1
|
Su H, Chen L, Wu J, Cheng Z, Li J, Ren Y, Xu J, Dang Y, Zheng M, Cao Y, Gao J, Dai C, Hu X, Xie H, Chen J, Luo T, Zhu J, Wu C, Sha W, Chen C, Liu H. Proteogenomic characterization reveals tumorigenesis and progression of lung cancer manifested as subsolid nodules. Nat Commun 2025; 16:2414. [PMID: 40069142 PMCID: PMC11897189 DOI: 10.1038/s41467-025-57364-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
Lung adenocarcinoma (LUAD) radiologically displayed as subsolid nodules (SSNs) is prevalent. Nevertheless, the precise clinical management of SSNs necessitates a profound understanding of their tumorigenesis and progression. Here, we analyze 66 LUAD displayed as SSNs covering 3 histological stages including adenocarcinoma in situ (AIS), minimally invasive adenocarcinoma (MIA) and invasive adenocarcinoma (IAC) by incorporating genomics, proteomics, phosphoproteomics and glycoproteomics. Intriguingly, cholesterol metabolism is aberrantly regulated in the preneoplastic AIS stage. Importantly, target ablation of proprotein convertase subtilisin/kexin type 9 (PCSK9) promotes the initiation of LUAD. Furthermore, sustained endoplasmic reticulum stress is demonstrated to be a hallmark and a reliable biomarker of AIS progression to IAC. Consistently, target promotion of ER stress profoundly retards LUAD progression. Our study provides comprehensive proteogenomic landscape of SSNs, sheds lights on the tumorigenesis and progression of SSNs and suggests preventive and therapeutic strategies for LUAD.
Collapse
Affiliation(s)
- Hang Su
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Li Chen
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Jun Wu
- Center for Bioinformatics and Computational Biology, and the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Zhongyi Cheng
- Jingjie PTM BioLab (Hangzhou). Co. Inc, Hangzhou, 310000, China
| | - Jing Li
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Yijiu Ren
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Junfang Xu
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Yifang Dang
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Mengge Zheng
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Yajuan Cao
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Jiani Gao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Chenyang Dai
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Xuefei Hu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Huikang Xie
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Jianxia Chen
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Tao Luo
- Jingjie PTM BioLab (Hangzhou). Co. Inc, Hangzhou, 310000, China
| | - Jun Zhu
- Jingjie PTM BioLab (Hangzhou). Co. Inc, Hangzhou, 310000, China
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| | - Wei Sha
- Department of tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| | - Haipeng Liu
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| |
Collapse
|
2
|
Su R, Sun X, Luo Y, Gu L, Wang F, Dong A, Yamamoto M, Tsukamoto T, Nomura S, Zhao Z, Dai C, Deng G, Zhuang B, He Y, Zhang C, Yin S. SUSD2 + cancer-associated fibroblasts in gastric cancer mediate the effect of immunosuppression and predict overall survival and the effectiveness of neoadjuvant immunotherapy. Gastric Cancer 2025; 28:245-263. [PMID: 39656339 DOI: 10.1007/s10120-024-01572-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/25/2024] [Indexed: 02/21/2025]
Abstract
BACKGROUND The expression patterns and functions of Sushi Domain Containing 2 (SUSD2) differ among various malignancies. This research aims to investigate the expression of SUSD2 and the role of the SUSD2+ cancer-associated fibroblasts (CAFs) for immunotherapy in gastric cancer. METHODS The expression of SUSD2 and specific markers (CD4, CD8, PD-1, TIGIT, TIM-3 and CD163) was determined using immunohistochemistry and multiplex immunofluorescence (mIHC) on paraffin sections. Flow cytometry and western blot were used to assess the expression of SUSD2 in fibroblasts from fresh samples. Also, analysis of single-cell and bulk RNA sequencing was employed to confirm the presence and characterize the function of SUSD2+ CAFs. The predictive power of indicators for neoadjuvant immunotherapy was evaluated via ROC curve analysis. Animal experiment was employed to validate the immunosuppressive effect of SUSD2+ CAFs. RESULTS SUSD2 is mainly expressed on fibroblasts within the tumors and the high infiltration of SUSD2+ CAFs went together with a poor survival and a more advanced tumor stage. Significantly, the joint use of SUSD2+ CAFs and CD8+ T cells demonstrated a remarkable ability to predict the efficacy of neoadjuvant immunotherapy superior to PD-L1 combined positive score. High SUSD2+ CAFs was correlated with resistance to immunotherapy as well as low CD8+ T infiltration and high exhausted T cell infiltration. CONCLUSIONS We have identified a novel subset of CAFs that could predict the survival and response to neoadjuvant immunotherapy of patients. The SUSD2+ CAFs have the potential to serve as a predictive biomarker and a promising target for immunotherapy.
Collapse
Affiliation(s)
- Rishun Su
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Xuezeng Sun
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Yusheng Luo
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Liang Gu
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Fulin Wang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Aoran Dong
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Masami Yamamoto
- Laboratory of Physiological Pathology, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Tetsuya Tsukamoto
- Department of Diagnostic Pathology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Sachiyo Nomura
- Department of Clinical Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Tokyo, Japan
| | - Zhenzhen Zhao
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Chen Dai
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Guofei Deng
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Baoding Zhuang
- Hepatic-Biliary-Pancreatic Surgery, The Second People's Hospital of Foshan, Foshan, Guangdong Province, China
| | - Yulong He
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Changhua Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Songcheng Yin
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China.
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China.
| |
Collapse
|
3
|
Jeong Y, Kim H, You D, Cho SY, Yoon SY, Kim SW, Nam SJ, Lee JE, Kim S. Induction of SUSD2 by STAT3 Activation Is Associated with Tumor Recurrence in HER2-Positive Breast Cancer. Cells 2024; 14:19. [PMID: 39791720 PMCID: PMC11719728 DOI: 10.3390/cells14010019] [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: 10/17/2024] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025] Open
Abstract
Sushi domain-containing protein 2 (SUSD2), a transmembrane protein containing a sushi motif, has been reported to have tumor-promoting functions in various types of cancer, including breast cancer. However, the regulatory mechanism of SUSD2 and its function in HER2-positive (HER2+) breast cancer have not been fully identified as yet. In this study, we explored the potential of targeting SUSD2 to overcome trastuzumab (TRZ) resistance in HER2+ breast cancer. SUSD2 expression was found to be significantly increased in HER2-overexpressing cells. Endogenous SUSD2 expression was observed in HER2+ breast cancer cells but not in estrogen receptor-positive or triple-negative breast cancer cells. We also found that SUSD2 expression was positively correlated with HER2 expression in a publicly available human primary breast cancer dataset. Although SUSD2 expression was associated with HER2, its expression levels were not affected by TRZ. Through kinase array experiments, we found that SUSD2 expression was modulated downstream of STAT3-dependent signaling in breast cancer cells overexpressing HER2. STAT3 activity was increased in EGFR+ HER2+ breast cancer cells compared to EGFR+ cells. Furthermore, we observed that SUSD2 expression was decreased by C188-9, a STAT3-specific inhibitor. Finally, we analyzed the association between patient survival and SUSD2 expression in breast cancer. Our results showed that SUSD2 expression had a negative correlation with the relapse-free survival of patients with EGFR+ HER2+ breast cancer when compared to EGFR+ breast cancer patients. Collectively, our results demonstrate that SUSD2 expression is mediated by STAT3 and imply the potential of using SUSD2 as a biomarker to stratify HER2+ breast cancer.
Collapse
Affiliation(s)
- Yisun Jeong
- Department of Breast Cancer Center, Samsung Medical Center, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea; (Y.J.); (H.K.)
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Hyungjoo Kim
- Department of Breast Cancer Center, Samsung Medical Center, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea; (Y.J.); (H.K.)
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Daeun You
- Department of Breast Cancer Center, Samsung Medical Center, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea; (Y.J.); (H.K.)
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Soo Youn Cho
- Department of Breast Cancer Center, Samsung Medical Center, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea; (Y.J.); (H.K.)
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Sun Young Yoon
- Department of Breast Cancer Center, Samsung Medical Center, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea; (Y.J.); (H.K.)
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Seok Won Kim
- Department of Breast Cancer Center, Samsung Medical Center, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea; (Y.J.); (H.K.)
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Seok Jin Nam
- Department of Breast Cancer Center, Samsung Medical Center, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea; (Y.J.); (H.K.)
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Jeong Eon Lee
- Department of Breast Cancer Center, Samsung Medical Center, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea; (Y.J.); (H.K.)
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Sangmin Kim
- Department of Breast Cancer Center, Samsung Medical Center, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea; (Y.J.); (H.K.)
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea
| |
Collapse
|
4
|
Bai H, Xian N, Zhao F, Zhou Y, Qin S. The dual role of SUSD2 in cancer development. Eur J Pharmacol 2024; 977:176754. [PMID: 38897441 DOI: 10.1016/j.ejphar.2024.176754] [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: 03/05/2024] [Revised: 06/04/2024] [Accepted: 06/16/2024] [Indexed: 06/21/2024]
Abstract
Sushi domain-containing protein 2 (SUSD2, also known as the complement control protein domain) is a representative and vital protein in the SUSD protein family involved in many physiological and pathological processes beyond complement regulation. Cancer is one of the leading causes of death worldwide. The complex role of SUSD2 in tumorigenesis and cancer progression has raised increasing concerns. Studies suggest that SUSD2 has different regulatory tendencies among different tumors and exerts its biological effects in a cancer type-specific manner; for instance, it has oncogenic effects on breast cancer, gastric cancer, and glioma and has tumor-suppression effects on lung cancer, bladder cancer, and colon cancer. Moreover, SUSD2 can be regulated by noncoding RNAs, its promoter methylation and other molecules, such as Galectin-1 (Gal-1), tropomyosin alpha-4 chain (TPM4), and p63. The therapeutic implications of targeting SUSD2 have already been preliminarily revealed in some malignancies, including melanoma, colon cancer, and breast cancer. This article reviews the role and regulatory mechanisms of SUSD2 in cancer development, as well as its structure and distribution. We hope that this review will advance the understanding of SUSD2 as a diagnostic and/or prognostic biomarker and provide new avenues for the development of novel cancer therapies.
Collapse
Affiliation(s)
- Han Bai
- The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an, 710000, China
| | - Ningyi Xian
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Fengyu Zhao
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yikun Zhou
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Sida Qin
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
5
|
Kuo WT, Lee YC, Yang YF, Cheng CF, Tseng CJ, Tsai KW. Sushi Domain Containing 2 Dysfunction Contributes to Cancer Progression in Patients with Bladder Cancer. J Cancer 2024; 15:5318-5328. [PMID: 39247587 PMCID: PMC11375537 DOI: 10.7150/jca.97537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/13/2024] [Indexed: 09/10/2024] Open
Abstract
Bladder cancer is the most prevalent type of cancer in Taiwan, and therefore, enhancing the diagnostic sensitivity of biomarkers for early-stage tumors and identifying therapeutic targets to improve patient survival rates are essential. Although Sushi Domain Containing 2 (SUSD2) dysfunction has been identified in several types of human cancer, its biological role in bladder cancer remains unclear. Analysis of The Cancer Genome Atlas revealed significantly higher expression of SUSD2 mRNA in bladder cancer tissues than in adjacent normal tissues. This elevated expression of SUSD2 significantly correlated with pathological stage (p = 0.029), pN stage (p < 0.001), and pM stage (p = 0.047). Univariate analysis revealed that high SUSD2 expression was associated with decreased overall survival (crude hazard ratio = 1.70, 95% confidence interval = 1.13-2.56, p = 0.01). Multivariate analysis revealed a significant correlation between high SUSD2 expression and poor survival outcomes (adjusted hazard ratio = 1.53, 95% confidence interval = 1.01-2.31, p = 0.043). IHC analysis revealed a significant correlation between elevated SUSD2 protein levels and unfavorable pathological stages (p < 0.001). SUSD2 suppression significantly reduced the proliferation, colony formation, and invasion of bladder cancer cells. In addition, cell cycle analysis revealed that SUSD2 knockdown induced G2/M phase arrestin bladder cancer cells. Tumor Immune Estimation Resource analysis indicated that expression of SUSD2 was significantly associated with macrophage infiltration and M2 macrophage polarization in bladder cancer. In addition, miR-383-5p directly targeted the 3'UTR of SUSD2, with its ectopic expression inhibiting the growth and motility of bladder cancer cells. Our study revealed that miR-383-5p/SUSD2 axis dysfunction may contribute to a poor prognosis for bladder cancer by affecting cell growth, metastasis, and the tumor microenvironment.
Collapse
Affiliation(s)
- Wei-Ting Kuo
- Division of Urology, Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taiwan
| | - Yi-Chen Lee
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Fang Yang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Ching-Feng Cheng
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Ching-Jiunn Tseng
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taiwan
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Kuo-Wang Tsai
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- Department of Nursing, Cardinal Tien Junior College of Healthcare and Management, Taiwan
| |
Collapse
|
6
|
Rivera M, Toledo-Jacobo L, Romero E, Oprea TI, Moses ME, Hudson LG, Wandinger-Ness A, Grimes MM. Agent-based modeling predicts RAC1 is critical for ovarian cancer metastasis. Mol Biol Cell 2022; 33:ar138. [PMID: 36200848 PMCID: PMC9727804 DOI: 10.1091/mbc.e21-11-0540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Experimental and computational studies pinpoint rate-limiting step(s) in metastasis governed by Rac1. Using ovarian cancer cell and animal models, Rac1 expression was manipulated, and quantitative measurements of cell-cell and cell-substrate adhesion, cell invasion, mesothelial clearance, and peritoneal tumor growth discriminated the tumor behaviors most highly influenced by Rac1. The experimental data were used to parameterize an agent-based computational model simulating peritoneal niche colonization, intravasation, and hematogenous metastasis to distant organs. Increased ovarian cancer cell survival afforded by the more rapid adhesion and intravasation upon Rac1 overexpression is predicted to increase the numbers of and the rates at which tumor cells are disseminated to distant sites. Surprisingly, crowding of cancer cells along the blood vessel was found to decrease the numbers of cells reaching a distant niche irrespective of Rac1 overexpression or knockdown, suggesting that sites for tumor cell intravasation are rate limiting and become accessible if cells intravasate rapidly or are displaced due to diminished viability. Modeling predictions were confirmed through animal studies of Rac1-dependent metastasis to the lung. Collectively, the experimental and modeling approaches identify cell adhesion, rapid intravasation, and survival in the blood as parameters in the ovarian metastatic cascade that are most critically dependent on Rac1.
Collapse
Affiliation(s)
- Melanie Rivera
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Leslie Toledo-Jacobo
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Elsa Romero
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Tudor I. Oprea
- Division of Translational Informatics, Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131,Translational Informatics, Roivant Discovery, Boston, MA 02210
| | - Melanie E. Moses
- Department of Computer Science, University of New Mexico, Albuquerque, NM 87131
| | - Laurie G. Hudson
- Cancer Research Facility, Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131,Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Angela Wandinger-Ness
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131,Cancer Research Facility, Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131,*Address correspondence to: Angela Wandinger-Ness ()
| | - Martha M. Grimes
- Cancer Research Facility, Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131,Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| |
Collapse
|
7
|
Zhao B, Gong W, Ma A, Chen J, Velegraki M, Dong H, Liu Z, Wang L, Okimoto T, Jones DM, Lei YL, Long M, Oestreich KJ, Ma Q, Xin G, Carbone DP, He K, Li Z, Wen H. SUSD2 suppresses CD8 + T cell antitumor immunity by targeting IL-2 receptor signaling. Nat Immunol 2022; 23:1588-1599. [PMID: 36266363 PMCID: PMC9669207 DOI: 10.1038/s41590-022-01326-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 09/08/2022] [Indexed: 11/09/2022]
Abstract
Dysfunctional CD8+ T cells, which have defective production of antitumor effectors, represent a major mediator of immunosuppression in the tumor microenvironment. Here, we show that SUSD2 is a negative regulator of CD8+ T cell antitumor function. Susd2-/- effector CD8+ T cells showed enhanced production of antitumor molecules, which consequently blunted tumor growth in multiple syngeneic mouse tumor models. Through a quantitative mass spectrometry assay, we found that SUSD2 interacted with interleukin (IL)-2 receptor α through sushi domain-dependent protein interactions and that this interaction suppressed the binding of IL-2, an essential cytokine for the effector functions of CD8+ T cells, to IL-2 receptor α. SUSD2 was not expressed on regulatory CD4+ T cells and did not affect the inhibitory function of these cells. Adoptive transfer of Susd2-/- chimeric antigen receptor T cells induced a robust antitumor response in mice, highlighting the potential of SUSD2 as an immunotherapy target for cancer.
Collapse
Affiliation(s)
- Bao Zhao
- Department of Microbial Infection and Immunity, Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Weipeng Gong
- Department of Microbial Infection and Immunity, Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
| | - Anjun Ma
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Jianwen Chen
- Department of Microbial Infection and Immunity, Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
| | - Maria Velegraki
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Hong Dong
- Department of Microbial Infection and Immunity, Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
| | - Zihao Liu
- Department of Microbial Infection and Immunity, Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
| | - Lingling Wang
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Internal Medicine, Division of Hematology, The Ohio State University, Columbus, OH, USA
| | - Tamio Okimoto
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
| | - Devin M Jones
- Department of Microbial Infection and Immunity, Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
| | - Yu L Lei
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Meixiao Long
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Internal Medicine, Division of Hematology, The Ohio State University, Columbus, OH, USA
| | - Kenneth J Oestreich
- Department of Microbial Infection and Immunity, Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
| | - Qin Ma
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Gang Xin
- Department of Microbial Infection and Immunity, Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - David P Carbone
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
| | - Kai He
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
| | - Zihai Li
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Haitao Wen
- Department of Microbial Infection and Immunity, Infectious Disease Institute, The Ohio State University, Columbus, OH, USA.
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
8
|
Neville MC, Webb PG, Baumgartner HK, Bitler BG. Claudin-4 localization in epithelial ovarian cancer. Heliyon 2022; 8:e10862. [PMID: 36237976 PMCID: PMC9552118 DOI: 10.1016/j.heliyon.2022.e10862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/06/2022] [Accepted: 09/28/2022] [Indexed: 11/19/2022] Open
Abstract
Claudin-4, a protein with the structure of classic claudins most often found in cell-cell junctions, is frequently overexpressed in epithelial cancers where its localization has not been studied. In this study we aimed to find out where this membrane protein is localized in an ovarian tumor model, OVCAR3 cells, that express high levels of the protein. Immunohistochemical studies showed claudin-4 staining in a perinuclear region, at most plasma membranes and in cytoplasmic puncta. Native claudin-4 did not overlap with phosphorylated claudin-4, which was partially located in focal adhesions. Using claudin-4 BioID technology we confirmed that large amounts of claudin-4 are localized to the Golgi compartment, including in dispersed Golgi in cells where claudin-4 is partially knocked down and in dividing cells. Claudin-4 appears to be present in the vicinity of several types of cell-cell junctions, but there is no evidence that it forms tight junctions in these tumor cells. Both claudin-4, the Golgi marker GM130, and the plasma membrane receptor Notch2 were found in dispersed Golgi in dividing cells. This definition of the cellular architecture of claudin-4 should provide a framework for better understanding of the function of claudin-4 in tumor cells and its molecular interactions.
Collapse
Affiliation(s)
- Margaret C. Neville
- Departments of Obstetrics and Gynecology and Physiology and Biophysics, University of Colorado School of Medicine, Aurora, CO, 80845, USA
- Corresponding author.
| | - Patricia G. Webb
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, 80845, USA
| | - Heidi K. Baumgartner
- University of Colorado Anschutz Medical Campus, 2700 E. 19th Ave., Aurora, CO, 80045, USA
| | - Benjamin G. Bitler
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, University of Colorado Denver Anschutz Medical Campus, Mail Stop 8613, 12700 E. 19 Ave., Aurora, CO, 80045, USA
| |
Collapse
|
9
|
Xing L, Tian S, Mi W, Zhang Y, Zhang Y, Zhang Y, Xu F, Zhang C, Lou G. PRSS1 Upregulation Predicts Platinum Resistance in Ovarian Cancer Patients. Front Cell Dev Biol 2021; 8:618341. [PMID: 33585454 PMCID: PMC7876278 DOI: 10.3389/fcell.2020.618341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/28/2020] [Indexed: 12/21/2022] Open
Abstract
Ovarian cancer is the most frequent cause of death among gynecologic malignancies. A total of 80% of patients who have completed platinum-based chemotherapy suffer from relapse and develop resistance within 2 years. In the present study, we obtained patients' complete platinum (cisplatin and carboplatin) medication information from The Cancer Genome Atlas database and then divided them into two categories: resistance and sensitivity. Difference analysis was performed to screen differentially expressed genes (DEgenes) related to platinum response. Subsequently, we annotated DEgenes into the protein–protein interaction network as seed nodes and analyzed them by random walk. Finally, second-ranking protease serine 1 gene (PRSS1) was selected as a candidate gene for verification analysis. PRSS1's expression pattern was continuously studied in Oncomine and cBio Cancer Genomic Portal databases, revealing the key roles of PRSS1 in ovarian cancer formation. Hereafter, we conducted in-depth explorations on PRSS1's platinum response to ovarian cancer through tissue and cytological experiments. Quantitative real-time polymerase chain reaction and Western blot assay results indicated that PRSS1 expression levels in platinum-resistant samples (tissue/cell) were significantly higher than in samples sensitive to platinum. By cell transfection assay, we observed that knockdown of PRSS1 reduced the resistance of ovarian cancer cells to cisplatin. Meanwhile, overexpression of PRSS1 increased the resistance to cisplatin. In conclusion, we identified a novel risk gene PRSS1 related to ovarian cancer platinum response and confirmed its key roles using multiple levels of low-throughput experiments, revealing a new treatment strategy based on a novel target factor for overcoming cisplatin resistance in ovarian cancer.
Collapse
Affiliation(s)
- Linan Xing
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Songyu Tian
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wanqi Mi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yongjian Zhang
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yunyan Zhang
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuxi Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Fengye Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Chunlong Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Ge Lou
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
10
|
Sheets JN, Patrick ME, Egland KA. SUSD2 expression correlates with decreased metastasis and increased survival in a high-grade serous ovarian cancer xenograft murine model. Oncotarget 2020; 11:2290-2301. [PMID: 32595828 PMCID: PMC7299533 DOI: 10.18632/oncotarget.27626] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 05/20/2020] [Indexed: 11/25/2022] Open
Abstract
The cause of death among high-grade serous ovarian cancer (HGSOC) patients involves passive dissemination of cancer cells within the peritoneal cavity and subsequent implantation of cancer spheroids into adjacent organs. Sushi DomainContaining 2 (SUSD2) encodes a type I transmembrane protein containing several functional domains inherent to adhesion molecules. Previous studies using in vitro methods have indicated that SUSD2 functions as a tumor suppressor in several cancers, including HGSOC. In this study, we generated a HGSOC xenograft mouse model to investigate SUSD2 expression in the context of HGSOC late-stage metastasis and overall survival. OVCAR3 cells with knock-down expression of SUSD2 (OVCAR3 SUSD2-KD) or endogenous expression of SUSD2 (OVCAR3-Non-Targeting (NT)) were injected into the peritoneal cavity of athymic nude mice. Immunohistochemistry analysis was utilized to identify infiltrating cancer cells and metastatic tumors in mouse ovaries, pancreas, spleen, omentum and liver. OVCAR3-NT mice developed significantly less cancer cell infiltrate and tumors in their pancreas and omentum compared to OVCAR3 SUSD2-KD mice. Furthermore, OVCAR3-NT mice displayed a longer median survival when compared to OVCAR3 SUSD2-KD mice (175 days and 185.5 days, respectively; p-value 0.0159). Altogether, the findings generated through the preclinical mouse model suggest that increased SUSD2 expression in HGSOC impedes in vivo metastasis to pancreas and omentum. These results correlate to longer median survival and prove to be consistent with previous findings showing prolonged survival of HGSOC patients with high SUSD2-expressing primary tumors.
Collapse
Affiliation(s)
- Jordan N Sheets
- Cancer Biology & Immunotherapies Group, Sanford Research, Sanford School of Medicine of The University of South Dakota, Sioux Falls, SD, USA
| | - Mitch E Patrick
- Cancer Biology & Immunotherapies Group, Sanford Research, Sanford School of Medicine of The University of South Dakota, Sioux Falls, SD, USA
| | - Kristi A Egland
- Cancer Biology & Immunotherapies Group, Sanford Research, Sanford School of Medicine of The University of South Dakota, Sioux Falls, SD, USA.,SAb Biotherapeutics, Sioux Falls, SD, USA
| |
Collapse
|
11
|
Guo W, Shao F, Sun S, Song P, Guo L, Xue X, Zhang G, Zhang H, Gao Y, Qiu B, Tan F, Gao S, He J. Loss of SUSD2 expression correlates with poor prognosis in patients with surgically resected lung adenocarcinoma. J Cancer 2020; 11:1648-1656. [PMID: 32194777 PMCID: PMC7052850 DOI: 10.7150/jca.39319] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 12/07/2019] [Indexed: 12/25/2022] Open
Abstract
There is limited evidence regarding the relationship between the expression of Sushi Domain Containing 2 (SUSD2) and prognosis of patients with surgically resected lung adenocarcinoma (LUAD). This retrospective study aimed to investigate the clinical significance of SUSD2 expression in LUAD. To assess SUSD2 expression in LUAD, we conducted both integrated bioinformatic analysis based on the TCGA database and also immunohistochemistry study using a tissue microarray encompassing 578 LUAD cases from our hospital. Reduced SUSD2 expression was associated with gender, smoking history, higher pathological grade, lymph node metastasis, larger tumor length, advanced TNM stage. LUAD patients with SUSD2-positive tumors showed significantly better overall survival (OS) than those with SUSD2-negative tumors (P = 0.000). When patients were stratified into those with stage I (218, 37.7%), II (152, 26.3%) and III (208, 36.0%) disease, and those without (254, 43.9%) and with (324, 56.1%) lymph node metastasis, the prognostic effect was almost consistent. The OS of patients with positive SUSD2 expression was significantly better in patients with stage I (P = 0.000), III (P = 0.000), without (P = 0.000) and with (P = 0.001) lymph node metastasis. Multivariate analysis showed that loss of SUSD2 predicted a shorter survival time and was an independent prognostic factor for LUAD patients. Our study indicated that SUSD2 may serve as a new prognostic and potential therapeutic target in LUAD.
Collapse
Affiliation(s)
- Wei Guo
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, The People's Republic of China
| | - Fei Shao
- The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, Shandong 266071,The People's Republic of China.,Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, The People's Republic of China
| | - Sijin Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, The People's Republic of China
| | - Peng Song
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, The People's Republic of China
| | - Lei Guo
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, The People's Republic of China
| | - Xuemin Xue
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, The People's Republic of China
| | - Guochao Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, The People's Republic of China
| | - Hao Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, The People's Republic of China
| | - Yibo Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, The People's Republic of China
| | - Bin Qiu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, The People's Republic of China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, The People's Republic of China
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, The People's Republic of China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, The People's Republic of China
| |
Collapse
|
12
|
Ovarian Cancer Dissemination-A Cell Biologist's Perspective. Cancers (Basel) 2019; 11:cancers11121957. [PMID: 31817625 PMCID: PMC6966436 DOI: 10.3390/cancers11121957] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 12/04/2019] [Indexed: 12/16/2022] Open
Abstract
Epithelial ovarian cancer (EOC) comprises multiple disease states representing a variety of distinct tumors that, irrespective of tissue of origin, genetic aberrations and pathological features, share common patterns of dissemination to the peritoneal cavity. EOC peritoneal dissemination is a stepwise process that includes the formation of malignant outgrowths that detach and establish widespread peritoneal metastases through adhesion to serosal membranes. The cell biology associated with outgrowth formation, detachment, and de novo adhesion is at the nexus of diverse genetic backgrounds that characterize the disease. Development of treatment for metastatic disease will require detailed characterization of cellular processes involved in each step of EOC peritoneal dissemination. This article offers a review of the literature that relates to the current stage of knowledge about distinct steps of EOC peritoneal dissemination, with emphasis on the cell biology aspects of the process.
Collapse
|
13
|
Rewiring of Cancer Cell Metabolism by Mitochondrial VDAC1 Depletion Results in Time-Dependent Tumor Reprogramming: Glioblastoma as a Proof of Concept. Cells 2019; 8:cells8111330. [PMID: 31661894 PMCID: PMC6912264 DOI: 10.3390/cells8111330] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/18/2019] [Accepted: 10/23/2019] [Indexed: 12/16/2022] Open
Abstract
Reprograming of the metabolism of cancer cells is an event recognized as a hallmark of the disease. The mitochondrial gatekeeper, voltage-dependent anion channel 1 (VDAC1), mediates transport of metabolites and ions in and out of mitochondria, and is involved in mitochondria-mediated apoptosis. Here, we compared the effects of reducing hVDAC1 expression in a glioblastoma xenograft using human-specific si-RNA (si-hVDAC1) for a short (19 days) and a long term (40 days). Tumors underwent reprograming, reflected in rewired metabolism, eradication of cancer stem cells (CSCs) and differentiation. Short- and long-term treatments of the tumors with si-hVDAC1 similarly reduced the expression of metabolism-related enzymes, and translocator protein (TSPO) and CSCs markers. In contrast, differentiation into cells expressing astrocyte or neuronal markers was noted only after a long period during which the tumor cells were hVDAC1-depleted. This suggests that tumor cell differentiation is a prolonged process that precedes metabolic reprograming and the “disappearance” of CSCs. Tumor proteomics analysis revealing global changes in the expression levels of proteins associated with signaling, synthesis and degradation of proteins, DNA structure and replication and epigenetic changes, all of which were highly altered after a long period of si-hVDAC1 tumor treatment. The depletion of hVDAC1 greatly reduced the levels of the multifunctional translocator protein TSPO, which is overexpressed in both the mitochondria and the nucleus of the tumor. The results thus show that VDAC1 depletion-mediated cancer cell metabolic reprograming involves a chain of events occurring in a sequential manner leading to a reversal of the unique properties of the tumor, indicative of the interplay between metabolism and oncogenic signaling networks.
Collapse
|
14
|
Patrick ME, Egland KA. SUSD2 Proteolytic Cleavage Requires the GDPH Sequence and Inter-Fragment Disulfide Bonds for Surface Presentation of Galectin-1 on Breast Cancer Cells. Int J Mol Sci 2019; 20:E3814. [PMID: 31387209 PMCID: PMC6696261 DOI: 10.3390/ijms20153814] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/24/2019] [Accepted: 07/31/2019] [Indexed: 11/17/2022] Open
Abstract
Galectin-1 (Gal-1) is a 14 kDa protein that has been well characterized for promoting cancer metastasis and tumor immune evasion. By localizing to the cancer cell surface, Gal-1 induces T cell apoptosis through binding T cell surface receptors. The transmembrane protein, Sushi Domain Containing 2 (SUSD2), has been previously shown to be required for Gal-1 surface presentation in breast cancer cells. Western immunoblot analysis revealed that SUSD2 is cleaved into two fragments. However, the significance of this cleavage for Gal-1 surface localization has not been investigated. To define the location of cleavage, a mutagenesis analysis of SUSD2 was performed. Our studies demonstrated that SUSD2 is cleaved at its glycine-aspartic acid-proline-histidine (GDPH) amino acid sequence. Generation of a noncleavable SUSD2 mutant (GDPH∆-SUSD2) showed that SUSD2 cleavage was required for SUSD2 and Gal-1 plasma membrane localization. Noncleavable cysteine mutants were also unable to present Gal-1 at the cell surface, further demonstrating that SUSD2 cleavage is required for Gal-1 surface presentation. Treatment with the serine protease inhibitor, Pefabloc SC, inhibited SUSD2 cleavage in a dose dependent manner, suggesting that SUSD2 is cleaved by a serine protease. Therefore, identification and inhibition of this protease may provide a new therapeutic tool for inhibiting SUSD2 and Gal-1's combined tumorigenic function in breast cancer.
Collapse
Affiliation(s)
- Mitch E Patrick
- Cancer Biology & Immunotherapies Group, Sanford Research, Sanford School of Medicine of the University of South Dakota, Sioux Falls, SD 57104, USA
| | - Kristi A Egland
- Cancer Biology & Immunotherapies Group, Sanford Research, Sanford School of Medicine of the University of South Dakota, Sioux Falls, SD 57104, USA.
| |
Collapse
|
15
|
Oikiri H, Asano Y, Matsusaki M, Akashi M, Shimoda H, Yokoyama Y. Inhibitory effect of carbonyl reductase 1 against peritoneal progression of ovarian cancer: evaluation by ex vivo 3D-human peritoneal model. Mol Biol Rep 2019; 46:4685-4697. [PMID: 31025149 DOI: 10.1007/s11033-019-04788-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 03/28/2019] [Indexed: 12/13/2022]
Abstract
The current authors previously reported that a carbonyl reductase 1 (CR1) DNA-dendrimer complex could potentially be used in gene therapy for peritoneal metastasis of ovarian cancer. The aims of the current study were to observe the cellular dynamics of peritoneal metastasis of epithelial ovarian cancer cells and to ascertain changes in the dynamics of ovarian cancer cells as a result of transfection of CR1 DNA. (1) Artificial human peritoneal tissue (AHPT) was seeded with serous ovarian cancer cells, and the process leading to development of peritoneal carcinomatosis was observed over time. (2) Peritoneal carcinomatosis was produced in mice and compared to a model using AHPT to determine the appropriateness of AHPT. (3) CR1 DNA was transfected into cancer cells seeded on AHPT, and the dynamics of cancer cells were observed over time. (1) Cancer cells perforated the mesothelium, leaving normal mesothelium intact. However, the cells proliferated between the layers of the mesothelium, forming a mass. After 24 h, cancer cells had invaded the lymphatics, and after 48-72 h cancer cells had invaded deep into the mesothelium, where they formed a mass. (2) Invasion of the peritoneum by cancer cells in a murine model of peritoneal carcinomatosis resembled that in a model using AHPT, and results substantiated the reproducibility of peritoneal carcinomatosis in AHPT. (3) Proliferation of cells transfected with CR1 DNA was significantly inhibited on AHPT, and necrosis was evident. Nevertheless, cancer cell invasion deep into the mesothelium was not inhibited. Use of a new tool, AHPT, in an in vitro model of peritoneal metastasis revealed that CR1 DNA inhibited cancer cell proliferation. CR1 DNA does not play a role in inhibiting invasion of the mesothelium during peritoneal metastasis, but it does affect cancer cell proliferation. Results suggested that CR1 DNA inhibits cancer cell proliferation via necrosis.
Collapse
Affiliation(s)
- Hiroe Oikiri
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Hirosaki University, 5 Zaifu, Hirosaki, 036-8562, Japan
| | - Yoshiya Asano
- Department of Neuroanatomy, Cell Biology and Histology, Graduate School of Medicine, Hirosaki University, 5 Zaifu, Hirosaki, 036-8562, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 1-3 Yamada-oka, Osaka, 565-0871, Japan
| | - Mitsuru Akashi
- Building Block Science, Graduate School of Frontier Biosciences, Osaka University, 2-1 Yamada-oka, Osaka, 565-0871, Japan
| | - Hiroshi Shimoda
- Department of Neuroanatomy, Cell Biology and Histology, Graduate School of Medicine, Hirosaki University, 5 Zaifu, Hirosaki, 036-8562, Japan.,Department of Anatomical Science, Graduate School of Medicine, Hirosaki University, 5 Zaifu, Hirosaki, 036-8562, Japan
| | - Yoshihito Yokoyama
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Hirosaki University, 5 Zaifu, Hirosaki, 036-8562, Japan.
| |
Collapse
|
16
|
Lager TW, Roetman JJ, Kunkel J, Thacker M, Sheets JN, Egland KA, Miles CM, Larson MK, Gubbels JAA. Sushi Domain Containing 2 (SUSD2) inhibits platelet activation and binding to high-grade serous ovarian carcinoma cells. Platelets 2018; 29:834-837. [PMID: 30335544 DOI: 10.1080/09537104.2018.1530345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Platelets play a central role in primary hemostasis affecting tumor survival and metastases. Tumors induce platelets to aggregate and bind to the cancer cells, resulting in protection from immune surveillance and often leading to thrombocytosis. In ovarian cancer (OvCa), one-third of patients present with thrombocytosis, a diagnosis that correlates with shorter survival. SUSD2 (SUShi Domain containing 2), a type I transmembrane protein, shown to inhibit metastatic processes in high-grade serous ovarian carcinoma (HGSOC), is expressed on endothelial cells and thus may influence platelet reactivity. As such, we hypothesized that SUSD2 levels in ovarian cancer-derived cell lines influence platelet activation. We incubated OvCa non-targeting (NT) and SUSD2 knockdown (KD) cell lines with labeled platelets and quantified platelet binding, as well as GPIIb/IIIa integrin activation. The role of GPIIb/IIIa in tumor cell/platelet interaction was also examined by measuring cell-cell adhesion in the presence of eptifibatide. We found that platelets exposed to OvCa cells with low SUSD2 expression display increased tumor cell-platelet binding along with an increase in GPIIb/IIIa receptor activation. As such, platelet activation and binding to HGSOC cells was inversely correlated with the presence of SUSD2. This represents one of the first tumor proteins known to provide differential platelet interaction based on protein status.
Collapse
Affiliation(s)
- Tyson W Lager
- a Department of Biology , Augustana University , Sioux Falls , SD , USA
| | - Jessica J Roetman
- a Department of Biology , Augustana University , Sioux Falls , SD , USA
| | - Jacob Kunkel
- a Department of Biology , Augustana University , Sioux Falls , SD , USA
| | - Megan Thacker
- a Department of Biology , Augustana University , Sioux Falls , SD , USA
| | - Jordan N Sheets
- b Cancer Biology Research Center, Sanford Research , Sanford School of Medicine of the University of South Dakota , Sioux Falls , SD , USA
| | - Kristi A Egland
- b Cancer Biology Research Center, Sanford Research , Sanford School of Medicine of the University of South Dakota , Sioux Falls , SD , USA
| | - Cecelia M Miles
- a Department of Biology , Augustana University , Sioux Falls , SD , USA
| | - Mark K Larson
- a Department of Biology , Augustana University , Sioux Falls , SD , USA
| | | |
Collapse
|
17
|
Umeda S, Kanda M, Miwa T, Tanaka H, Tanaka C, Kobayashi D, Suenaga M, Hattori N, Hayashi M, Yamada S, Nakayama G, Fujiwara M, Kodera Y. Expression of sushi domain containing two reflects the malignant potential of gastric cancer. Cancer Med 2018; 7:5194-5204. [PMID: 30259711 PMCID: PMC6198216 DOI: 10.1002/cam4.1793] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 08/07/2018] [Accepted: 08/31/2018] [Indexed: 12/19/2022] Open
Abstract
Hepatic recurrence of gastric cancer (GC) is uncontrollable. Discovery of causative oncogenes and the development of sensitive biomarkers to predict hepatic recurrence are required to improve patients’ outcomes. In this study, recurrence pattern‐specific transcriptome analysis of 57 749 genes was conducted to identify mRNAs specifically associated with hepatic metastasis of patients with stage III GC who underwent curative resection. GC cell lines were subjected to mRNA expression analysis, PCR array analysis, and siRNA‐mediated knockdown. The expression levels of primary cancer tissues from 154 patients with resectable GC were determined and correlated with clinicopathological variables. Among 21 genes significantly overexpressed specifically in patients with hepatic recurrence, Sushi domain containing 2 (SUSD2) was selected as a promising target. PCR array analysis revealed that SUSD2 mRNA levels positively correlated with those of FZD7, CDH2, TGFB1, SPARC, ITGA5, and ZEB1. Functional analysis revealed that knockdown of SUSD2 significantly reduced the proliferation, migration, and invasiveness GC cell lines. Patients with high SUSD2 expression were more likely to experience shorter disease‐free and overall survival. Analysis of the relation between disease recurrence pattern and SUSD2 levels revealed that significantly more patients with hepatic metastases expressed higher levels of SUSD2 mRNA. The cumulative incidence of hepatic recurrence was greater in patients with high SUSD2 expression. In conclusion, SUSD2 likely contributes to the malignant potential of GC and may serve as a novel biomarker that predicts hepatic recurrence after curative resection.
Collapse
Affiliation(s)
- Shinichi Umeda
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mitsuro Kanda
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Miwa
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Haruyoshi Tanaka
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Chie Tanaka
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Kobayashi
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masaya Suenaga
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norifumi Hattori
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masamichi Hayashi
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Suguru Yamada
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Goro Nakayama
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Michitaka Fujiwara
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
18
|
Tang J, Pulliam N, Özeş A, Buechlein A, Ding N, Keer H, Rusch D, O'Hagan H, Stack MS, Nephew KP. Epigenetic Targeting of Adipocytes Inhibits High-Grade Serous Ovarian Cancer Cell Migration and Invasion. Mol Cancer Res 2018; 16:1226-1240. [PMID: 29759990 PMCID: PMC6072573 DOI: 10.1158/1541-7786.mcr-17-0406] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 11/16/2017] [Accepted: 04/27/2018] [Indexed: 01/17/2023]
Abstract
Ovarian cancer (OC) cells frequently metastasize to the omentum, and adipocytes play a significant role in ovarian tumor progression. Therapeutic interventions targeting aberrant DNA methylation in ovarian tumors have shown promise in the clinic, but the effects of epigenetic therapy on the tumor microenvironment are understudied. Here, we examined the effect of adipocytes on OC cell behavior in culture and impact of targeting DNA methylation in adipocytes on OC metastasis. The presence of adipocytes increased OC cell migration and invasion, and proximal and direct coculture of adipocytes increased OC proliferation alone or after treatment with carboplatin. Treatment of adipocytes with hypomethylating agent guadecitabine decreased migration and invasion of OC cells toward adipocytes. Subcellular protein fractionation of adipocytes treated with guadecitabine revealed decreased DNA methyltransferase 1 (DNMT1) levels even in the presence of DNA synthesis inhibitor, aphidicolin. Methyl-Capture- and RNA-sequencing analysis of guadecitabine-treated adipocytes revealed derepression of tumor-suppressor genes and epithelial-mesenchymal transition inhibitors. SUSD2, a secreted tumor suppressor downregulated by promoter CpG island methylation in adipocytes, was upregulated after guadecitabine treatment, and recombinant SUSD2 decreased OC cell migration and invasion. Integrated analysis of the methylomic and transcriptomic data identified pathways associated with inhibition of matrix metalloproteases and fatty acid α-oxidation, suggesting a possible mechanism of how epigenetic therapy of adipocytes decreases metastasis. In conclusion, the effect of DNMT inhibitor on fully differentiated adipocytes suggests that hypomethylating agents may affect the tumor microenvironment to decrease cancer cell metastasis.Implications: Epigenetic targeting of tumor microenvironment can affect metastatic behavior of ovarian cancer cells. Mol Cancer Res; 16(8); 1226-40. ©2018 AACR.
Collapse
Affiliation(s)
- Jessica Tang
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana
| | - Nicholas Pulliam
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana
- Molecular and Cellular Biochemistry Department, Indiana University, Bloomington, Indiana
| | - Ali Özeş
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana
- Molecular and Cellular Biochemistry Department, Indiana University, Bloomington, Indiana
| | - Aaron Buechlein
- Center of Genomics and Bioinformatics, Indiana University, Bloomington, Indiana
| | - Ning Ding
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana
| | - Harold Keer
- Astex Pharmaceuticals Inc., Pleasanton, California
| | - Doug Rusch
- Center of Genomics and Bioinformatics, Indiana University, Bloomington, Indiana
| | - Heather O'Hagan
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana University Simon Cancer Center, Indianapolis, Indiana
| | - M Sharon Stack
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
| | - Kenneth P Nephew
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana.
- Molecular and Cellular Biochemistry Department, Indiana University, Bloomington, Indiana
- Indiana University Simon Cancer Center, Indianapolis, Indiana
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
19
|
Novel potential inhibitors of complement system and their roles in complement regulation and beyond. Mol Immunol 2018; 102:73-83. [PMID: 30217334 DOI: 10.1016/j.molimm.2018.05.023] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/02/2018] [Accepted: 05/25/2018] [Indexed: 12/20/2022]
Abstract
The complement system resembles a double-edged sword since its activation can either benefit or harm the host. Thus, regulation of this system is of utmost importance and performed by several circulating and membrane-bound complement inhibitors. The pool of well-established regulators has recently been enriched with proteins that either share structural homology to known complement inhibitors such as Sushi domain-containing (SUSD) protein family and Human CUB and Sushi multiple domains (CSMD) families or extracellular matrix (ECM) macromolecules that interact with and modulate complement activity. In this review, we summarize the current knowledge about newly discovered complement inhibitors and discuss their implications in complement regulation, as well as in processes beyond complement regulation such cancer development. Understanding the behavior of these proteins will introduce new mechanisms of complement regulation and may provide new avenues in the development of novel therapies.
Collapse
|
20
|
Xu Y, Miao C, Jin C, Qiu C, Li Y, Sun X, Gao M, Lu N, Kong B. SUSD2 promotes cancer metastasis and confers cisplatin resistance in high grade serous ovarian cancer. Exp Cell Res 2018; 363:160-170. [DOI: 10.1016/j.yexcr.2017.12.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/26/2017] [Accepted: 12/29/2017] [Indexed: 01/27/2023]
|
21
|
Coelho R, Marcos-Silva L, Ricardo S, Ponte F, Costa A, Lopes JM, David L. Peritoneal dissemination of ovarian cancer: role of MUC16-mesothelin interaction and implications for treatment. Expert Rev Anticancer Ther 2017; 18:177-186. [PMID: 29241375 DOI: 10.1080/14737140.2018.1418326] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Peritoneal dissemination is a particular form of malignant progression in ovarian cancer, preceding hematogenic or lymphatic dissemination. Thus, prevention of peritoneal implantation of cancer cells is envisioned to inhibit neoplastic dissemination and therefore prolong disease remission and patient's survival. Areas covered: An extended review on the role of MUC16 (CA125) and mesothelin (MSLN), expressed in a high percentage of ovarian carcinomas, indicate that this duet is relevant for the contact between cancer cells and mesothelial cells in homotypic (cancer cell-cancer cell) and heterotypic (cancer cell-mesothelial cell) interactions. This review discusses the reasons underlying the clinical failure of immunotherapeutic strategies targeting MUC16. Clinical data on MSLN targeting agents such as antibody-based immunotoxins or antibody drug conjugates are also reviewed. The promising anti-tumor effect of CAR-T cells directed to MUC16 or MSLN is emphasized. New emerging strategies specifically disrupting the MUC16-MSLN interaction are at the forefront of this review, including TRAIL ligands bound to MSLN targeting MUC16 expressing cells and single chain monoclonal antibodies and immunoadhesins recognizing MSLN-MUC16 binding domains. Expert commentary: Based on existing evidences the authors advocate that agents targeting MUC16-MSLN may add to the therapeutic armamentarium directed to abrogate peritoneal homing of ovarian cancer.
Collapse
Affiliation(s)
- Ricardo Coelho
- a Differentiation and Cancer Group, IPATIMUP/i3S , Institute of Molecular Pathology and Immunology of the University of Porto/Institute for Research and Innovation in Health of University of Porto , Porto , Portugal.,b FMUP , Faculty of Medicine of University of Porto , Porto , Portugal
| | - Lara Marcos-Silva
- a Differentiation and Cancer Group, IPATIMUP/i3S , Institute of Molecular Pathology and Immunology of the University of Porto/Institute for Research and Innovation in Health of University of Porto , Porto , Portugal.,c Animal Cell Technology Unit, ITQB, Instituto de Tecnologia Química e Biológica António Xavier , Universidade Nova de Lisboa, Lisboa, Portugal and iBET, Instituto de Biologia Experimental e Tecnológica , Oeiras , Portugal
| | - Sara Ricardo
- a Differentiation and Cancer Group, IPATIMUP/i3S , Institute of Molecular Pathology and Immunology of the University of Porto/Institute for Research and Innovation in Health of University of Porto , Porto , Portugal.,b FMUP , Faculty of Medicine of University of Porto , Porto , Portugal
| | - Filipa Ponte
- a Differentiation and Cancer Group, IPATIMUP/i3S , Institute of Molecular Pathology and Immunology of the University of Porto/Institute for Research and Innovation in Health of University of Porto , Porto , Portugal
| | - Antonia Costa
- b FMUP , Faculty of Medicine of University of Porto , Porto , Portugal.,d Gynecology and Obstetrics Department , Centro hospitalar de São João , Porto , Portugal.,e Monitoring and simulation of perinatal asphyxia group, INEB/i3S, Instituto de Engenharia Biomédica , Universidade do Porto, Porto, Portugal/Institute for Research and Innovation in Health of University of Porto , Porto , Portugal
| | - Jose Manuel Lopes
- b FMUP , Faculty of Medicine of University of Porto , Porto , Portugal.,f Pathology Department , Centro hospitalar de São João , Porto , Portugal.,g Cancer Cell Signalling and Metabolism Group, IPATIMUP/i3S , Institute of Molecular Pathology and Immunology of the University of Porto/Institute for Research and Innovation in Health of University of Porto , Porto , Portugal
| | - Leonor David
- a Differentiation and Cancer Group, IPATIMUP/i3S , Institute of Molecular Pathology and Immunology of the University of Porto/Institute for Research and Innovation in Health of University of Porto , Porto , Portugal.,b FMUP , Faculty of Medicine of University of Porto , Porto , Portugal
| |
Collapse
|
22
|
Downregulation of endometrial mesenchymal marker SUSD2 causes cell senescence and cell death in endometrial carcinoma cells. PLoS One 2017; 12:e0183681. [PMID: 28841682 PMCID: PMC5571916 DOI: 10.1371/journal.pone.0183681] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/09/2017] [Indexed: 12/13/2022] Open
Abstract
The cause of death among the majority of endometrial cancer patients involves migration of cancer cells within the peritoneal cavity and subsequent implantation of cancer spheroids into neighbouring organs. It is, thereby, important to identify factors that mediate metastasis. Cell adhesion and migration are modified by the mesenchymal stem cell (MSC) marker Sushi domain containing 2 (SUSD2), a type I transmembrane protein that participates in the orchestration of cell adhesion and migration through interaction with its partner Galactosidase-binding soluble-1 (LGALS1). MSCs have emerged as attractive targets in cancer therapy. Human endometrial adenocarcinoma (Ishikawa) cells were treated with TGFβ (10 ng/ml) for 72h. SUSD2, LGALS1 and MKI67 transcript levels were quantified using qRT-PCR. The proportion of SUSD2 positive (SUSD2+) cells and SMAD2/3 abundance were quantified by FACS and Western blotting, respectively. Senescent cells were identified with β-galactosidase staining; cell cycle and cell death were quantified using Propidium Iodide staining. Treatment of endometrial cancer cells (Ishikawa cells) with TGFβ (10 ng/ml) significantly decreased SUSD2 transcript levels and the proportion of SUSD2 positive cells. Silencing of SUSD2 using siRNA resulted in senescence and cell death of Ishikawa cells via activation of SMAD2/3. These findings suggest that SUSD2 counteracts senescence and cell death and is thus a potential chemotherapeutic target in human endometrial cancer.
Collapse
|