1
|
Wang C, Ji J, Jin Y, Sun Y, Cai Q, Jiang J, Guo L, Zhou C, Zhang J. Tumor-mesothelium HOXA11-PDGF BB/TGF β1-miR-181a-5p-Egr1 feedforward amplifier circuity propels mesothelial fibrosis and peritoneal metastasis of gastric cancer. Oncogene 2024; 43:171-188. [PMID: 37989866 PMCID: PMC10786717 DOI: 10.1038/s41388-023-02891-4] [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: 05/19/2023] [Revised: 11/02/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023]
Abstract
A proportion of gastric cancer (GC) patients suffer from peritoneal metastasis (PM) in the late stage of tumor and these patients have a poor prognosis. To provide more care for GC patient with PM, a deeper exploration of the molecular characteristics of GC-PM is needed. Here we performed the in vitro and in vivo study to illustrate the effect of HOXA11 over-expressed GC cells on peritoneal mesothelial cells (HMrSV5), transcriptomics analyses of HMrSV5 cells co-cultured with HOXA11 over-expressed GC cells, counterparts or alone, cytokine array analyses of serum-free culture medium of HOXA11 over-expressed GC cells, we validated our findings through genetic manipulation of HMrSV5 cells and neutralizing antibodies targeting cytokines secreted by HOXA11 over-expressed GC cells in vitro, as well as utilized human peritoneal metastatic lesions to validate expression of potential targets. We identified that HOXA11 over-expressed GC cells strongly propelled mesothelial fibrosis in vivo and in vitro, and HOXA11 regulated paracrine and autocrine of PDGF BB and TGF β1 in GC cells to propel mesothelial fibrosis. Meanwhile, HOXA11 over-expressed GC cells drove PDGF BB and TGF β1 secretion to activate developmental-process related genes in HMrSV5 cells, including Egr1, which processes dependent on miR-181a-5p. Then, Egr1 could mediate peritoneal mesothelial fibrosis. Correspondingly, Egr1 over-expressed HMrSV5 cells supported migration and peritoneal dissemination of GC cells. Together our results suggest that a feedforward amplifier circuity governing GC cells and mesothelial cells in peritoneum contribute to peritoneal metastasis of GC cells.
Collapse
Affiliation(s)
- Chao Wang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Jun Ji
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Yangbing Jin
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Ying Sun
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Qu Cai
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Jinling Jiang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Liting Guo
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Chenfei Zhou
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China.
| | - Jun Zhang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China.
| |
Collapse
|
2
|
Lv ZD, Wang HB, Dong Q, Kong B, Li JG, Yang ZC, Qu HL, Cao WH, Xu HM. Retraction Note to: Mesothelial cells differentiate into fibroblast-like cells under the scirrhous gastric cancer microenvironment and promote peritoneal carcinomatosis in vitro and in vivo. Mol Cell Biochem 2024; 479:197-198. [PMID: 38038799 DOI: 10.1007/s11010-023-04911-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Affiliation(s)
- Zhi-Dong Lv
- Department of Breast Surgery, The Affiliated Hospital of Medical College, Qingdao University, Qingdao, 266003, Shandong, People's Republic of China
- Department of Surgical Oncology, The First Affiliated Hospital, China Medical University, Shenyang, 110001, Liaoning, People's Republic of China
| | - Hai-Bo Wang
- Department of Breast Surgery, The Affiliated Hospital of Medical College, Qingdao University, Qingdao, 266003, Shandong, People's Republic of China.
| | - Qian Dong
- Department of Pediatric Surgery, The Affiliated Hospital of Medical College, Qingdao University, Qingdao, 266003, Shandong, People's Republic of China
| | - Bin Kong
- Department of Breast Surgery, The Affiliated Hospital of Medical College, Qingdao University, Qingdao, 266003, Shandong, People's Republic of China
| | - Jian-Guo Li
- Department of Breast Surgery, The Affiliated Hospital of Medical College, Qingdao University, Qingdao, 266003, Shandong, People's Republic of China
| | - Zhao-Chuan Yang
- Department of Child Health Care, The Affiliated Hospital of Medical College, Qingdao University, Qingdao, 266003, Shandong, People's Republic of China
| | - Hui-Li Qu
- Department of Breast Surgery, The Affiliated Hospital of Medical College, Qingdao University, Qingdao, 266003, Shandong, People's Republic of China
| | - Wei-Hong Cao
- Department of Breast Surgery, The Affiliated Hospital of Medical College, Qingdao University, Qingdao, 266003, Shandong, People's Republic of China
| | - Hui-Mian Xu
- Department of Surgical Oncology, The First Affiliated Hospital, China Medical University, Shenyang, 110001, Liaoning, People's Republic of China.
| |
Collapse
|
3
|
Kimura Y, Ohzawa H, Miyato H, Kaneko Y, Kuchimaru T, Takahashi R, Yamaguchi H, Kurashina K, Saito S, Hosoya Y, Lefor AK, Sata N, Kitayama J. Intraperitoneal transfer of microRNA-29b-containing small extracellular vesicles can suppress peritoneal metastases of gastric cancer. Cancer Sci 2023; 114:2939-2950. [PMID: 36939028 PMCID: PMC10323101 DOI: 10.1111/cas.15793] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 03/21/2023] Open
Abstract
Small extracellular vesicles (sEV) contain various microRNAs (miRNAs) and play crucial roles in the tumor metastatic process. Although miR-29b levels in peritoneal exosomes were markedly reduced in patients with peritoneal metastases (PM), their role has not been fully clarified. In this study, we asked whether the replacement of miR-29b can affect the development of PM in a murine model. UE6E7T-12, human bone marrow-derived mesenchymal stem cells (BMSCs), were transfected with miR-29b-integrating recombinant lentiviral vector and sEV were isolated from culture supernatants using ultracentrifugation. The sEV contained markedly increased amounts of miR-29b compared with negative controls. Treatment with transforming growth factor-β1 decreased the expression of E-cadherin and calretinin with increased expression of vimentin and fibronectin on human omental tissue-derived mesothelial cells (HPMCs). However, the effects were totally abrogated by adding miR-29b-rich sEV. The sEV inhibited proliferation and migration of HPMCs by 15% (p < 0.005, n = 6) and 70% (p < 0.005, n = 6), respectively, and inhibited adhesion of NUGC-4 and MKN45 to HPMCs by 90% (p < 0.0001, n = 5) and 77% (p < 0.0001, n = 5), respectively. MicroRNA-29b-rich murine sEV were similarly obtained using mouse BMSCs and examined for in vivo effects with a syngeneic murine model using YTN16P, a highly metastatic clone of gastric cancer cell. Intraperitoneal (IP) transfer of the sEV every 3 days markedly reduced the number of PM from YTN16P in the mesentery (p < 0.05, n = 6) and the omentum (p < 0.05, n = 6). Bone marrow mesenchymal stem cell-derived sEV are a useful carrier for IP administration of miR-29b, which can suppress the development of PM of gastric cancer.
Collapse
Affiliation(s)
- Yuki Kimura
- Department of SurgeryJichi Medical University HospitalShimotsukeJapan
| | - Hideyuki Ohzawa
- Department of Clinical OncologyJichi Medical University HospitalShimotsukeJapan
| | - Hideyo Miyato
- Department of SurgeryJichi Medical University HospitalShimotsukeJapan
| | - Yuki Kaneko
- Department of SurgeryJichi Medical University HospitalShimotsukeJapan
| | | | - Rei Takahashi
- Department of SurgeryJichi Medical University HospitalShimotsukeJapan
| | - Hironori Yamaguchi
- Department of Clinical OncologyJichi Medical University HospitalShimotsukeJapan
| | - Kentaro Kurashina
- Department of SurgeryJichi Medical University HospitalShimotsukeJapan
| | - Shin Saito
- Department of SurgeryJichi Medical University HospitalShimotsukeJapan
| | - Yoshinori Hosoya
- Department of SurgeryJichi Medical University HospitalShimotsukeJapan
| | | | - Naohiro Sata
- Department of SurgeryJichi Medical University HospitalShimotsukeJapan
| | - Joji Kitayama
- Department of SurgeryJichi Medical University HospitalShimotsukeJapan
- Center for Clinical ResearchJichi Medical University HospitalShimotsukeJapan
| |
Collapse
|
4
|
Ye X, Li J, Liu Z, Sun X, Wei D, Song L, Wu C. Peptide mediated therapy in fibrosis: Mechanisms, advances and prospects. Biomed Pharmacother 2023; 157:113978. [PMID: 36423541 DOI: 10.1016/j.biopha.2022.113978] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/30/2022] [Accepted: 11/03/2022] [Indexed: 11/22/2022] Open
Abstract
Fibrosis, a disease characterized by an excess accumulation of extracellular matrix components, could lead to organ failure and death, and is to blame for up to 45 % of all fatalities in developed nations. These disorders all share the common trait of an unchecked and increasing accumulation of fibrotic tissue in the affected organs, which leads to their malfunction and eventual failure, even if their underlying causes are highly diverse and, in some cases, remain unclear. Numerous studies have identified activated myofibroblasts as the common cellular elements ultimately responsible for the replacement of normal tissues with nonfunctional fibrotic tissue. The transforming growth factor-β pathway, for instance, plays a significant role in practically all kinds of fibrosis. However, there is no specific drug for the treatment of fibrosis, several medications with anti-hepatic fibrosis properties are still in the research and development stages. Peptide, which refers to a substance consisting of 2-50 amino acids, is characterized by structural diversity, low toxicity, biological activities, easy absorption, specific targeting, few side effects, and has been proven to be effective in anti-fibrosis. Here, we summarized various anti-fibrosis peptides in fibrosis including the liver, lungs, kidneys, and other organs. This review will provide a new insight into peptide mediated anti-fibrosis and is helpful to creation of antifibrotic medications.
Collapse
Affiliation(s)
- Xun Ye
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Jinhu Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Zibo Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Xue Sun
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Daneng Wei
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Linjiang Song
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, PR China.
| | - Chunjie Wu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China.
| |
Collapse
|
5
|
Wen Z, Yang C, Zou D, Liu J, Wang S, Liu X, Zhang Y, Zhang Y. Pan-cancer analysis of PSAP identifies its expression and clinical relevance in gastric cancer. Pathol Res Pract 2022; 238:154027. [PMID: 36084426 DOI: 10.1016/j.prp.2022.154027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 11/23/2022]
Abstract
Prosaposin (PSAP) plays a critical role in sphingolipid and cancer metabolism. Reports have shown that PSAP was involved in proliferation, tumorigenesis, and metastasis. However, the expression pattern of PSAP and its prognostic roles in gastric cancer remain elusive. PSAP expression pattern and its prognostic roles in gastric cancer (GC) were explored using data from the TCGA and Kaplan-Meier Plotter. Immunohistochemical staining of GC tissues was performed to validate the prognostic role of PSAP. TISIDB was used to analyze its correlation with immunomodulators. PSAP-associated genes, PDCD1, TGFB1, and CSF1R were used to build a risk model to evaluate immunotherapy outcomes of patients with stomach adenocarcinoma (STAD). Results showed that PSAP was highly expressed in GC. High PSAP expression in GC patients also significantly indicated a poor prognosis. The results of immunohistochemical staining showed that PSAP was an independent prognostic factor in GC patients. Based on three PSAP-associated genes, a risk model that could predict the prognosis and immunotherapy outcome of STAD was bulit. PSAP was an independent prognostic factor in GC. Our results have identified three prognosis-related genes which were useful to evaluate immunotherapy outcomes of STAD patients.
Collapse
Affiliation(s)
- Zhenpeng Wen
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| | - Chunjiao Yang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| | - Dan Zou
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Insititute, Shenyang, Liaoning Province 110042, PR China.
| | - Jiaqing Liu
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| | - Song Wang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| | - Xuqin Liu
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| | - Yi Zhang
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| | - Ye Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| |
Collapse
|
6
|
Jiang Y, Chen F, Ren X, Yang Y, Luo J, Yuan J, Yuan J, Tong Q. RNA-Binding Protein COL14A1, TNS1, NUSAP1 and YWHAE Are Valid Biomarkers to Predict Peritoneal Metastasis in Gastric Cancer. Front Oncol 2022; 12:830688. [PMID: 35515139 PMCID: PMC9062106 DOI: 10.3389/fonc.2022.830688] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/25/2022] [Indexed: 01/15/2023] Open
Abstract
Gastric cancer (GC) is the third leading cause of tumor related mortality worldwide. Peritoneal metastasis (PM) occurs in more than half of advanced GC patients, leading to poor prognosis. Therefore, the GSE62254 cohort was used to construct a signature consisting of four RNA-binding proteins (RBP) to predict the possibility of PM in GC patients. Then, ROC curves were plotted followed by calculation of AUCs, showing that the signature had a similar predictive accuracy compared with the TNM staging system. Importantly, the capability of prediction was enhanced by combining the classifier and TNM staging. In order to validate the expression of the four RBPs in GC tissues with and without PM, immunohistochemistry was further performed on samples from 108 patients. The differential expression of COL14A1, TNS1, NUSAP1 and YWHAE was in accordance with the emergence of PM. Afterwards, we produced Kaplan–Meier curves according to the signature and differential expression of the RBPs in patients. Finally, CCK-8 assays were performed to verify the effect on cell proliferation, finding that COL14A1 and TNS1 promoted cell proliferation, while NUSAP1 and YWHAE led to suppressed cell proliferation. In conclusion, the four-RBP-based signature, combined with TNM staging, has the potential to predict risk of PM in GC.
Collapse
Affiliation(s)
- Yue Jiang
- Department of Gastrointestinal Surgery I Section, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fangfang Chen
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xunshan Ren
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yu Yang
- Department of Gastrointestinal Surgery I Section, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiajun Luo
- Department of Gastrointestinal Surgery I Section, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingwen Yuan
- Department of Gastrointestinal Surgery I Section, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qiang Tong
- Department of Gastrointestinal Surgery I Section, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
7
|
Lin XM, Wang ZJ, Lin YX, Chen H. Decreased exosome-delivered miR-486-5p is responsible for the peritoneal metastasis of gastric cancer cells by promoting EMT progress. World J Surg Oncol 2021; 19:312. [PMID: 34686196 PMCID: PMC8539850 DOI: 10.1186/s12957-021-02381-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/26/2021] [Indexed: 11/10/2022] Open
Abstract
Background The present study aims to investigate the preliminary mechanism underlying the peritoneal metastasis of gastric cancer cells. Methods Exosomes from GC9811 cells (Con-Exo) and from GC9811-P cells (PM-Exo) were extracted by ultracentrifugation, which were identified with transmission electron microscopy (TEM) and nanoparticle trafficking analysis, as well as the expression of CD9, CD63, and CD81 detected by Western blot assay. α-SMA expression was determined by immunofluorescence assay and Western blot assay. The levels of Snail1, E-cadherin, and Actin-related protein 3 (ACTR3) were evaluated by Western blot assay. MiRNA array was performed on exosomes to screen the differentially expressed miRNAs. The expressions of miRNAs, SMAD2, CDK4, and ACTR3 were determined by QRT-PCR. The delivery of miR-486-5p was confirmed by laser confocal detection. Results Firstly, TEM, nanoparticle trafficking analysis, and Western blot assays were used to confirm the successful extraction of Con-Exo and PM-Exo. The incubation of Con-Exo and PM-Exo could decrease E-cadherin expression and increase of α-SMA respectively in HMrSV5 cells, with the increased proportion of fusiform cells. More significant changes were observed in PM-Exo-treated HMrSV5 cells. Secondary, compared to Con-Exo, miR-486-5p and miR-132-3p were found downregulated, and miR-132-5p was found upregulated in PM-Exo. The transfection of miR-486-5p and miR-132-3p was observed to suppress EMT, and the transfection of miR-132-3p was observed to induce EMT. Laser confocal detection confirmed the delivery of miR-486-5p from gastric cancer cells to HMrSV5 cells through exosomes. Lastly, the expression of Mothers against decapentaplegic homolog 2 (SMAD2), cyclin-dependent kinase 4 (CDK4), and ACTR3 was found to be downregulated via miR-486-5p. Conclusion Decreased delivery of miR-486-5p via exosomes might be responsible for the peritoneal metastasis of gastric cancer cells by promoting epithelial-mesenchymal transition progress.
Collapse
Affiliation(s)
- Xian-Ming Lin
- Department of General Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, People's Republic of China.
| | - Zhi-Jiang Wang
- Department of General Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - Yu-Xiao Lin
- Next Generation Sequencing, DIAN Diagnostics Group Co., Ltd., Hangzhou, 310030, Zhejiang, People's Republic of China.,Next Generation Sequencing, Hangzhou Dian Huayin Biotechnology Co., Ltd., Hangzhou, 310030, Zhejiang, People's Republic of China
| | - Hao Chen
- Safety Evaluation Center, Zhejiang Academy of Medical Sciences, Hangzhou, 310007, Zhejiang, People's Republic of China
| |
Collapse
|
8
|
Veen LM, Skrabanja TLP, Derks S, de Gruijl TD, Bijlsma MF, van Laarhoven HWM. The role of transforming growth factor β in upper gastrointestinal cancers: A systematic review. Cancer Treat Rev 2021; 100:102285. [PMID: 34536730 DOI: 10.1016/j.ctrv.2021.102285] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/27/2021] [Accepted: 08/29/2021] [Indexed: 01/02/2023]
Abstract
Esophageal and gastric malignancies are associated with poor prognosis, in part due to development of recurrences or metastases after curative treatment. The transforming growth factor β (TGF-β) pathway might play a role in the development of treatment resistance. In this systematic review, we provide an overview of preclinical studies investigating the role of TGF-β in esophageal and gastric malignancies. We systematically searched MEDLINE/PubMed and EMBASE for eligible preclinical studies describing the effect of TGF-β or TGF-β inhibition on hallmarks of cancer, such as proliferation, migration, invasion, angiogenesis and immune evasion. In total, 2107 records were screened and 45 articles were included, using mouse models and 45 different cell lines. TGF-β failed to induce apoptosis in twelve of sixteen tested cell lines. TGF-β could either decrease (five cell lines) or increase proliferation (seven cell lines) in gastric cancer cells, but had no effect in esophageal cancer cells. In all esophageal and all but two gastric cancer cell lines, TGF-β increased migratory, adhesive and invasive capacities. In vivo studies showed increased metastasis in response to TGF-β treatment. Additionally, TGF-β was shown to induce vascular endothelial growth factor production and differentiation of cancer-associated fibroblasts and regulatory T-cells. In conclusion, we found that TGF-β enhances hallmarks of cancer in most gastric and esophageal cancer cell lines, but not in all. Therefore, targeting the TGF-β pathway could be an attractive strategy in patients with gastric or esophageal cancer, but additional clinical trials are needed to define patient groups who would benefit most.
Collapse
Affiliation(s)
- Linde M Veen
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, De Boelelaan 1117-1118, 1081 HV Amsterdam, The Netherlands.
| | - Tim L P Skrabanja
- Laboratory of Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Sarah Derks
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, De Boelelaan 1117-1118, 1081 HV Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, De Boelelaan 1117-1118, 1081 HV Amsterdam, The Netherlands
| | - Maarten F Bijlsma
- Laboratory of Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Hanneke W M van Laarhoven
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, De Boelelaan 1117-1118, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
9
|
Shurygina IA, Shurygin МG, Rodionova LV, Ayushinova NI. Growth factors in the regulation of reparative response in the presence of peritoneal damage. Pleura Peritoneum 2020; 5:20200114. [PMID: 33575460 PMCID: PMC7823155 DOI: 10.1515/pp-2020-0114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 10/10/2020] [Indexed: 11/15/2022] Open
Abstract
Objectives To study the expression of growth factors in the regulation of tissue repair after peritoneal damage tissue response to peritoneal damage. Methods Experimental study in 35 male Wistar rats determining the evolution over time of the tissue response to aseptic peritoneal damage. A standardized bowel and peritoneal lesions were created in the right lower quadrant by laparotomy. Then, tissular expression of growth factors was evaluated by multiplex polymerase chain reaction at seven timepoints between 6 h and 30 days, postoperatively. Results Tissular responses of granulocyte-stimulating factors (Csf2, Csf3), connective tissue growth factor (Ctgf), epidermal growth factors and receptor (Egf, Egfr), fibroblast growth factors (Fgf2, 7 and 10), heparin binding EGF-like growth factor (Hbegf), hepatocyte growth factor (Hgf), insulin-like growth factor-1 (Igf1), mitogenic transforming growth factors (Tgfa, Tgfb1, Tgfbr3), and vascular endothelial growth factor A (Vegfa) were biphasic with a first expression peak at day 3, followed by a more pronounced peak at day 14. Conclusions We observed a long-lasting, widespread response of tissular growth factors for at least two weeks after peritoneal damage. To be clinically effective, the prophylaxis of postoperative adhesions might be needed for an extended period of time.
Collapse
|
10
|
Yao X, Ajani JA, Song S. Molecular biology and immunology of gastric cancer peritoneal metastasis. Transl Gastroenterol Hepatol 2020; 5:57. [PMID: 33073052 DOI: 10.21037/tgh.2020.02.08] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 02/03/2020] [Indexed: 12/24/2022] Open
Abstract
Peritoneal metastases occur in 55-60% of patients with gastric cancer (GC) and are associated with a 2% 5-year overall survival rate. There are limited treatment options for these patients, and no targeted therapy or immunotherapy is available. Rational therapeutic targets remain to be found. In this review, we present the published literature and our own recent experience in molecular biology to identify important molecules and signaling pathways as well as cellular immunity involved in the peritoneal metastasis of GC. We also suggest potential novel strategies for improving the outcomes of GC patients with peritoneal metastasis.
Collapse
Affiliation(s)
- Xiaodan Yao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shumei Song
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
11
|
Yang Z, Qu Z, Yi M, Lv Z, Wang Y, Shan Y, Ran N, Liu X. MiR-204-5p Inhibits Transforming Growth Factor-β1-Induced Proliferation and Extracellular Matrix Production of Airway Smooth Muscle Cells by Regulating Six1 in Asthma. Int Arch Allergy Immunol 2020; 181:239-248. [PMID: 31955160 DOI: 10.1159/000505064] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 11/27/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Transforming growth factor-β1 (TGF-β1)-in-duced proliferation of airway smooth muscle cells plays critical roles in the development of airway remodeling. Six1 (sine oculis homeobox homolog 1) has been demonstrated to be involved in airway inflammation and remodeling in asthmatic mice. OBJECTIVES The aim of this work was to investigate the potential role of miR-204-5p in the proliferation and extracellular matrix (ECM) production of airway smooth muscle cells in asthma. METHODS Real-time PCR was used to measure the expression of miR-204-5p in asthmatic airway smooth muscle cells. Cell viability and apoptosis were detected to evaluate the effect of miR-204-5p on airway smooth muscle cells. Dual-luciferase reporter experiments were applied to identify the target genes of miR-204-5p. RESULTS MiR-204-5p was downregulated notably in asthmatic airway smooth muscle cells as well as cells stimulated with TGF-β1. Overexpression of miR-204-5p markedly suppressed the TGF-β1-induced proliferation of airway smooth muscle cells and the deposition of ECM, whereas the inhibition of miR-204-5p significantly enhanced the proliferation of airway smooth muscle cells and upregulated the level of fibronectin and collagen III. Furthermore, subsequent analyses demonstrated that Six1 was a direct target of miR-204-5p, and Western blot further indicated that miR-204-5p negatively regulated the expression of Six1. Most importantly, the restoration of Six1 expression reversed the inhibitory effect of miR-204-5p on TGF-β1-induced proliferation and ECM production. CONCLUSIONS MiR-204-5p inhibits TGF-β1-in-duced proliferation and ECM production of airway smooth muscle cells by regulating Six1, identifying a potential therapeutic target for preventing airway remodeling in asthma.
Collapse
Affiliation(s)
- Zhaochuan Yang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China.,Department of Child Health Care, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhenghai Qu
- Center of Pediatrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mingji Yi
- Department of Child Health Care, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhidong Lv
- Breast Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanxia Wang
- Department of Child Health Care, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanchun Shan
- Department of Child Health Care, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ni Ran
- Department of Child Health Care, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xinjie Liu
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China,
| |
Collapse
|
12
|
Role of Growth Factors in the Adhesive Process in the Abdominal Cavity. ACTA BIOMEDICA SCIENTIFICA 2019. [DOI: 10.29413/abs.2019-4.5.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Fibroproliferative diseases have been described in the lungs, kidneys, liver, eyes, heart, skin, and abdomen. Each fibrous pathology has causal factors and pathological manifestations characteristic of this organ and this condition. However, there are common mechanisms underlying many fibrous pathologies. This gives potential value to studies focused on specific processes, among which is the adhesive process in the abdominal cavity. The study of growth factors in the formation of connective tissue contributes to a better understanding of the pathogenetic picture in this pathology. It is shown that vascular endothelial growth factor, fibroblast growth factor, transforming growth factor, platelet growth factor, insulin-like growth factor and keratinocyte growth factor participate in the development of abdominal adhesions. Each of these factors contributes significantly to the formation of peritoneal fibrosis.Thus, damage to any tissue initiates a complex multistage process, which is regulated by a large number of cytokines and growth factors. Growth factors control cell migration, proliferation, differentiation, and survival. In addition, they are able to influence the expression of other factors involved in the regenerative response. Understanding the process that develops during the formation of the adhesive process in the abdominal cavity and the growth factors affecting it is important for their further use in order to prevent the pathological process.
Collapse
|
13
|
Ashizawa N, Miyazaki T, Abe S, Takazono T, Saijo T, Obata Y, Shimamura S, Yamamoto K, Imamura Y, Koji T, Nishino T, Izumikawa K, Yanagihara K, Kohno S, Mukae H. Evaluation of Candida peritonitis with underlying peritoneal fibrosis and efficacy of micafungin in murine models of intra-abdominal candidiasis. Sci Rep 2019; 9:9331. [PMID: 31249356 PMCID: PMC6597535 DOI: 10.1038/s41598-019-45776-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 06/14/2019] [Indexed: 12/30/2022] Open
Abstract
Candida peritonitis is a crucial disease, however the optimal antifungal therapy regimen has not been clearly defined. Peritoneal fibrosis (PF) can be caused by abdominal surgery, intra-abdominal infection, and malignant diseases, and is also widely recognized as a crucial complication of long-term peritoneal dialysis. However, the influence of PF on Candida peritonitis prognosis remains unknown. Here, we evaluated the severity of Candida peritonitis within the context of PF and the efficacy of micafungin using mice. A PF mouse model was generated by intraperitoneally administering chlorhexidine gluconate. Candida peritonitis, induced by intraperitoneal inoculation of Candida albicans, was treated with a 7-day consecutive subcutaneous administration of micafungin. Candida infection caused a higher mortality rate in the PF mice compared with the control mice on day 7. Proliferative Candida invasion into the peritoneum and intra-abdominal organs was confirmed pathologically only in the PF mice. However, all mice in both groups treated with micafungin survived until day 20. Micafungin treatment tends to suppress inflammatory cytokines in the plasma 12 h after infection in both groups. Our results suggest that PF enhances early mortality in Candida peritonitis. Prompt initiation and sufficient doses of micafungin had good efficacy for Candida peritonitis, irrespective of the underlying PF.
Collapse
Affiliation(s)
- Nobuyuki Ashizawa
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, Japan.,Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, Japan
| | - Taiga Miyazaki
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, Japan. .,Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, Japan.
| | - Shinichi Abe
- Department of Nephrology Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, Japan
| | - Takahiro Takazono
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, Japan.,Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, Japan
| | - Tomomi Saijo
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, Japan
| | - Yoko Obata
- Department of Nephrology Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, Japan.,Medical Education Development Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, Japan
| | - Shintaro Shimamura
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, Japan
| | - Kazuko Yamamoto
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, Japan
| | - Yoshifumi Imamura
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, Japan
| | - Takehiko Koji
- Department of Histology and Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, Japan
| | - Tomoya Nishino
- Department of Nephrology Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, Japan
| | - Koichi Izumikawa
- Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, Japan
| | - Katsunori Yanagihara
- Department of Laboratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, Japan
| | - Shigeru Kohno
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, Japan
| | - Hiroshi Mukae
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, Japan.,Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, Japan
| |
Collapse
|
14
|
Yin S, Miao Z, Tan Y, Wang P, Xu X, Zhang C, Hou W, Huang J, Xu H. SPHK1-induced autophagy in peritoneal mesothelial cell enhances gastric cancer peritoneal dissemination. Cancer Med 2019; 8:1731-1743. [PMID: 30791228 PMCID: PMC6488120 DOI: 10.1002/cam4.2041] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 01/10/2019] [Accepted: 01/31/2019] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer peritoneal dissemination (GCPD) has been recognized as the most common form of metastasis in advanced gastric cancer (GC), and the survival is pessimistic. The injury of mesothelial cells plays an important role in GCPD. However, its molecular mechanism is not entirely clear. Here, we focused on the sphingosine kinase 1 (SPHK1) in human peritoneal mesothelial cells (HPMCs) which regulates HPMCs autophagy in GCPD progression. Initially, we analyzed SPHK1 expression immunohistochemically in 120 GC peritoneal tissues, and found high SPHK1 expression to be significantly associated with LC3B expression and peritoneal recurrence, leading to poor prognosis. Using a coculture system, we observed that GC cells promoted HPMCs autophagy and this process was inhibited by blocking TGF-β1 secreted from GC cells. Autophagic HPMCs induced adhesion and invasion of GC cells. We also confirmed that knockdown of SPHK1 expression in HPMCs inhibited TGF-β1-induced autophagy. In addition, SPHK1-driven autophagy of HPMCs accelerated GC cells occurrence of GCPD in vitro and in vivo. Moreover, we explored the relationship between autophagy and fibrosis in HPMCs, observing that overexpression of SPHK1 induced HPMCs fibrosis, while the inhibition of autophagy weakened HPMCs fibrosis. Taken together, our results provided new insights for understanding the mechanisms of GCPD and established SPHK1 as a novel target for GCPD.
Collapse
Affiliation(s)
- Songcheng Yin
- Department of Surgical Oncology, First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Gastric Cancer Molecular Pathology of Liaoning Province, Heping District, Shenyang, China
| | - Zhifeng Miao
- Department of Surgical Oncology, First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Gastric Cancer Molecular Pathology of Liaoning Province, Heping District, Shenyang, China
| | - Yuen Tan
- Department of Surgical Oncology, First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Gastric Cancer Molecular Pathology of Liaoning Province, Heping District, Shenyang, China
| | - Pengliang Wang
- Department of Surgical Oncology, First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Gastric Cancer Molecular Pathology of Liaoning Province, Heping District, Shenyang, China
| | - Xiaoyu Xu
- Department of Gynecology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Chao Zhang
- Department of Surgical Oncology, First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Gastric Cancer Molecular Pathology of Liaoning Province, Heping District, Shenyang, China
| | - Wenbin Hou
- Department of Surgical Oncology, First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Gastric Cancer Molecular Pathology of Liaoning Province, Heping District, Shenyang, China
| | - Jinyu Huang
- Department of Surgical Oncology, First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Gastric Cancer Molecular Pathology of Liaoning Province, Heping District, Shenyang, China
| | - Huimian Xu
- Department of Surgical Oncology, First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Gastric Cancer Molecular Pathology of Liaoning Province, Heping District, Shenyang, China
| |
Collapse
|
15
|
Zheng P, Wang W, Ji M, Zhu Q, Feng Y, Zhou F, He Q. TMEM119 promotes gastric cancer cell migration and invasion through STAT3 signaling pathway. Onco Targets Ther 2018; 11:5835-5844. [PMID: 30271166 PMCID: PMC6145364 DOI: 10.2147/ott.s164045] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Objective TMEM119 is a member of transmembrane proteins family, which is abnormally expressed in human cancers and associated with tumorigenesis. In this study, we focused on the expression of TMEM119 and its role in cell invasion and migration in gastric cancer. Methods Real-time polymerase chain reaction, Western blotting, and immunohistochemistry were performed to examine the expression of TMEM119 in gastric cancer tissues and cell lines. After transfection with TMEM119 siRNA or recombined TMEM119-expressing vector, the invasion and migration ability of MKN45 and SGC-7901 cells was measured by transwell assay. The expression of TMEM119, p-STAT3, STAT3, VEGF, MMP2, and MMP9 proteins in SGC-7901 and MKN45 cells treated with TMEM119 siRNA, TMEM119-expressing vector, or AG490 was measured by Western blotting. Results We found that higher TMEM119 expression was found in gastric cancer tissues and cell lines and was associated with lower survival rate. TMEM119 knockdown inhibited SGC-7901 cell invasion and migration, along with the expression of p-STAT3, VEGF, MMP2, and MMP9. TMEM119 overexpression promoted MKN45 cell invasion and migration, along with the expression of p-STAT3, VEGF, MMP2, and MMP9. Additionally, AG490 treatment significantly corrected TMEM119-induced MKN45 cell migration and invasion and expression of p-STAT3, VEGF, MMP9, and MMP2 proteins. Conclusion The results indicated that TMEM119 promotes gastric cancer cell migration and invasion through activation of STAT3 signaling pathway, and TMEM119 may therefore act as a novel therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Peifen Zheng
- Department of Gastroenterology, Zhejiang Hospital, Hangzhou, People's Republic of China,
| | - Weifeng Wang
- Department of Gastroenterology, Zhejiang Hospital, Hangzhou, People's Republic of China,
| | - Muxi Ji
- Department of Gastroenterology, Zhejiang Hospital, Hangzhou, People's Republic of China,
| | - Qin Zhu
- Department of Gastroenterology, Zhejiang Hospital, Hangzhou, People's Republic of China,
| | - Yuliang Feng
- Department of Gastroenterology, Zhejiang Hospital, Hangzhou, People's Republic of China,
| | - Feng Zhou
- Department of Gastroenterology, Zhejiang Hospital, Hangzhou, People's Republic of China,
| | - Qiaona He
- Department of Gastroenterology, Zhejiang Hospital, Hangzhou, People's Republic of China,
| |
Collapse
|
16
|
Chen KB, Chen J, Jin XL, Huang Y, Su QM, Chen L. Exosome-mediated peritoneal dissemination in gastric cancer and its clinical applications. Biomed Rep 2018; 8:503-509. [PMID: 29774141 DOI: 10.3892/br.2018.1088] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/11/2018] [Indexed: 12/21/2022] Open
Abstract
The prognosis of patients with peritoneal dissemination from gastric cancer is poor, and the underlying molecular mechanism remains unclear. Exosomes, as macromolecular phospholipid bilayer vesicles comprising of proteins, nucleic acids and lipids, serve as mediators of cell-cell communication. Gastric cancer tumor-derived exosomes may be involved in the pathological process of peritoneal dissemination by mediating crosstalk between cancer cells and mesothelial cells, to result in the induction of enhanced tumor growth, migratory, adhesive and invasive abilities, peritoneal fibrosis and apoptosis, mesothelial-to-mesenchymal transition, angiogenesis and chemoresistance. The present review focuses on previous studies addressing the exosome-dependent molecular transfer in peritoneal dissemination in gastric cancer and the potential clinical applications.
Collapse
Affiliation(s)
- Kai-Bo Chen
- Department of General Surgery, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Jian Chen
- Department of General Surgery, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Xiao-Li Jin
- Department of General Surgery, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Yi Huang
- Department of General Surgery, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Qiu-Ming Su
- Department of General Surgery, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Li Chen
- Department of General Surgery, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| |
Collapse
|
17
|
Sharma A, Thakur R, Lingaraju MC, Kumar D, Mathesh K, Telang AG, Singh TU, Kumar D. Betulinic acid attenuates renal fibrosis in rat chronic kidney disease model. Biomed Pharmacother 2017; 89:796-804. [DOI: 10.1016/j.biopha.2017.01.181] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 01/26/2017] [Accepted: 01/31/2017] [Indexed: 12/20/2022] Open
|