induced and GPX4-inhibition-induced cell death could partially be rescued by deferoxamine, but not Z-VAD-FMK and olaparib, which indicated that erastin and GPX4-inhibition induced ferroptosis in the radioresistant cells. Erastin decreased radioresistance of NSCLC cells partially by inducing GPX4-mediated ferroptosis.


Niclosamide sensitizes nasopharyngeal carcinoma to radiation by downregulating Ku70/80 expression

February 2018

·

122 Reads

·

9 Citations

Journal of Cancer

The aim of the present study was to investigate whether niclosamide could sensitize the nasopharyngeal carcinoma cells to radiation and further explore the underlying mechanisms. CCK-8 assay was used to determine the effect of niclosamide on the proliferation of NPC cells. Colony formation assay was used to evaluate the radiosensitizing effect of niclosamide on NPC cells. Flow cytometry analysis was used to determine the apoptosis of NPC cells induced by niclosamide. Immunofluorescent staining was used to detect the formation of γ-H2AX foci and the localization of Ku70/80 proteins in NPC cells. Real-time PCR quantification analysis was used to examine the level of Ku70/80 mRNA. DNA damage repair-related proteins were detected by western blot analysis. Our results showed that niclosamide markedly suppressed the proliferation of NPC cells. Niclosamide pretreatment followed by irradiation reduced the colony forming ability of NPC cells. Niclosamide in combination with irradiation significantly increased the apoptotic rate of NPC cells. Niclosamide significantly reduced the transcriptional level of K70/80 but not the translocation of Ku70/80 protein induced by irradiation. In conclusion, our study demonstrated that niclosamide could inhibit the growth of NPC cells and sensitize the NPC cells to radiation via suppressing the transcription of Ku70/80.


Niclosamide enhances the cytotoxic effect of cisplatin in cisplatin-resistant human Lung cancer cells via suppression of Lung resistance-related protein and c-myc

December 2017

·

168 Reads

·

24 Citations

Molecular Medicine Reports

Lung cancer is a leading cause of cancer-associated mortality worldwide. The cisplatin (DDP)‑based chemotherapy remains the foundation of treatment for the majority of patients affected by advanced non‑small cell lung cancer (NSCLC). However, DDP‑resistance limits the clinical utility of this drug in patients with advanced NSCLC. The aim of the present study was to investigate the inhibitory effect of niclosamide on human lung cancer cell growth and to investigate the possible underlying mechanism. The effects of niclosamide on the proliferation of human lung adenocarcinoma (A549) and DDP‑resistant (CR) human lung adenocarcinoma (A549/DDP) cells were examined by Cell Counting kit‑8 assay. The impact of niclosamide on the apoptosis of A549/DDP cells was detected by Annexin V‑fluorescein isothiocyanate/propidium iodide assay. The expression levels of cisplatin‑resistant‑associated molecules (lung resistance‑related protein and c‑myc) following niclosamide treatment in A549/DDP cells were evaluated by western blot analysis. The results indicated that niclosamide in combination with DDP demonstrated a synergistic effect in A549/DDP cells and directly induced apoptosis, which may be associated with caspase‑3 activation. Furthermore, niclosamide decreased the expression level of c‑myc protein, which may influence DDP sensitivity of A549/DDP cells. Thus, the present study indicates that niclosamide combined with DDP exerts a synergistic effect in cisplatin‑resistant lung cancer cells and may present as a promising drug candidate in lung cancer therapy.


Figure 1 of 2
Figure 2 of 2
Niclosamide enhances the antitumor effects of radiation by inhibiting the hypoxia-inducible factor-1α/vascular endothelial growth factor signal

June 2017

·

47 Reads

·

11 Citations

Oncology Letters

Lung cancer is one of the leading causes of cancer-associated mortality, worldwide. The overall survival rate remains low, but progress has been made in improving the diagnosis and treatment of lung cancer over the past decades. Niclosamide, a salicylanilide derivative used for the treatment of tapeworm infections, is safe, well tolerated, inexpensive and readily available. Previous studies have identified niclosamide as a potential anticancer agent. The present study demonstrated that niclosamide enhanced the effect of irradiation by inhibiting the hypoxia-inducible factor-1α/vascular endothelial growth factor signaling pathway. These findings suggest that niclosamide may be a promising candidate for clinical evaluation as part of a combined regimen for the treatment of non-small cell lung cancer.


Figure 2 ga induces g0/g1 cell cycle arrest in a549 (A) and a549/DDP (B) cells. Notes: a549 and a549/DDP cells were treated with 0, 1.0, and 2.0 μM GA for 24 hours. After staining with propidium iodide, cell cycle stage was analyzed by flow cytometry. The cell cycle distribution was quantified in GA-treated and -untreated control cells. All data are presented as the mean ± sD of at least three independent experiments. **P,0.01 vs control cells; bars, sD. Abbreviations: ga, gambogic acid; sD, standard deviation; Pi, propium iodide.
Figure 6 ga downregulates the protein expression of MrP2 and lrP in a dose-dependent way. Notes: (A) The basic protein expression of MrP2 and lrP in untreated a549 and a549/DDP cells. (B) cells were treated with ga at the indicated concentrations (0, 0.5, 1.0, 2.0, and 4.0 μM) for 24 hours. The levels of MrP2 and lrP were determined by Western blot analysis. Data are shown as the mean values ± sD from three independent experiments. **P,0.01; bars, sD. Abbreviations: ga, gambogic acid; lrP, lung resistance protein; MrP2, multidrug resistance-associated protein 2; sD, standard deviation.
Combination of gambogic acid with cisplatin enhances the antitumor effects on cisplatin-resistant lung cancer cells by downregulating MRP2 and LRP expression

June 2016

·

32 Reads

·

26 Citations

Cisplatin resistance is a main clinical problem of lung cancer therapy. Gambogic acid (GA) could prohibit the proliferation of a variety of human cancer cells. However, the effects of GA on cisplatin-resistant lung cancer are still unclear. The objective of the present study was to find out the antitumor effects of GA on cisplatin-resistant human lung cancer A549/DDP cells and further explore its underlying mechanisms. Cell Counting Kit-8 assay was used to observe the impacts of GA and/or cisplatin on the proliferation of lung cancer cells; flow cytometry was used to detect the effects of GA on cell cycle and apoptosis; Western blot was used to examine the effects of GA on the expression of lung resistance protein (LRP) and multidrug resistance-associated protein 2 (MRP2) protein in A549/DDP cells. Our results showed that GA dose- and time-dependently prohibited the proliferation and induced significant cell apoptosis in A549 and A549/DDP cells. GA also induced G0/G1 arrest in both A549/DDP and A549 cells. Moreover, GA upregulated protein expression level of cleaved caspase-3 and Bax and downregulated protein expression level of pro-caspase-9 and Bcl-2 in time- and dose-dependent way in A549/DDP cells. GA combined with cisplatin enhanced the cells apoptotic rate and reduced the cisplatin resistance index in A549/DDP cells. In addition, GA reduced the MRP2 and LRP protein expression level in A549/DDP cells. GA inhibits the proliferation, induces cell cycle arrest and apoptosis in A549/DDP cells. Combination of GA with cisplatin enhances the antitumor effects on cisplatin-resistant lung cancer cells by downregulating MRP2 and LRP expression.

Citations (6)


... Glioblastoma (GBM) is the most prevalent form of glioma and the most common primary intracranial tumor [1]. Despite multiple treatment options available, such as surgical resection, chemotherapy, and radiotherapy, the prognosis for GBM patients is very unfavorable, with a median survival of only 15 months [2]. ...

Reference:

Bioinformatics Analysis and Experimental Validation for Exploring Key Molecular Markers for Glioblastoma
Comprehensive analysis of oxidative stress-related lncRNA signatures in glioma reveals the discrepancy of prognostic and immune infiltration

Scientific Reports

... After the application of ionizing radiation, the rays cause the emergence of free radicals through the radiolysis of water to control cell proliferation [59]. Many studies have determined that sorafenib and sulfasalazine, which are among the FDA-approved chemotherapeutics, stimulate the ferroptosis mechanism with radiation application in different types of cancer such as lung cancer, melanoma and breast cancer [60][61][62]. It has been revealed in studies in the literature that RSL3 and erastin, which are ferroptosis inducers, increase the radiation efficiency in different cancer types [43,62]. ...

Erastin decreases radioresistance of NSCLC cells partially by inducing GPX4‑mediated ferroptosis
  • Citing Article
  • January 2019

Oncology Letters

... In chronic myeloid leukemia (CML), NIC inhibits the proliferation of CML cells by downregulating the expression of the signaling molecules STAT5 and Akt [39]. In NPC, NIC inhibits cell proliferation by downregulating ku70/80 expression, thereby increasing the radiotherapy sensitivity of NPC cells [40]. NIC inhibits ...

Niclosamide sensitizes nasopharyngeal carcinoma to radiation by downregulating Ku70/80 expression

Journal of Cancer

... We further demonstrated that MYC is the downstream target of niclosamide in osteosarcoma, most likely via the β-catenin/Myc axis. This is supported by previous findings that niclosamide dysregulates MYC in oral squamous cell carcinoma and lung cancer (Wang LH et al., 2018;Zuo et al., 2018). Given the important role of β-catenin in cancer stem cells, it is likely that niclosamide also targets osteosarcoma stem cells which serve as a reservoir for osteosarcoma relapse. ...

Niclosamide enhances the cytotoxic effect of cisplatin in cisplatin-resistant human Lung cancer cells via suppression of Lung resistance-related protein and c-myc

Molecular Medicine Reports

... Owing to its weak solubility in water, Nic was dissolved in a solvent mixture of 3/15/82 (v/v/v) dimethyl sulfoxide (DMSO)/Cremophor EL/water for animal administration [28]. Several studies have demonstrated that Nic uncouples oxidative phosphorylation and reduces ATP levels in various tumor cells, including CRC cells [29,30]. Nic can indirectly inhibit DNA DSB (DNA doublestrand break) repair [31]. ...

Niclosamide enhances the antitumor effects of radiation by inhibiting the hypoxia-inducible factor-1α/vascular endothelial growth factor signal

Oncology Letters

... As reported, treatment with oxaliplatin at 1, 2, and 4µM concentration exhibited platinum levels of ~0.25, 0.45, and 0.48 ng/10 7 cells, while co-treatment with GA reduced the oxaliplatin concentration almost 4-folds and the platinum levels increased to 0.45, 0.8, and 1.6ng/10 7 cells in a time-dependent manner [60][61][62][63][64][65][66]. When GA was used in combination with cisplatin, it showed higher anticancer activity by downregulation of LRP and MRP2 proteins, thereby promoting cell cycle arrest and increased apoptosis in cisplatin-resistant (A549/DDP) lung cancer cells [67]. GA when combined with cisplatin and rapamycin induced autophagy which ultimately suppressed the Akt/mTOR signaling probably by synergistic effect. ...

Combination of gambogic acid with cisplatin enhances the antitumor effects on cisplatin-resistant lung cancer cells by downregulating MRP2 and LRP expression
OncoTargets and Therapy

OncoTargets and Therapy