1
|
Wei S, Xu G, Zhao S, Zhang C, Feng Y, Yang W, Lu R, Zhou J, Ma Y. EGR2 promotes liver cancer metastasis by enhancing IL-8 expression through transcription regulation of PDK4 in M2 macrophages. Int Immunopharmacol 2025; 153:114484. [PMID: 40139095 DOI: 10.1016/j.intimp.2025.114484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025]
Abstract
Liver tumor is a common digestive system tumor, and its development is closely related to complex cytokines, tumor microenvironment and immunoregulatory mechanisms. Tumor-associated macrophages play a great role in a series of liver cancer development by secreting various cytokines and transmitting multiple signals, but how macrophages regulate the various biological behaviors of liver cancer cells at the microscopic level is a challenge we still need to overcome. In this research, we first identified the Early Growth Response 2 (EGR2) gene, which exhibited significant expression in M2 macrophages in comparison to M0 and M1 cell types, utilizing RNA sequencing. Subsequently, we validated this finding through a battery of methodologies, including WB, qRT-PCR, and immunofluorescence assays. We further employed a co-culture model involving MHCC97L and macrophages to investigate the impact of EGR2 downregulation within M2 cells on the in vivo and in vitro metastatic and invasive capabilities of MHCC97L cells. Subsequently, we directed our attention to investigating the impact of EGR2 on the levels of interleukin-8 (IL-8). Through comprehensive analyses including RNA sequencing, CUT-and-Tag, and ChIP techniques, we demonstrated that EGR2 can bind to the promoter region of the Pyruvate Dehydrogenase Kinase 4 (PDK4) gene. Finally, we introduced a virus overexpressing PDK4 and demonstrated that EGR2 could regulate the transcriptional level of PDK4 to affect the expression of IL-8, and ultimately alter the metastatic ability of hepatocellular carcinoma cells. Our study demonstrates that EGR2 may be a valuable target for future intervention in the disease process of hepatocellular carcinoma and refines the mechanism at the microscopic level of Tumor-associated macrophages.
Collapse
Affiliation(s)
- Song Wei
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Gaoxin Xu
- Department of General Surgery, Affiliated Kunshan Hospital of Jiangsu University,Kunshan,Suzhou,China
| | - Siqi Zhao
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, , Zhejiang, China
| | - Chenwei Zhang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yongheng Feng
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Weijun Yang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Renhe Lu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jin Zhou
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| | - Yong Ma
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
2
|
Wu S, Luo Y, Wei F, Li Y, Fan J, Chen Y, Zhang W, Li X, Xu Y, Chen Z, Xia C, Hu M, Li P, Gu Q. Lactic acid bacteria target NF-κB signaling to alleviate gastric inflammation. Food Funct 2025; 16:3101-3119. [PMID: 40152095 DOI: 10.1039/d4fo06308b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Helicobacter pylori (H. pylori) infection and the resulting gastric inflammation are major contributors to gastric cancer development. Probiotics, particularly Lactobacillus, are promising for their anti-inflammatory potential, yet their exact mechanisms in inhibiting H. pylori-induced inflammation are unclear. In our previous study, Lactiplantibacillus plantarum ZJ316 (L. plantarum ZJ316) demonstrated strong anti-inflammatory effects against H. pylori infection in vivo, but its precise mechanisms were not fully understood. Here, we aimed to investigate how L. plantarum ZJ316 inhibits the inflammatory response to H. pylori infection. Our results demonstrated that L. plantarum ZJ316 effectively reduced the expression of pro-inflammatory cytokines in H. pylori-infected AGS cells. Mechanistically, L. plantarum ZJ316 inhibited the NF-κB signaling pathway by preventing the degradation of IκBα, suppressing p65 phosphorylation, and blocking the nuclear translocation of phosphorylated p65. Treatment with the NF-κB inhibitor BAY 11-7082 further decreased tumor necrosis factor-α (TNF-α), interleukin-8 (IL-8), and interleukin-1β (IL-1β) levels, confirming the inhibitory effect of L. plantarum ZJ316 on the NF-κB pathway. In H. pylori-infected mice, oral administration of L. plantarum ZJ316 significantly alleviated inflammatory cell infiltration, reduced TNF-α and pepsinogen II (PGII) levels, and increased interleukin-10 (IL-10) levels in serum. A comparative metagenomic analysis of the gastric microbiota revealed a decrease in Prevotella and Desulfovibrio, alongside an increase in Ligilactobacillus and Akkermansia, supporting the protective effects of L. plantarum ZJ316 and correlating with their decreased inflammatory response. In summary, administration of L. plantarum ZJ316 demonstrated robust anti-inflammatory effects against H. pylori infection by suppressing NF-κB signaling and promoting favorable changes in the gastric microbiota composition. Therefore, L. plantarum ZJ316 holds promise as a novel functional food for protecting the body against H. pylori infection.
Collapse
Affiliation(s)
- Shiying Wu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Yuenuo Luo
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Fangtong Wei
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Yanan Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, Jiangsu 210023, China
| | - Jiayi Fan
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Yongqiang Chen
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Wenjie Zhang
- Hangzhou Helixinjian Industry Co., Ltd, No. 48 Zijinghua Road, Gudang Street, Xihu District, Hangzhou, Zhejiang 310050, China
| | - Xuelong Li
- Hangzhou Helixinjian Industry Co., Ltd, No. 48 Zijinghua Road, Gudang Street, Xihu District, Hangzhou, Zhejiang 310050, China
| | - Yang Xu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Ziqi Chen
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Chenlan Xia
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Mingyang Hu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Ping Li
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Qing Gu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| |
Collapse
|
3
|
Cui M, Yao G, Zhang Y, Wen M, Zhang S, Jin J, Lin Z, Ren X, An R, Piao Y. The molecular mechanisms of Caulophyllum robustum Maxim extract inhibition by regulating FAK/PI3K signaling pathway in gastric cancer HGC-27 cells. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118867. [PMID: 39369918 DOI: 10.1016/j.jep.2024.118867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/12/2024] [Accepted: 09/28/2024] [Indexed: 10/08/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Caulophyllumrobustum Maxim extract (CRME), as recorded in traditional Chinese medicine, has the function of dispelling Feng, regulating Qi and dredging collaterals, promoting blood circulation and regulating menstruation, gingering up and relieving pain, clearing heat simultaneously detoxifying, lowering blood pressure and hemostasis. CRME is often used as Chinese materia medica preparation for rheumatoid arthritis, traumatic injury, irregular menstruation, abdominal pain, and hypertension treatment. Since gastric cancer (GC) existed as a health problem of human over the years, we are committed to the development of potential components of Chinese herbal medicine curing cancer, and we found CRME is expected to be one of the effective anti-tumor traditional Chinese medicine preparations. AIMS OF THE STUDY To investigate the molecular mechanisms of CRME anticancer effects and the potential links between CRME and FAK. MATERIALS AND METHODS Caulophyllumrobustum Maxim was extracted to obtain CRME, high-performance liquid chromatography (HPLC) was used for qualitative analysis. Information about CRME was collected from traditional Chinese medicine records and local surveys unpublished internationally. Series of cellular function experiments were applied to detect cell proliferation, migration, apoptosis, autophagy, cell cycle, angiogenesis. The xenograft model is employed in vivo. RESULTS CRME can significantly inhibit HGC-27 cells on proliferation, migration and angiogenic capacity. Xenograft model indicated CRME inhibited cell proliferation in vivo. Annexin V-FITC/PI double staining assay and PI single staining assay depicted that CRME induces cell apoptosis, and arrests cell cycle at G0/G1 phase. AO (acridine orange) staining assay showed that CRME promoted autophagosome formation and inhibited autophagic flow. HPLC indicated Cauloside A and Cauloside C are components of CRME. Western blot indicated that FAK/PI3K signaling pathway is critical in the inhibition of CRME on HGC-27 cells. CONCLUSIONS The anti-tumor components of CRME, Cauloside A and Cauloside C, inhibited tumor progression in HGC-27 cells. This inhibition is achieved by decreasing the phosphorylation levels of FAK, thereby modulating PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Minghua Cui
- The key laboratory of pathobiology on the tumors with high incidence in Ethics area, State Ethnic Affairs Commission; Cancer Research Center of Yanbian University, Yanji, China
| | - Guangyuan Yao
- The key laboratory of pathobiology on the tumors with high incidence in Ethics area, State Ethnic Affairs Commission; Cancer Research Center of Yanbian University, Yanji, China
| | - Yingying Zhang
- The key laboratory of pathobiology on the tumors with high incidence in Ethics area, State Ethnic Affairs Commission; Cancer Research Center of Yanbian University, Yanji, China; Chifeng Municipal Hospital, Chifeng, China
| | - Meixin Wen
- The key laboratory of pathobiology on the tumors with high incidence in Ethics area, State Ethnic Affairs Commission; Cancer Research Center of Yanbian University, Yanji, China
| | - Shengjun Zhang
- The key laboratory of pathobiology on the tumors with high incidence in Ethics area, State Ethnic Affairs Commission; Cancer Research Center of Yanbian University, Yanji, China
| | - Jingchun Jin
- The key laboratory of pathobiology on the tumors with high incidence in Ethics area, State Ethnic Affairs Commission; Cancer Research Center of Yanbian University, Yanji, China; Yanbian University Hospital, Yanji, China
| | - Zhenhua Lin
- The key laboratory of pathobiology on the tumors with high incidence in Ethics area, State Ethnic Affairs Commission; Cancer Research Center of Yanbian University, Yanji, China
| | - Xiangshan Ren
- The key laboratory of pathobiology on the tumors with high incidence in Ethics area, State Ethnic Affairs Commission; Cancer Research Center of Yanbian University, Yanji, China
| | - Renbo An
- Key Laboratory of Changbai Mountain Natural Medicine Research, Yanji, China
| | - Yingshi Piao
- The key laboratory of pathobiology on the tumors with high incidence in Ethics area, State Ethnic Affairs Commission; Cancer Research Center of Yanbian University, Yanji, China.
| |
Collapse
|
4
|
Nasier-Hussain M, Samanje JN, Mokhtari K, Nabi-Afjadi M, Fathi Z, Hoseini A, Bahreini E. Serum levels of oxidative stress, IL-8, and pepsinogen I/II ratio in Helicobacter pylori and gastric cancer patients: potential diagnostic biomarkers. BMC Gastroenterol 2025; 25:2. [PMID: 39748276 PMCID: PMC11697901 DOI: 10.1186/s12876-024-03564-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND AND AIM Helicobacter pylori (H.pylori), a gram-negative bacterial pathogen associated with an increased risk of gastric cancer. This study investigates potential factors in the incidence of gastric cancer in patients with H.pylori, including oxidative stress, inflammatory biomarkers, serum pepsinogens (PG) of I and II, and PG-I/PG-II ratio. METHODS The study comprised individuals with Helicobacter pylori (H.pylori) infection, gastric cancer patients, and healthy individuals. Biochemical parameters such as FBS (fasting blood sugar), lipid profile, and liver and kidney functional factors were evaluated using colorimetric techniques. Oxidative markers such as total oxidant status (TOS) and malondialdehyde (MDA) were quantified through colorimetric methods. IL-8, PG-II, and PG-II levels were also determined using the ELISA technique. RESULTS Individuals with H. pylori infection exhibited elevated levels of IL-8 (940.5 ± 249.7 vs. 603.4 ± 89.1 pg/ml, P < 0.0001) and oxidative species (5.47 ± 0.7 vs. 1.64 ± 0.7 nM, P < 0.05) compared to gastric cancer patients, who, despite having lower levels of IL-8 and oxidative species, showed higher levels of MDA. H.pylori patients exhibited significantly higher levels of PG-I (7.28 ± 2.1 vs. 2.61 ± 1.4 ng/ml, P < 0.001), PG-II (3.21 ± 1 vs. 2.6 ± 0.6 ng/ml, P < 0.001), and the PG-I/PG-II ratio (2.27 ± 1.2 vs. 1 ± 0.4, P < 0.001) compared to gastric cancer patients. The findings were substantiated using various data analysis platforms such as Gene Expression Profiling Interactive Analysis (GEPIA), UALCAN (The University of ALabama at Birmingham CANcer data analysis), cBioPortal, and TIMER (Tumor IMmune Estimation Resource). These parameters could serve as potential diagnostic biomarkers for screening and therapeutic interventions based on the cut-off values derived from ROC (receiver operating characteristic) curves for IL-8, PGI, PGII, and PGI/PGII across the three groups. CONCLUSIONS IL-8, PGI, PGII, and PGI/PGII parameters could serve as potential diagnostic markers for the screening and treatment of gastric conditions.
Collapse
Affiliation(s)
- Marwa Nasier-Hussain
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614525, Tehran, Iran
| | - Jaleel Najah Samanje
- Collage of Health and Medical Technology, Middle Technical University, Baghdad, Iraq
| | - Khatere Mokhtari
- Department of Modern Biology, ACECR Institute of Higher Education (Isfahan Branch), Isfahan, Iran
- Department of Cellular and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zeinab Fathi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Asieh Hoseini
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Bahreini
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614525, Tehran, Iran.
| |
Collapse
|
5
|
Tang ZY, Wang XM, Xu CW, Sun QQ, Hua YX, Zhou QY, Hu HY, Liu SB, Guo YJ, Ao L, Che X, Zhang XC, Heger M, Zheng X, Liu AJ, Wang Q, Zhan ZJ, Cheng SQ, Pan WW. DCAF13 promotes ovarian cancer progression by activating FRAS1-mediated FAK signaling pathway. Cell Mol Life Sci 2024; 81:421. [PMID: 39367995 PMCID: PMC11455852 DOI: 10.1007/s00018-024-05446-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 08/09/2024] [Accepted: 09/10/2024] [Indexed: 10/07/2024]
Abstract
Cullin-RING ubiquitin ligase 4 (CRL4) is closely correlated with the incidence and progression of ovarian cancer. DDB1- and CUL4-associated factor 13 (DCAF13), a substrate-recognition protein in the CRL4 E3 ubiquitin ligase complex, is involved in the occurrence and development of ovarian cancer. However, its precise function and the underlying molecular mechanism in this disease remain unclear. In this study, we confirmed that DCAF13 is highly expressed in human ovarian cancer and its expression is negatively correlated with the overall survival rate of patients with ovarian cancer. We then used CRISPR/Cas9 to knockout DCAF13 and found that its deletion significantly inhibited the proliferation, colony formation, and migration of human ovarian cancer cells. In addition, DCAF13 deficiency inhibited tumor proliferation in nude mice. Mechanistically, CRL4-DCAF13 targeted Fraser extracellular matrix complex subunit 1 (FRAS1) for polyubiquitination and proteasomal degradation. FRAS1 influenced the proliferation and migration of ovarian cancer cell through induction of the focal adhesion kinase (FAK) signaling pathway. These findings collectively show that DCAF13 is an important oncogene that promotes tumorigenesis in ovarian cancer cells by mediating FRAS1/FAK signaling. Our findings provide a foundation for the development of targeted therapeutics for ovarian cancer.
Collapse
Affiliation(s)
- Ze-Yi Tang
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, P. R. China
- Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, P. R. China
- Department of Pharmacy, Xinhua Hospital, School of Medicine, Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Xiao-Min Wang
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, P. R. China
| | - Chun-Wei Xu
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, No. 1 Banshan East Street, Gongshu District, Hangzhou, 310022, China
| | - Qing-Qing Sun
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, P. R. China
- Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, P. R. China
| | - Yu-Xin Hua
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, P. R. China
- Zhejiang Chinese Medicine University and Jiaxing University Master Degree Cultivation Base, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, P. R. China
| | - Qi-Yin Zhou
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, P. R. China
- Zhejiang Chinese Medicine University and Jiaxing University Master Degree Cultivation Base, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, P. R. China
| | - Han-Yin Hu
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, P. R. China
- Zhejiang Chinese Medicine University and Jiaxing University Master Degree Cultivation Base, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, P. R. China
| | - Sheng-Bing Liu
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, P. R. China
| | - Yan-Jun Guo
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, P. R. China
| | - Lei Ao
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, P. R. China
| | - Xuan Che
- Department of Anesthesiology, Jiaxing Maternity and Child Health Care Hospital, Affiliated Women and Children Hospital, Jiaxing University, Jiaxing, 314001, P. R. China
| | - Xian-Chao Zhang
- Institute of Information Network and Artificial Intelligence, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, P. R. China
| | - Michal Heger
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, P. R. China
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, The Netherlands
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Dr. Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands
| | - Xin Zheng
- Department of Gynecology and Obstetrics, Affiliated Hospital of Jiaxing University, Jiaxing, 314000, P. R. China
| | - Ai-Jun Liu
- Department of Pathology, The 7th Medical Center, General Hospital of PLA, Beijing, 100700, P. R. China
| | - Qian Wang
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, P. R. China
| | - Zha-Jun Zhan
- Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, P. R. China.
| | - Shu-Qun Cheng
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, P. R. China.
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, 200438, P. R. China.
| | - Wei-Wei Pan
- Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, P. R. China.
- G60 STI Valley Industry & Innovation Institute, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001, P. R. China.
| |
Collapse
|
6
|
Li X, Xie G, Chen J, Wang Y, Zhai J, Shen L. Tumour cell-derived serglycin promotes IL-8 secretion of CAFs in gastric cancer. Br J Cancer 2024; 131:271-282. [PMID: 38862740 PMCID: PMC11263384 DOI: 10.1038/s41416-024-02735-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/19/2024] [Accepted: 05/21/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs)-derived IL-8 plays important roles in chemoresistance, immunosuppression, and lymph node metastasis of gastric cancer. However, the mechanisms underlying IL-8 production in CAFs remains unclear. METHODS DNA pulldown assay was performed to identify the transcription factors responsible for IL-8 expression in CAFs, which was further verified using CHIP-qPCR and DNA agarose gel electrophoresis assays. The cellular localisation of IL-8 was analysed using multiplex immunofluorescence (MxIF). RESULTS MxIF demonstrated that IL-8 was mainly produced by CAFs in gastric cancer. Lysine[K]-specific demethylase 5B (KDM5B) was identified as an IL-8 transcription factor in CAFs, and the binding of KDM5B to phosphorylated RB1 limited the transcriptional regulation of IL-8 in gastric cancer cells. Serglycin (SRGN) secreted by tumour cells activated the CD44/c-Myc pathway to upregulate KDM5B expression, thereby promoting IL-8 production in CAFs. Furthermore, tumour-associated neutrophils (TANs)-derived regenerating family member 4 (REG4) upregulates SRGN expression by activating cAMP-responsive element binding protein 1 (CREB1) in gastric cancer cells. Thus, the SRGN-IL-8-TANs-SRGN loop, which facilitates tumour progression, has been explored in gastric cancer. CONCLUSIONS This study revealed the mechanisms of the preferential production of IL-8 by CAFs in gastric cancer, and paves the way for potential new therapeutic strategies for gastric cancer.
Collapse
Affiliation(s)
- Xiang Li
- Department of Surgical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Guiping Xie
- Department of Surgical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Jia Chen
- Department of General Surgery, the First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China
- Department of General Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Yaohui Wang
- Department of Pathology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Jing Zhai
- Department of Surgical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Lizong Shen
- Department of Surgical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
- Department of General Surgery, the First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
7
|
Chen R, Liu L, Chen H, Xing C, Zhang T, Pang Y, Yang X. Evaluation of the clinical application value of cytokine expression profiles in the differential diagnosis of prostate cancer. Cancer Immunol Immunother 2024; 73:139. [PMID: 38833027 PMCID: PMC11150366 DOI: 10.1007/s00262-024-03723-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/02/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND The significance of tumor-secreted cytokines in tumor development has gained substantial attention. Nevertheless, the precise role of tumor-related inflammatory cytokines in prostate cancer (PCa) remains ambiguous. OBJECTIVES To gain deeper insights into the inflammatory response in the process of PCa. METHODS A total of 233 cases were collected, including 80 cases of prostate hyperplasia as disease control, 65 cases of postoperative prostate cancer and 36 cases of prostate cancer as PCa group. Additionally, 52 patients undergoing physical examinations during the same period were collected as the healthy control. The levels of 12 inflammatory cytokines in peripheral blood samples were analyzed using flow cytometric bead array technology. The levels of total prostate-specific antigen (TPSA) and free prostate-specific antigen (FPSA) in peripheral blood samples were analyzed using electrochemiluminescence technology. RESULTS Our findings revealed significant increases in serum IL-8 levels in PCa group compared to the healthy control group. Additionally, IL-6, IL-10, IFN-γ and IL-12p70 levels were markedly elevated in the PCa group compared to the disease control group (all p < 0.05). Conversely, the level of IL-4, TNF-α, IL-1β, IL-17A and IFN-α were lower in the PCa group compared to those in control group. Following surgery, the concentration of IL-6 decreased; whereas, the concentrations of IL-4, TNF-α, IL-17A, IL-1β, IL-12p70, and IFN-α increased, demonstrating significant differences (p < 0.05). The differential upregulation of IL-6 or downregulation of IL-17A in peripheral blood exhibited diagnostic efficacy in PCa patients. Moreover, we observed a significant increase in IL-17A levels, accompanied by decreased of IL-2, IL-4, IL-10, TNF-a, IFN-γ, IL-1β, and IL-12P70 in patients with distant metastasis. CONCLUSION The peripheral blood cytokines are closely associated with the occurrence and development of prostate cancer, especially the serum levels of IL-6 and IL-17A may be useful as potential predictors of PCa diagnosis.
Collapse
Affiliation(s)
- Rongfa Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Linna Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Hui Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Chao Xing
- Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Tingting Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yilin Pang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xunjun Yang
- Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
8
|
Jiang Y, Fu L, Liu B, Li F. YAP induces FAK phosphorylation to inhibit gastric cancer cell proliferation via upregulation of HMGB1. Int J Biol Macromol 2024; 262:130037. [PMID: 38331059 DOI: 10.1016/j.ijbiomac.2024.130037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/29/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
Yes associated protein (YAP) is the main effector protein in the Hippo pathway, regulating cell growth by binding to transcription factors in the nucleus. However, the mechanisms by which YAP regulates the development and progression of gastric cancer (GC) remain largely unknown. In this study, bioinformatics analysis determined that YAP was significantly upregulated in GC and associated with poor prognosis. In addition, YAP deletion inhibits proliferation and migration of GC cells in vitro, while overexpression of YAP has the opposite effect. Mechanistically, overexpression of YAP induced FAK phosphorylation in gastric cancer cells, whereas knockdown of YAP had the opposite effect. Importantly, translocation expressed mutant plasmid YAP-S94A (YAP1 mutant without TEAD binding site) did not significantly change the level of FAK phosphorylation. Furthermore, Verteporfin (a small molecule inhibitor of YAP) interrupted the YAP-TEAD interaction and inhibited FAK phosphorylation, confirming that YAP can induce FAK phosphorylation in a TEAD-dependent manner. In addition, the silencing of FAK or the use of FAK inhibitors inhibited the aggregation of YAP proteins in the nucleus, forming a FAK-YAP positive feedback loop. Finally, we identify the FAK upstream gene, HMGB1, as a direct transcriptional target of YAP-TEAD. Silencing HMGB1 reversed YAP-induced FAK activation as well as cell proliferation and migration. Collectively, our results reveal a new signalling axis, YAP/HMGB1/FAK, in the regulation of cell proliferation and migration, and provide new insights into the crosstalk between Hippo signalling and cell proliferation.
Collapse
Affiliation(s)
- Yunhe Jiang
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese, Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Lifu Fu
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese, Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Bin Liu
- Cardiovascular Disease Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Fan Li
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese, Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China; The Key Laboratory for Bionics Engineering, Ministry of Education, Jilin University, Changchun, China; Engineering Research Center for Medical Biomaterials of Jilin Province, Jilin University, Changchun, China; Key Laboratory for Health Biomedical Materials of Jilin Province, Jilin University, Changchun, China; State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang, China.
| |
Collapse
|
9
|
He Y, Gang B, Zhang M, Bai Y, Wan Z, Pan J, Liu J, Liu G, Gu W. ACE2 improves endothelial cell function and reduces acute lung injury by downregulating FAK expression. Int Immunopharmacol 2024; 128:111535. [PMID: 38246001 DOI: 10.1016/j.intimp.2024.111535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/01/2024] [Accepted: 01/10/2024] [Indexed: 01/23/2024]
Abstract
Endothelial cell (EC) barrier dysfunction and increased adhesion of immune inflammatory cells to ECs crucially contribute to acute lung injury (ALI). Angiotensin-converting enzyme 2 (ACE2) is an essential regulator of the renin-angiotensin system (RAS) and exerts characteristic vasodilatory and anti-inflammatory effects. SARS-COV-2 infects the lungs by binding to ACE2, which can lead to dysregulation of ACE2 expression, further leading to ALI with predominantly vascular inflammation and eventually to more severe acute respiratory distress syndrome (ARDS). Therefore, restoration of ACE2 expression represents a valuable therapeutic approach for SARS-COV-2-related ALI/ARDS. In this study, we used polyinosinic-polycytidylic acid (Poly(I:C)), a double-stranded RNA analog, to construct a mouse ALI model that mimics virus infection. After Poly(I:C) exposure, ACE2 was downregulated in mouse lung tissues and in cultured ECs. Treatment with DIZE, an ACE2-activating compound, upregulated ACE2 expression and relieved ALI in mice. DIZE also improved barrier function and reduced the number of THP-1 monocytes adhering to cultured ECs. Focal adhesion kinase (FAK) and phosphorylated FAK (p-FAK) levels were increased in lung tissues of ALI mice as well as in Poly(I:C)-treated ECs in vitro. Both DIZE and the FAK inhibitor PF562271 decreased FAK/p-FAK expression in both ALI models, attenuating ALI severity in vivo and increasing barrier function and reducing monocyte adhesion in cultured ECs. Furthermore, in vivo experiments using ANG 1-7 and the MAS inhibitor A779 corroborated that DIZE-mediated ACE2 activation stimulated the activity of the ANG 1-7/MAS axis, which inhibited FAK/p-FAK expression in the mouse lung. These findings provide further evidence that activation of ACE2 in ECs may be a valuable therapeutic strategy for ALI.
Collapse
Affiliation(s)
- Yixuan He
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, and Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, 2600 Donghai Avenue, Bengbu, Anhui Province 233030, PR China
| | - Baocai Gang
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, and Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, 2600 Donghai Avenue, Bengbu, Anhui Province 233030, PR China
| | - Mengjie Zhang
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, and Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, 2600 Donghai Avenue, Bengbu, Anhui Province 233030, PR China
| | - Yuting Bai
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, and Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, 2600 Donghai Avenue, Bengbu, Anhui Province 233030, PR China
| | - Ziyu Wan
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, and Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, 2600 Donghai Avenue, Bengbu, Anhui Province 233030, PR China
| | - Jiesong Pan
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, and Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, 2600 Donghai Avenue, Bengbu, Anhui Province 233030, PR China
| | - Jie Liu
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, Sichuan Province, PR China
| | - Guoquan Liu
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, and Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, 2600 Donghai Avenue, Bengbu, Anhui Province 233030, PR China.
| | - Wei Gu
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, and Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, 2600 Donghai Avenue, Bengbu, Anhui Province 233030, PR China.
| |
Collapse
|
10
|
Tatsuno R, Ichikawa J, Komohara Y, Pan C, Kawasaki T, Enomoto A, Aoki K, Hayakawa K, Iwata S, Jubashi T, Haro H. Pivotal role of IL-8 derived from the interaction between osteosarcoma and tumor-associated macrophages in osteosarcoma growth and metastasis via the FAK pathway. Cell Death Dis 2024; 15:108. [PMID: 38302407 PMCID: PMC10834992 DOI: 10.1038/s41419-024-06487-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/03/2024]
Abstract
The prognosis of osteosarcoma (OS) has remained stagnant over the past two decades, requiring the exploration of new therapeutic targets. Cytokines, arising from tumor-associated macrophages (TAMs), a major component of the tumor microenvironment (TME), have garnered attention owing to their impact on tumor growth, invasion, metastasis, and resistance to chemotherapy. Nonetheless, the precise functional role of TAMs in OS progression requires further investigation. In this study, we investigated the interaction between OS and TAMs, as well as the contribution of TAM-produced cytokines to OS advancement. TAMs were observed to be more prevalent in lung metastases compared with that in primary tumors, suggesting their potential support for OS progression. To simulate the TME, OS and TAMs were co-cultured, and the cytokines resulting from this co-culture could stimulate OS proliferation, migration, and invasion. A detailed investigation of cytokines in the co-culture conditioned medium (CM) revealed a substantial increase in IL-8, establishing it as a pivotal cytokine in the process of enhancing OS proliferation, migration, and invasion through the focal adhesion kinase (FAK) pathway. In an in vivo model, co-culture CM promoted OS proliferation and lung metastasis, effects that were mitigated by anti-IL-8 antibodies. Collectively, IL-8, generated within the TME formed by OS and TAMs, accelerates OS proliferation and metastasis via the FAK pathway, thereby positioning IL-8 as a potential novel therapeutic target in OS.
Collapse
Affiliation(s)
- Rikito Tatsuno
- Department of Orthopaedic Surgery, University of Yamanashi, Yamanashi, Japan
| | - Jiro Ichikawa
- Department of Orthopaedic Surgery, University of Yamanashi, Yamanashi, Japan.
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Cheng Pan
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tomonori Kawasaki
- Department of Pathology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Kaoru Aoki
- Physical Therapy Division, School of Health Sciences, Shinshu University, Nagano, Japan
| | - Keiko Hayakawa
- Department of Orthopaedic Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shintaro Iwata
- Department of Musculoskeletal Oncology and Rehabilitation, National Cancer Center Hospital, Tokyo, Japan
| | - Takahiro Jubashi
- Department of Orthopaedic Surgery, University of Yamanashi, Yamanashi, Japan
| | - Hirotaka Haro
- Department of Orthopaedic Surgery, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
11
|
Sun G, Ye H, Yang Q, Zhu J, Qiu C, Shi J, Dai L, Wang K, Zhang J, Wang P. Using Proteome Microarray and Gene Expression Omnibus Database to Screen Tumour-Associated Antigens to Construct the Optimal Diagnostic Model of Oesophageal Squamous Cell Carcinoma. Clin Oncol (R Coll Radiol) 2023; 35:e582-e592. [PMID: 37433700 DOI: 10.1016/j.clon.2023.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 05/09/2023] [Accepted: 06/30/2023] [Indexed: 07/13/2023]
Abstract
AIMS Autoantibodies against tumour-associated antigens (TAAs) are promising biomarkers for early immunodiagnosis of cancers. This study was designed to screen and verify autoantibodies against TAAs in sera as diagnostic biomarkers for oesophageal squamous cell carcinoma (ESCC). MATERIALS AND METHODS The customised proteome microarray based on cancer driver genes and the Gene Expression Omnibus database were used to identify potential TAAs. The expression levels of the corresponding autoantibodies in serum samples obtained from 243 ESCC patients and 243 healthy controls were investigated by enzyme-linked immunosorbent assay (ELISA). In total, 486 serum samples were randomly divided into the training set and the validation set in the ratio of 2:1. Logistic regression analysis, recursive partition analysis and support vector machine were performed to establish different diagnostic models. RESULTS Five and nine candidate TAAs were screened out by proteome microarray and bioinformatics analysis, respectively. Among these 14 anti-TAAs autoantibodies, the expression level of nine (p53, PTEN, GNA11, SRSF2, CXCL8, MMP1, MSH6, LAMC2 and SLC2A1) anti-TAAs autoantibodies in the cancer patient group was higher than that in the healthy control group based on the results from ELISA. In the three constructed models, a logistic regression model including four anti-TAA autoantibodies (p53, SLC2A1, GNA11 and MMP1) was considered to be the optimal diagnosis model. The sensitivity and specificity of the model in the training set and the validation set were 70.4%, 72.8% and 67.9%, 67.9%, respectively. The area under the receiver operating characteristic curve for detecting early patients in the training set and the validation set were 0.84 and 0.85, respectively. CONCLUSIONS This approach to screen novel TAAs is feasible, and the model including four autoantibodies could pave the way for the diagnosis of ESCC.
Collapse
Affiliation(s)
- G Sun
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan Province, China; Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, China
| | - H Ye
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan Province, China; Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Q Yang
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan Province, China; Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, China
| | - J Zhu
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan Province, China; Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, China
| | - C Qiu
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, China
| | - J Shi
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, China; Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - L Dai
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, China; Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - K Wang
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, China; Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - J Zhang
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, China; Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - P Wang
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan Province, China; Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, China.
| |
Collapse
|