1
|
Weng C, Jin R, Jin X, Yang Z, He C, Zhang Q, Xu J, Lv B. Exploring the Mechanisms, Biomarkers, and Therapeutic Targets of TRIM Family in Gastrointestinal Cancer. Drug Des Devel Ther 2024; 18:5615-5639. [PMID: 39654601 PMCID: PMC11626976 DOI: 10.2147/dddt.s482340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 11/23/2024] [Indexed: 12/12/2024] Open
Abstract
Gastrointestinal region (GI) cancers are closely linked to the ubiquitination system, with the E3 ubiquitin ligase playing a crucial role by targeting various substrates. As E3 ubiquitin ligases, proteins of tripartite motif (TRIM) family play a role in cancer signaling, development, apoptosis, and formation. These proteins regulate diverse biological activities and signaling pathways. This study comprehensively outlines the functions of TRIM proteins in gastrointestinal physiology, contributing to our knowledge of the molecular pathways involved in gastrointestinal tumors. Gastrointestinal region (GI) cancers are closely linked to the ubiquitination system, with the E3 ubiquitin ligase playing a crucial role by targeting various substrates.
Collapse
Affiliation(s)
- Chunyan Weng
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Rijuan Jin
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Xiaoliang Jin
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Zimei Yang
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Chenghai He
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
- Department of Gastroenterology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Qiuhua Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Jingli Xu
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Bin Lv
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
| |
Collapse
|
2
|
Zeng M, Wang Y, Tao X, Fan T, Yin X, Shen C, Wang X. Novel Perspectives in the Management of Colorectal Cancer: Mechanistic Investigations Into the Reversal of Drug Resistance via Active Constituents Derived From Herbal Medicine. Phytother Res 2024; 38:5962-5984. [PMID: 39462152 DOI: 10.1002/ptr.8363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 09/03/2024] [Accepted: 10/05/2024] [Indexed: 10/29/2024]
Abstract
The high incidence and mortality rate of colorectal cancer have become a significant global health burden. Chemotherapy has been the traditional treatment for colorectal cancer and has demonstrated promising antitumor effects, leading to significant improvements in patient survival. However, the development of chemoresistance poses a major challenge during chemotherapy in colorectal cancer, significantly impeding treatment efficacy and affecting patient prognosis. Despite the development of a variety of novel anticolorectal cancer chemotherapy agents, their effectiveness and side effects vary, possibly due to the complex mechanisms of resistance in colorectal cancer. Abnormal drug metabolism or protein targets are the most direct causes of resistance. Further studies have revealed that these resistance mechanisms involve biochemical processes such as altered protein expression, autophagy, and epithelial-mesenchymal transitions. Herbal active ingredients offer an alternative treatment option and have shown promise in reversing colorectal cancer drug resistance. This paper aims to summarize the role of various biochemical processes and key protein targets in the occurrence and maintenance of resistance mechanisms in colorectal cancer. Additionally, it elaborates on the mechanisms of action of herbal active ingredients in reversing colorectal cancer drug resistance. The article also discusses the limitations and opportunities in developing novel anticolorectal cancer drugs based on herbal medicine.
Collapse
Affiliation(s)
- Mingtang Zeng
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Yao Wang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelin Tao
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Tianfei Fan
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Xi Yin
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Chao Shen
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Xueyan Wang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Zhang X, Yi K, Wang B, Chu K, Liu J, Zhang J, Fang J, Zhao T. EZH2 Activates HTLV-1 bZIP Factor-Mediated TGF-β Signaling in Adult T-Cell Leukemia. J Med Virol 2024; 96:e70025. [PMID: 39530290 DOI: 10.1002/jmv.70025] [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: 08/05/2024] [Revised: 09/20/2024] [Accepted: 10/13/2024] [Indexed: 11/16/2024]
Abstract
Adult T-cell leukemia (ATL) is an aggressive malignancy caused by human T-cell leukemia virus type 1 (HTLV-1) infection. Enhancer of zeste homolog 2 (EZH2) has been implicated in the development and progression of multiple cancers, including virus-induced malignancies. However, the potential function of EZH2 in HTLV-1-induced oncogenesis has not been clearly elucidated. In the present study, we showed that EZH2 was overexpressed and activated in HTLV-1-infected cell lines, potentially due to the activation of EZH2 promoter by HTLV-1 Tax and NF-κB p65 subunit. In addition, we found that EZH2 enhanced the HBZ-induced activation of TGF-β signaling in a histone methyltransferase-independent manner. As a mechanism for these actions, we found that EZH2 targeted Smad3/Smad4 to form a ternary complex, and the association between Smad3 and Smad4 was markedly enhanced in the presence of EZH2. Knockdown of EZH2 in ATL cells indeed repressed the expressions of the TGF-β target genes. In particular, EZH2 synergistically enhanced the HBZ/TGF-β-induced Foxp3 expression. Treatment of 3-Deazaneplanocin A, a specific inhibitor of EZH2 significantly inhibited the Foxp3 expression. Taken together, our results suggest that EZH2 may be involved in the differentiation of regulatory T cells through activating the HBZ-Smad3-TGF-β signaling axis, which is considered to be a key strategy for viral persistence.
Collapse
Affiliation(s)
- Xu Zhang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Kaining Yi
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Bingbing Wang
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Kaifei Chu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Jie Liu
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Jie Zhang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Jiaqi Fang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Tiejun Zhao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| |
Collapse
|
4
|
Nishimura K, Takahara K, Komura K, Ishida M, Hirosuna K, Maenosono R, Ajiro M, Sakamoto M, Iwatsuki K, Nakajima Y, Tsujino T, Taniguchi K, Tanaka T, Inamoto T, Hirose Y, Ono F, Kondo Y, Yoshimi A, Azuma H. Mechanistic insights into lethal hyper progressive disease induced by PD-L1 inhibitor in metastatic urothelial carcinoma. NPJ Precis Oncol 2024; 8:206. [PMID: 39289546 PMCID: PMC11408499 DOI: 10.1038/s41698-024-00707-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024] Open
Abstract
Hyper progressive disease (HPD) is a paradoxical phenomenon characterized by accelerated tumor growth following treatment with immune checkpoint inhibitors. However, the pathogenic causality and its predictor remain unknown. We herein report a fatal case of HPD in a 50-year-old man with metastatic bladder cancer. He had achieved a complete response (CR) through chemoradiation therapy followed by twelve cycles of chemotherapy, maintaining CR for 24 months. Three weeks after initiating maintenance use of a PD-L1 inhibitor, avelumab, a massive amount of metastases developed, leading to the patient's demise. Omics analysis, utilizing metastatic tissues obtained from an immediate autopsy, implied the contribution of M2 macrophages, TGF-β signaling, and interleukin-8 to HPD pathogenesis.
Collapse
Affiliation(s)
- Kazuki Nishimura
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
- Division of Cancer RNA Research, National Cancer Center Research Institute, Chuo-Ku, Tokyo, Japan
| | - Kiyoshi Takahara
- Department of Urology, Fujita-Health University School of Medicine, Toyoake City, Aichi, Japan
| | - Kazumasa Komura
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan.
- Division of Translational Research, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan.
| | - Mitsuaki Ishida
- Department of Pathology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Kensuke Hirosuna
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama City, Okayama, Japan
| | - Ryoichi Maenosono
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
- Division of Cancer RNA Research, National Cancer Center Research Institute, Chuo-Ku, Tokyo, Japan
| | - Masahiko Ajiro
- Division of Cancer RNA Research, National Cancer Center Research Institute, Chuo-Ku, Tokyo, Japan
| | - Moritoshi Sakamoto
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
- Division of Cancer RNA Research, National Cancer Center Research Institute, Chuo-Ku, Tokyo, Japan
| | - Kengo Iwatsuki
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Yuki Nakajima
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Takuya Tsujino
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Kohei Taniguchi
- Division of Translational Research, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Tomohito Tanaka
- Division of Translational Research, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Teruo Inamoto
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Yoshinobu Hirose
- Department of Pathology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Fumihito Ono
- Division of Translational Research, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Yoichi Kondo
- Department of Anatomy and Cell Biology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Akihide Yoshimi
- Division of Cancer RNA Research, National Cancer Center Research Institute, Chuo-Ku, Tokyo, Japan.
| | - Haruhito Azuma
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| |
Collapse
|
5
|
Jiang YL, Li X, Tan YW, Fang YJ, Liu KY, Wang YF, Ma T, Ou QJ, Zhang CX. Docosahexaenoic acid inhibits the invasion and migration of colorectal cancer by reversing EMT through the TGF-β1/Smad signaling pathway. Food Funct 2024; 15:9420-9433. [PMID: 39189524 DOI: 10.1039/d4fo02346c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
The primary cause of mortality in colorectal cancer (CRC) patients is tumor metastasis. The epithelial-mesenchymal transition (EMT) stands out as a crucial factor promoting the metastasis of CRC. Previous findings suggest a potential inhibitory effect of docosahexaenoic acid (DHA) on CRC metastasis, but the precise mechanism remains unknown, this study aims to explore this issue. We assessed metastasis and recurrence, all-cause mortality, and cancer-related mortality rates according to DHA intake in independent CRC cohorts (n = 367) by survival analysis. The ability of DHA to block CRC cell migration and invasion was tested using transwell and wound-healing assays. The regulation of EMT marker genes in CRC by DHA was detected by quantitative real-time PCR (qPCR) and immunoblotting, and the effect of DHA on the TGF-β1/Smad signaling pathway was further investigated. These cellular findings were validated using a subcutaneous CRC mouse model. Survival analyses showed that lower DHA intake was associated with a higher risk of CRC metastasis and a poorer prognosis. In vitro experiments showed that DHA inhibits the TGF-β1/Smad signaling pathway and regulates downstream transcription factors, thereby reversing the EMT and inhibiting invasion and migration. In the mouse model, dietary DHA supplementation effectively increased blood DHA concentrations and inhibited CRC metastasis. Our study demonstrated that DHA inhibits CRC invasion and metastasis by inhibiting the TGF-β1/Smad signaling pathway. Increased intake of DHA among CRC patients may provide additional benefits to the prognosis of colorectal cancer.
Collapse
Affiliation(s)
- Yi-Ling Jiang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Xue Li
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Ya-Wen Tan
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Yu-Jing Fang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Kai-Yan Liu
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Yi-Fan Wang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Ting Ma
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Qing-Jian Ou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Cai-Xia Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
6
|
Yang M, Mu Y, Yu X, Gao D, Zhang W, Li Y, Liu J, Sun C, Zhuang J. Survival strategies: How tumor hypoxia microenvironment orchestrates angiogenesis. Biomed Pharmacother 2024; 176:116783. [PMID: 38796970 DOI: 10.1016/j.biopha.2024.116783] [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: 03/15/2024] [Revised: 05/07/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024] Open
Abstract
During tumor development, the tumor itself must continuously generate new blood vessels to meet their growth needs while also allowing for tumor invasion and metastasis. One of the most common features of tumors is hypoxia, which drives the process of tumor angiogenesis by regulating the tumor microenvironment, thus adversely affecting the prognosis of patients. In addition, to overcome unsuitable environments for growth, such as hypoxia, nutrient deficiency, hyperacidity, and immunosuppression, the tumor microenvironment (TME) coordinates angiogenesis in several ways to restore the supply of oxygen and nutrients and to remove metabolic wastes. A growing body of research suggests that tumor angiogenesis and hypoxia interact through a complex interplay of crosstalk, which is inextricably linked to the TME. Here, we review the TME's positive contribution to angiogenesis from an angiogenesis-centric perspective while considering the objective impact of hypoxic phenotypes and the status and limitations of current angiogenic therapies.
Collapse
Affiliation(s)
- Mengrui Yang
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Yufeng Mu
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Xiaoyun Yu
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Dandan Gao
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Wenfeng Zhang
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Ye Li
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, 999078, Macao Special Administrative Region of China
| | - Jingyang Liu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, 999078, Macao Special Administrative Region of China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang 261053, China; Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261000, China.
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261000, China.
| |
Collapse
|
7
|
Wu Z, Ke Q, Jiang L, Hong H, Pan W, Chen W, Abudukeremu X, She F, Chen Y. TGF-β1 facilitates gallbladder carcinoma metastasis by regulating FOXA1 translation efficiency through m 6A modification. Cell Death Dis 2024; 15:422. [PMID: 38886389 PMCID: PMC11183149 DOI: 10.1038/s41419-024-06800-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 05/22/2024] [Accepted: 05/31/2024] [Indexed: 06/20/2024]
Abstract
TGF-β1 plays a pivotal role in the metastatic cascade of malignant neoplasms. N6-methyladenosine (m6A) stands as one of the most abundant modifications on the mRNA transcriptome. However, in the metastasis of gallbladder carcinoma (GBC), the effect of TGF-β1 with mRNA m6A modification, especially the effect of mRNA translation efficiency associated with m6A modification, remains poorly elucidated. Here we demonstrated a negative correlation between FOXA1 and TGF-β1 expression in GBC. Overexpression of FOXA1 inhibited TGF-β1-induced migration and epithelial-mesenchymal transition (EMT) in GBC cells. Mechanistically, we confirmed that TGF-β1 suppressed the translation efficiency of FOXA1 mRNA through polysome profiling analysis. Importantly, both in vivo and in vitro experiments showed that TGF-β1 promoted m6A modification on the coding sequence (CDS) region of FOXA1 mRNA, which was responsible for the inhibition of FOXA1 mRNA translation by TGF-β1. We demonstrated through MeRIP and RIP assays, dual-luciferase reporter assays and site-directed mutagenesis that ALKBH5 promoted FOXA1 protein expression by inhibiting m6A modification on the CDS region of FOXA1 mRNA. Moreover, TGF-β1 inhibited the binding capacity of ALKBH5 to the FOXA1 CDS region. Lastly, our study confirmed that overexpression of FOXA1 suppressed lung metastasis and EMT in a nude mice lung metastasis model. In summary, our research findings underscore the role of TGF-β1 in regulating TGF-β1/FOXA1-induced GBC EMT and metastasis by inhibiting FOXA1 translation efficiency through m6A modification.
Collapse
Affiliation(s)
- Zhenheng Wu
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
- Fujian Medical University Cancer Center, Fuzhou, Fujian, 350122, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fujian Medical University, Fuzhou, Fujian, 350122, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, Fujian, 350122, China
| | - Qiming Ke
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
- Fujian Medical University Cancer Center, Fuzhou, Fujian, 350122, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fujian Medical University, Fuzhou, Fujian, 350122, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, Fujian, 350122, China
| | - Lei Jiang
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
- Fujian Medical University Cancer Center, Fuzhou, Fujian, 350122, China
| | - Haijie Hong
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
- Fujian Medical University Cancer Center, Fuzhou, Fujian, 350122, China
| | - Wei Pan
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
- Fujian Medical University Cancer Center, Fuzhou, Fujian, 350122, China
| | - Wen Chen
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
- Fujian Medical University Cancer Center, Fuzhou, Fujian, 350122, China
| | - Xiahenazi Abudukeremu
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
- Fujian Medical University Cancer Center, Fuzhou, Fujian, 350122, China
| | - Feifei She
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fujian Medical University, Fuzhou, Fujian, 350122, China.
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, Fujian, 350122, China.
| | - Yanling Chen
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China.
- Fujian Medical University Cancer Center, Fuzhou, Fujian, 350122, China.
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fujian Medical University, Fuzhou, Fujian, 350122, China.
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, Fujian, 350122, China.
| |
Collapse
|
8
|
Li L, Sun X, Zhang M, Zhang B, Yang Y, Wang S. FOXN2, identified as a novel biomarker in serum, modulates the transforming growth factor-beta signaling pathway through its interaction with partitioning defective 6 homolog alpha, contributing to the pathogenesis of gastric cancer. Growth Factors 2024; 42:62-73. [PMID: 38954805 DOI: 10.1080/08977194.2023.2297700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 12/07/2023] [Indexed: 07/04/2024]
Abstract
BACKGROUND AND OBJECTIVE Dysregulated expression of Forkhead Box N2 (FOXN2) has been detected in various cancer types. However, the underlying mechanisms by which FOXN2 contributes to the onset and progression of gastric cancer (GC) remain largely unexplored. This study aimed to elucidate the potential role of FOXN2 within GC, its downstream molecular mechanisms, and its feasibility as a novel serum biomarker for GC. METHODS Tissue samples from GC patients and corresponding non-cancerous tissues were collected. Peripheral blood samples were obtained from GC patients and healthy controls. The expression of FOXN2 was determined using quantitative real-time PCR, western blotting, and immunohistochemistry. The expression of FOXN2 in GC cells was modulated by transfection with small interfering RNA (siRNA) or the pcDNA 3.1 expression vector. Cell proliferation was assessed using the Cell Counting Kit-8 and 5-ethynyl-2'-deoxyuridine incorporation assays. The migratory and invasive capacities of cells were evaluated by Transwell assays, apoptosis rates were measured by flow cytometry, and the expression of proliferative, apoptotic, and epithelial-mesenchymal transition (EMT) markers were assessed by western blot analysis. RESULTS FOXN2 was found to be overexpressed in the serum, tissues, and cells of GC, correlating with distant metastasis and TNM staging. FOXN2 demonstrated diagnostic value in differentiating GC patients from healthy individuals, with higher levels of FOXN2 being indicative of poorer survival rates. Silencing FOXN2 in vitro inhibited the proliferation, invasion, migration, and EMT of GC cells, while promoting apoptosis. FOXN2 was shown to regulate the transforming growth factor-beta (TGFβ) receptor signaling pathway in GC cells via its interaction with Partitioning Defective 6 Homolog Alpha (PARD6A). CONCLUSION In summary, our data suggest that FOXN2 acts as an oncogenic factor in GC, modulating the TGFβ pathway by binding to PARD6A, thereby influencing gastric carcinogenesis. This study underscores the functional significance of FOXN2 as a potential serum biomarker and therapeutic target in GC.
Collapse
Affiliation(s)
- Liang Li
- Department of Gastroenterology, Three Gorges Hospital Affiliated to Chongqing University, Chongqing City, China
| | - XueFeng Sun
- Department of Gastroenterology, Three Gorges Hospital Affiliated to Chongqing University, Chongqing City, China
| | - Mei Zhang
- Department of Gastroenterology, Three Gorges Hospital Affiliated to Chongqing University, Chongqing City, China
| | - BangShuo Zhang
- Department of Hematology, Three Gorges Hospital Affiliated to Chongqing University, Chongqing City, China
| | - Yi Yang
- Department of Hematology, Three Gorges Hospital Affiliated to Chongqing University, Chongqing City, China
| | - Sheng Wang
- Department of Rheumatology and Immunology, Three Gorges Hospital Affiliated to Chongqing University, Chongqing City, China
| |
Collapse
|
9
|
Shichijo T, Yasunaga JI, Sato K, Nosaka K, Toyoda K, Watanabe M, Zhang W, Koyanagi Y, Murphy EL, Bruhn RL, Koh KR, Akari H, Ikeda T, Harris RS, Green PL, Matsuoka M. Vulnerability to APOBEC3G linked to the pathogenicity of deltaretroviruses. Proc Natl Acad Sci U S A 2024; 121:e2309925121. [PMID: 38502701 PMCID: PMC10990082 DOI: 10.1073/pnas.2309925121] [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: 06/13/2023] [Accepted: 01/29/2024] [Indexed: 03/21/2024] Open
Abstract
Human retroviruses are derived from simian ones through cross-species transmission. These retroviruses are associated with little pathogenicity in their natural hosts, but in humans, HIV causes AIDS, and human T-cell leukemia virus type 1 (HTLV-1) induces adult T-cell leukemia-lymphoma (ATL). We analyzed the proviral sequences of HTLV-1, HTLV-2, and simian T-cell leukemia virus type 1 (STLV-1) from Japanese macaques (Macaca fuscata) and found that APOBEC3G (A3G) frequently generates G-to-A mutations in the HTLV-1 provirus, whereas such mutations are rare in the HTLV-2 and STLV-1 proviruses. Therefore, we investigated the mechanism of how HTLV-2 is resistant to human A3G (hA3G). HTLV-1, HTLV-2, and STLV-1 encode the so-called antisense proteins, HTLV-1 bZIP factor (HBZ), Antisense protein of HTLV-2 (APH-2), and STLV-1 bZIP factor (SBZ), respectively. APH-2 efficiently inhibits the deaminase activity of both hA3G and simian A3G (sA3G). HBZ and SBZ strongly suppress sA3G activity but only weakly inhibit hA3G, suggesting that HTLV-1 is incompletely adapted to humans. Unexpectedly, hA3G augments the activation of the transforming growth factor (TGF)-β/Smad pathway by HBZ, and this activation is associated with ATL cell proliferation by up-regulating BATF3/IRF4 and MYC. In contrast, the combination of APH-2 and hA3G, or the combination of SBZ and sA3G, does not enhance the TGF-β/Smad pathway. Thus, HTLV-1 is vulnerable to hA3G but utilizes it to promote the proliferation of infected cells via the activation of the TGF-β/Smad pathway. Antisense factors in each virus, differently adapted to control host cellular functions through A3G, seem to dictate the pathogenesis.
Collapse
Affiliation(s)
- Takafumi Shichijo
- Department of Hematology, Rheumatology and Infectious Diseases, Faculty of Life Sciences, Kumamoto University, Kumamoto860-8556, Japan
- Laboratory of Virus Control, Institute for Life and Medical Sciences, Kyoto University, Kyoto606-8507, Japan
| | - Jun-ichirou Yasunaga
- Department of Hematology, Rheumatology and Infectious Diseases, Faculty of Life Sciences, Kumamoto University, Kumamoto860-8556, Japan
- Laboratory of Virus Control, Institute for Life and Medical Sciences, Kyoto University, Kyoto606-8507, Japan
| | - Kei Sato
- Division of Systems Virology, Institute of Medical Science, The University of Tokyo, Tokyo108-8639, Japan
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Saitama332-0012, Japan
| | - Kisato Nosaka
- Department of Hematology, Rheumatology and Infectious Diseases, Faculty of Life Sciences, Kumamoto University, Kumamoto860-8556, Japan
| | - Kosuke Toyoda
- Department of Hematology, Rheumatology and Infectious Diseases, Faculty of Life Sciences, Kumamoto University, Kumamoto860-8556, Japan
- Laboratory of Virus Control, Institute for Life and Medical Sciences, Kyoto University, Kyoto606-8507, Japan
| | - Miho Watanabe
- Department of Hematology, Rheumatology and Infectious Diseases, Faculty of Life Sciences, Kumamoto University, Kumamoto860-8556, Japan
| | - Wenyi Zhang
- Department of Hematology, Rheumatology and Infectious Diseases, Faculty of Life Sciences, Kumamoto University, Kumamoto860-8556, Japan
| | - Yoshio Koyanagi
- Laboratory of Systems Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto606-8507, Japan
| | - Edward L. Murphy
- Department of Laboratory Medicine, University of California, San Francisco94158
- Department of Epidemiology/Biostatistics, University of California, San Francisco
- Vitalant Research Institute, San Francisco94105
| | | | - Ki-Ryang Koh
- Department of Hematology, Osaka General Hospital of West Japan Railway Company, Osaka545-0053, Japan
| | - Hirofumi Akari
- Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama, Aichi484-8506, Japan
| | - Terumasa Ikeda
- Division of Molecular Virology and Genetics, Joint Research Center for Human Retrovirus infection, Kumamoto University, Kumamoto860-0811, Japan
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX78229
- HHMI, University of Texas Health San Antonio, San Antonio, TX78229
| | - Reuben S. Harris
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX78229
- HHMI, University of Texas Health San Antonio, San Antonio, TX78229
| | - Patrick L. Green
- Center for Retrovirus Research, Department of Veterinary Biosciences, The Ohio State University, Columbus, OH43210
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH
| | - Masao Matsuoka
- Department of Hematology, Rheumatology and Infectious Diseases, Faculty of Life Sciences, Kumamoto University, Kumamoto860-8556, Japan
- Laboratory of Virus Control, Institute for Life and Medical Sciences, Kyoto University, Kyoto606-8507, Japan
| |
Collapse
|
10
|
Lughmani H, Patel H, Chakravarti R. Structural Features and Physiological Associations of Human 14-3-3ζ Pseudogenes. Genes (Basel) 2024; 15:399. [PMID: 38674334 PMCID: PMC11049341 DOI: 10.3390/genes15040399] [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: 03/05/2024] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
There are about 14,000 pseudogenes that are mutated or truncated sequences resembling functional parent genes. About two-thirds of pseudogenes are processed, while others are duplicated. Although initially thought dead, emerging studies indicate they have functional and regulatory roles. We study 14-3-3ζ, an adaptor protein that regulates cytokine signaling and inflammatory diseases, including rheumatoid arthritis, cancer, and neurological disorders. To understand how 14-3-3ζ (gene symbol YWHAZ) performs diverse functions, we examined the human genome and identified nine YWHAZ pseudogenes spread across many chromosomes. Unlike the 32 kb exon-to-exon sequence in YWHAZ, all pseudogenes are much shorter and lack introns. Out of six, four YWHAZ exons are highly conserved, but the untranslated region (UTR) shows significant diversity. The putative amino acid sequence of pseudogenes is 78-97% homologous, resulting in striking structural similarities with the parent protein. The OMIM and Decipher database searches revealed chromosomal loci containing pseudogenes are associated with human diseases that overlap with the parent gene. To the best of our knowledge, this is the first report on pseudogenes of the 14-3-3 family protein and their implications for human health. This bioinformatics-based study introduces a new insight into the complexity of 14-3-3ζ's functions in biology.
Collapse
Affiliation(s)
| | | | - Ritu Chakravarti
- Department of Physiology and Pharmacology, The University of Toledo, Toledo, OH 43614, USA; (H.L.); (H.P.)
| |
Collapse
|
11
|
Boudreault J, Wang N, Ghozlan M, Lebrun JJ. Transforming Growth Factor-β/Smad Signaling Inhibits Melanoma Cancer Stem Cell Self-Renewal, Tumor Formation and Metastasis. Cancers (Basel) 2024; 16:224. [PMID: 38201651 PMCID: PMC10778361 DOI: 10.3390/cancers16010224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 12/20/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
The secreted protein transforming growth factor-beta (TGFβ) plays essential roles, ranging from cell growth regulation and cell differentiation in both normal and cancer cells. In melanoma, TGFβ acts as a potent tumor suppressor in melanoma by blocking cell cycle progression and inducing apoptosis. In the present study, we found TGFβ to regulate cancer stemness in melanoma through the Smad signaling pathway. We discovered that TGFβ/Smad signaling inhibits melanosphere formation in multiple melanoma cell lines and reduces expression of the CD133+ cancer stem cell subpopulation in a Smad3-dependent manner. Using preclinical models of melanoma, we further showed that preventing Smad3/4 signaling, by means of CRISPR knockouts, promoted both tumorigenesis and lung metastasis in vivo. Collectively, our results define new functions for the TGFβ/Smad signaling axis in melanoma stem-cell maintenance and open avenues for new therapeutic approaches to this disease.
Collapse
Affiliation(s)
| | | | | | - Jean-Jacques Lebrun
- Cancer Research Program, Department of Medicine, Research Institute of McGill University Health Center, Montreal, QU H4A 3J1, Canada; (J.B.); (N.W.); (M.G.)
| |
Collapse
|
12
|
Arnhold J. Inflammation-Associated Cytotoxic Agents in Tumorigenesis. Cancers (Basel) 2023; 16:81. [PMID: 38201509 PMCID: PMC10778456 DOI: 10.3390/cancers16010081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 12/16/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Chronic inflammatory processes are related to all stages of tumorigenesis. As inflammation is closely associated with the activation and release of different cytotoxic agents, the interplay between cytotoxic agents and antagonizing principles is highlighted in this review to address the question of how tumor cells overcome the enhanced values of cytotoxic agents in tumors. In tumor cells, the enhanced formation of mitochondrial-derived reactive species and elevated values of iron ions and free heme are antagonized by an overexpression of enzymes and proteins, contributing to the antioxidative defense and maintenance of redox homeostasis. Through these mechanisms, tumor cells can even survive additional stress caused by radio- and chemotherapy. Through the secretion of active agents from tumor cells, immune cells are suppressed in the tumor microenvironment and an enhanced formation of extracellular matrix components is induced. Different oxidant- and protease-based cytotoxic agents are involved in tumor-mediated immunosuppression, tumor growth, tumor cell invasion, and metastasis. Considering the special metabolic conditions in tumors, the main focus here was directed on the disturbed balance between the cytotoxic agents and protective mechanisms in late-stage tumors. This knowledge is mandatory for the implementation of novel anti-cancerous therapeutic approaches.
Collapse
Affiliation(s)
- Jürgen Arnhold
- Institute of Medical Physics and Biophysics, Medical Faculty, Leipzig University, Härtelstr. 16-18, 04107 Leipzig, Germany
| |
Collapse
|
13
|
Kim JY, Hong N, Park S, Ham SW, Kim EJ, Kim SO, Jang J, Kim Y, Kim JK, Kim SC, Park JW, Kim H. Jagged1 intracellular domain/SMAD3 complex transcriptionally regulates TWIST1 to drive glioma invasion. Cell Death Dis 2023; 14:822. [PMID: 38092725 PMCID: PMC10719344 DOI: 10.1038/s41419-023-06356-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 11/25/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023]
Abstract
Jagged1 (JAG1) is a Notch ligand that correlates with tumor progression. Not limited to its function as a ligand, JAG1 can be cleaved, and its intracellular domain translocates to the nucleus, where it functions as a transcriptional cofactor. Previously, we showed that JAG1 intracellular domain (JICD1) forms a protein complex with DDX17/SMAD3/TGIF2. However, the molecular mechanisms underlying JICD1-mediated tumor aggressiveness remains unclear. Here, we demonstrate that JICD1 enhances the invasive phenotypes of glioblastoma cells by transcriptionally activating epithelial-to-mesenchymal transition (EMT)-related genes, especially TWIST1. The inhibition of TWIST1 reduced JICD1-driven tumor aggressiveness. Although SMAD3 is an important component of transforming growth factor (TGF)-β signaling, the JICD1/SMAD3 transcriptional complex was shown to govern brain tumor invasion independent of TGF-β signaling. Moreover, JICD1-TWIST1-MMP2 and MMP9 axes were significantly correlated with clinical outcome of glioblastoma patients. Collectively, we identified the JICD1/SMAD3-TWIST1 axis as a novel inducer of invasive phenotypes in cancer cells.
Collapse
Affiliation(s)
- Jung Yun Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Nayoung Hong
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Sehyeon Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Seok Won Ham
- MEDIFIC Inc., Hwaseong-si, Gyeonggi-do, 18469, Republic of Korea
| | - Eun-Jung Kim
- MEDIFIC Inc., Hwaseong-si, Gyeonggi-do, 18469, Republic of Korea
| | - Sung-Ok Kim
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Junseok Jang
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Yoonji Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Jun-Kyum Kim
- MEDIFIC Inc., Hwaseong-si, Gyeonggi-do, 18469, Republic of Korea
| | - Sung-Chan Kim
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Jong-Whi Park
- Department of Life Sciences, Gachon University, Incheon, 21999, Republic of Korea.
| | - Hyunggee Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
14
|
Sato J, Nakano K, Miyazaki H. Decreased intracellular chloride enhances cell migration and invasion via activation of the ERK1/2 signaling pathway in DU145 human prostate carcinoma cells. Biochem Biophys Res Commun 2023; 685:149170. [PMID: 37924777 DOI: 10.1016/j.bbrc.2023.149170] [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: 06/29/2023] [Revised: 10/16/2023] [Accepted: 10/26/2023] [Indexed: 11/06/2023]
Abstract
Our previous study revealed that changes of the intracellular Cl- concentration ([Cl-]i) affected cell proliferation in cancer cells. However, the role of Cl- on cell migration and invasion in cancer cells remains unanalyzed. Therefore, the aim of the present study is to investigate whether changes of [Cl-]i affects cell migration and invasion of cancer cells. In human prostate cancer DU145 cells, cell migration and invasion were enhanced by culturing in the low Cl- medium (replacement of Cl- by NO3-). We also found that DU145 cells in the low Cl- condition caused significant transient ERK1/2 activation followed by an increase of MMP-1 mRNA levels. Inhibition of ERK1/2 activation in the low Cl- condition reduced enhancement of MMP-1 mRNA levels and decreased cell migration and invasion. These observations indicate that [Cl-]i plays important roles in metastatic function by regulating the ERK1/2 signaling pathway in human prostate cancer cells, and intracellular Cl- would be one of the key targets for anti-cancer therapy.
Collapse
Affiliation(s)
- Junichi Sato
- Department of Life Science, Faculty of Science and Engineering, Setsunan University, Neyagawa, Osaka, Japan
| | - Koya Nakano
- Department of Life Science, Faculty of Science and Engineering, Setsunan University, Neyagawa, Osaka, Japan
| | - Hiroaki Miyazaki
- Department of Life Science, Faculty of Science and Engineering, Setsunan University, Neyagawa, Osaka, Japan.
| |
Collapse
|
15
|
Wu Y, Liang H, Luo A, Li Y, Liu Z, Li X, Li W, Liang K, Li J, Liu Z, Du Y. Gelatin-based 3D biomimetic scaffolds platform potentiates culture of cancer stem cells in esophageal squamous cell carcinoma. Biomaterials 2023; 302:122323. [PMID: 37717405 DOI: 10.1016/j.biomaterials.2023.122323] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/02/2023] [Accepted: 09/09/2023] [Indexed: 09/19/2023]
Abstract
Cancer stem cells (CSCs) are crucial for tumorigenesis, metastasis, and therapy resistance in esophageal squamous cell carcinoma (ESCC). To further elucidate the mechanism underlying characteristics of CSCs and develop CSCs-targeted therapy, an efficient culture system that could expand and maintain CSCs is needed. CSCs reside in a complex tumor microenvironment, and three-dimensional (3D) culture systems of biomimetic scaffolds are expected to better support the growth of CSCs by recapitulating the biophysical properties of the extracellular matrix (ECM). Here, we established gelatin-based 3D biomimetic scaffolds mimicking the stiffness and collagen content of ESCC, which could enrich ESCC CSCs efficiently. Biological changes of ESCC cells laden in scaffolds with three different viscoelasticity emulating physiological stiffness of esophageal tissues were thoroughly investigated in varied aspects such as cell morphology, viability, cell phenotype markers, and transcriptomic profiling. The results demonstrated the priming effects of viscoelasticity on the stemness of ESCC. The highly viscous scaffolds (G': 6-403 Pa; G'': 2-75 Pa) better supported the enrichment of ESCC CSCs, and the TGF-beta signaling pathway might be involved in regulating the stemness of ESCC cells. Compared to two-dimensional (2D) cultures, highly viscous scaffolds significantly promoted the clonal expansion of ESCC cells in vitro and tumor formation ability in vivo. Our findings highlight the crucial role of biomaterials' viscoelasticity for the 3D culture of ESCC CSCs in vitro, and this newly-established culture system represents a valuable platform to support their growth, which could facilitate the CSCs-targeted therapy in the future.
Collapse
Affiliation(s)
- Yenan Wu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Haiwei Liang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Aiping Luo
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yong Li
- 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, China
| | - Zhiqiang Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xin Li
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wenxin Li
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Kaini Liang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Junyang Li
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
16
|
Cai X, Li M, Zhong Y, Yang W, Liang Z. COMP Improves Ang-II-Induced Atrial Fibrillation via TGF-β Signaling Pathway. Cardiovasc Toxicol 2023; 23:305-316. [PMID: 37584842 DOI: 10.1007/s12012-023-09799-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 06/27/2023] [Indexed: 08/17/2023]
Abstract
Cartilage oligomeric matrix protein (COMP) regulates transforming growth factor-β (TGF-β) signaling pathway, which has been proved to be associated with skin fibrosis and pulmonary fibrosis. Atrial fibrosis is a major factor of atrial fibrillation (AF). Nevertheless, the interaction between COMP and TGF-β as well as their role in AF remains undefined. The purpose of this study is to clarify the role of COMP in AF and explore its potential mechanism. The hub gene of AF was identified from two datasets using bioinformatics. Furthermore, it was verified by the downregulation of COMP in angiotensin-II (Ang-II)-induced AF in mice. Moreover, the effect on AF was examined using CCK8 assay, ELISA, and western blot. The involvement of TGF-β pathway was further discussed. The expression of COMP was the most significant among all these hub genes. Our experimental results revealed that the protein levels of TGF-β1, phosphorylated Smad2 (P-Smad2), and phosphorylated Smad3 (P-Smad3) were decreased after silencing COMP, which indicated that COMP knockdown could inhibit the activation of TGF-β pathway in AF cells. However, the phenomenon was reversed when the activator SRI was added. COMP acts as a major factor and can improve Ang-II-induced AF via TGF-β signaling pathway. Thus, our research enriches the understanding of the interaction between COMP and TGF-β in AF, and provides reference for the pathogenesis and diagnosis of AF.
Collapse
Affiliation(s)
- XiaoBi Cai
- Department of Cardiovascular Surgery, The Affiliated Hospital of Guangdong Medical University, No. 57, Renmin Avenue South, Xiashan District, Zhangjian City, 524001, Guangdong Province, China
| | - Mingliang Li
- Department of Cardiovascular Surgery, The Affiliated Hospital of Guangdong Medical University, No. 57, Renmin Avenue South, Xiashan District, Zhangjian City, 524001, Guangdong Province, China
| | - Ying Zhong
- Department of Cardiovascular Surgery, The Affiliated Hospital of Guangdong Medical University, No. 57, Renmin Avenue South, Xiashan District, Zhangjian City, 524001, Guangdong Province, China
| | - Wenkun Yang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Guangdong Medical University, No. 57, Renmin Avenue South, Xiashan District, Zhangjian City, 524001, Guangdong Province, China
| | - Zhu Liang
- Department of Cardiovascular and Thoracic Surgery, The Affiliated Hospital of Guangdong Medical University, No. 57, Renmin Avenue South, Xiashan District, Zhangjian City, 524001, Guangdong Province, China.
| |
Collapse
|
17
|
Rana M, Kansal RG, Bisunke B, Fang J, Shibata D, Bajwa A, Yang J, Glazer ES. Bromo- and Extra-Terminal Domain Inhibitors Induce Mitochondrial Stress in Pancreatic Ductal Adenocarcinoma. Mol Cancer Ther 2023; 22:936-946. [PMID: 37294884 PMCID: PMC10527726 DOI: 10.1158/1535-7163.mct-23-0149] [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: 03/09/2023] [Revised: 04/28/2023] [Accepted: 06/06/2023] [Indexed: 06/11/2023]
Abstract
Identifying novel, unique, and personalized molecular targets for patients with pancreatic ductal adenocarcinoma (PDAC) remains the greatest challenge in altering the biology of fatal tumors. Bromo- and extra-terminal domain (BET) proteins are activated in a noncanonical fashion by TGFβ, a ubiquitous cytokine in the PDAC tumor microenvironment (TME). We hypothesized that BET inhibitors (BETi) represent a new class of drugs that attack PDAC tumors via a novel mechanism. Using a combination of patient and syngeneic murine models, we investigated the effects of the BETi drug BMS-986158 on cellular proliferation, organoid growth, cell-cycle progression, and mitochondrial metabolic disruption. These were investigated independently and in combination with standard cytotoxic chemotherapy (gemcitabine + paclitaxel [GemPTX]). BMS-986158 reduced cell viability and proliferation across multiple PDAC cell lines in a dose-dependent manner, even more so in combination with cytotoxic chemotherapy (P < 0.0001). We found that BMS-986158 reduced both human and murine PDAC organoid growth (P < 0.001), with associated perturbations in the cell cycle leading to cell-cycle arrest. BMS-986158 disrupts normal cancer-dependent mitochondrial function, leading to aberrant mitochondrial metabolism and stress via dysfunctional cellular respiration, proton leakage, and ATP production. We demonstrated mechanistic and functional data that BETi induces metabolic mitochondrial dysfunction, abrogating PDAC progression and proliferation, alone and in combination with systemic cytotoxic chemotherapies. This novel approach improves the therapeutic window in patients with PDAC and offers another treatment approach distinct from cytotoxic chemotherapy that targets cancer cell bioenergetics.
Collapse
Affiliation(s)
- Manjul Rana
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Rita G. Kansal
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Bijay Bisunke
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Jie Fang
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, TN
| | - David Shibata
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
- Center for Cancer Research, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Amandeep Bajwa
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
- Transplant Research Institute, James D. Eason Transplant Institute, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
- Department of Genetics, Genomics, and Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Jun Yang
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, TN
- Transplant Research Institute, James D. Eason Transplant Institute, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
- Department of Genetics, Genomics, and Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
- Comprehensive Cancer Center, St. Jude Children’s Research Hospital, Memphis, TN
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, St. Jude Children’s Research Hospital, Memphis, TN
| | - Evan S. Glazer
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
- Center for Cancer Research, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
| |
Collapse
|
18
|
Matsuguchi S, Hirai Y. Syntaxin4, P-cadherin, and CCAAT enhancer binding protein β as signaling elements in the novel differentiation pathway for cultured embryonic stem cells. Biochem Biophys Res Commun 2023; 672:27-35. [PMID: 37331168 DOI: 10.1016/j.bbrc.2023.06.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/15/2023] [Accepted: 06/13/2023] [Indexed: 06/20/2023]
Abstract
Pluripotent stem cells possess the potential to differentiate into all three germ layers. However, upon removal of the stemness factors, pluripotent stem cells, such as embryonic stem cells (ESCs), exhibit EMT-like cell behavior and lose stemness signatures. This process involves the membrane translocation of the t-SNARE protein syntaxin4 (Stx4) and the expression of the intercellular adhesion molecule P-cadherin. The forced expression of either of these elements induces the emergence of such phenotypes even in the presence of stemness factors. Interestingly, extracellular Stx4, but not P-cadherin, appears to induce a significant upregulation of the gastrulation-related gene brachyury, along with a slight upregulation of the smooth muscle cell-related gene ACTA2 in ESCs. Furthermore, our findings reveal that extracellular Stx4 plays a role in preventing the elimination of CCAAT enhancer binding protein β (C/EBPβ). Notably, the forced overexpression of C/EBPβ led to the downregulation of brachyury and a significant upregulation of ACTA2 in ESCs. These observations suggest that extracellular Stx4 contributes to early mesoderm induction while simultaneously activating an element that alters the differentiation state. The fact that a single differentiation cue can elicit multiple differentiation responses may reflect the challenges associated with achieving sensitive and directed differentiation in cultured stem cells.
Collapse
Affiliation(s)
- Shuji Matsuguchi
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen-Uegahara, Sanda, 669-1330, Japan.
| | - Yohei Hirai
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen-Uegahara, Sanda, 669-1330, Japan.
| |
Collapse
|
19
|
Cao Y. Neural induction drives body axis formation during embryogenesis, but a neural induction-like process drives tumorigenesis in postnatal animals. Front Cell Dev Biol 2023; 11:1092667. [PMID: 37228646 PMCID: PMC10203556 DOI: 10.3389/fcell.2023.1092667] [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: 11/08/2022] [Accepted: 04/17/2023] [Indexed: 05/27/2023] Open
Abstract
Characterization of cancer cells and neural stem cells indicates that tumorigenicity and pluripotency are coupled cell properties determined by neural stemness, and tumorigenesis represents a process of progressive loss of original cell identity and gain of neural stemness. This reminds of a most fundamental process required for the development of the nervous system and body axis during embryogenesis, i.e., embryonic neural induction. Neural induction is that, in response to extracellular signals that are secreted by the Spemann-Mangold organizer in amphibians or the node in mammals and inhibit epidermal fate in ectoderm, the ectodermal cells lose their epidermal fate and assume the neural default fate and consequently, turn into neuroectodermal cells. They further differentiate into the nervous system and also some non-neural cells via interaction with adjacent tissues. Failure in neural induction leads to failure of embryogenesis, and ectopic neural induction due to ectopic organizer or node activity or activation of embryonic neural genes causes a formation of secondary body axis or a conjoined twin. During tumorigenesis, cells progressively lose their original cell identity and gain of neural stemness, and consequently, gain of tumorigenicity and pluripotency, due to various intra-/extracellular insults in cells of a postnatal animal. Tumorigenic cells can be induced to differentiation into normal cells and integrate into normal embryonic development within an embryo. However, they form tumors and cannot integrate into animal tissues/organs in a postnatal animal because of lack of embryonic inducing signals. Combination of studies of developmental and cancer biology indicates that neural induction drives embryogenesis in gastrulating embryos but a similar process drives tumorigenesis in a postnatal animal. Tumorigenicity is by nature the manifestation of aberrant occurrence of pluripotent state in a postnatal animal. Pluripotency and tumorigenicity are both but different manifestations of neural stemness in pre- and postnatal stages of animal life, respectively. Based on these findings, I discuss about some confusion in cancer research, propose to distinguish the causality and associations and discriminate causal and supporting factors involved in tumorigenesis, and suggest revisiting the focus of cancer research.
Collapse
Affiliation(s)
- Ying Cao
- Shenzhen Research Institute of Nanjing University, Shenzhen, China
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
20
|
Tu SM, Aydin AM, Maraboyina S, Chen Z, Singh S, Gokden N, Langford T. Stem Cell Origin of Cancer: Implications of Oncogenesis Recapitulating Embryogenesis in Cancer Care. Cancers (Basel) 2023; 15:cancers15092516. [PMID: 37173982 PMCID: PMC10177345 DOI: 10.3390/cancers15092516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/18/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
From this perspective, we wonder about the clinical implications of oncology recapturing ontogeny in the contexts of neoantigens, tumor biomarkers, and cancer targets. We ponder about the biological ramifications of finding remnants of mini-organs and residuals of tiny embryos in some tumors. We reminisce about classical experiments showing that the embryonic microenvironment possesses antitumorigenic properties. Ironically, a stem-ness niche-in the wrong place at the wrong time-is also an onco-niche. We marvel at the paradox of TGF-beta both as a tumor suppressor and a tumor promoter. We query about the dualism of EMT as a stem-ness trait engaged in both normal development and abnormal disease states, including various cancers. It is uncanny that during fetal development, proto-oncogenes wax, while tumor-suppressor genes wane. Similarly, during cancer development, proto-oncogenes awaken, while tumor-suppressor genes slumber. Importantly, targeting stem-like pathways has therapeutic implications because stem-ness may be the true driver, if not engine, of the malignant process. Furthermore, anti-stem-like activity elicits anti-cancer effects for a variety of cancers because stem-ness features may be a universal property of cancer. When a fetus survives and thrives despite immune surveillance and all the restraints of nature and the constraints of its niche, it is a perfect baby. Similarly, when a neoplasm survives and thrives in an otherwise healthy and immune-competent host, is it a perfect tumor? Therefore, a pertinent narrative of cancer depends on a proper perspective of cancer. If malignant cells are derived from stem cells, and both cells are intrinsically RB1 negative and TP53 null, do the absence of RB1 and loss of TP53 really matter in this whole narrative and an entirely different perspective of cancer?
Collapse
Affiliation(s)
- Shi-Ming Tu
- Division of Hematology and Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Ahmet Murat Aydin
- Department of Urology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Sanjay Maraboyina
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Zhongning Chen
- Division of Hematology and Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Sunny Singh
- Division of Hematology and Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Neriman Gokden
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Timothy Langford
- Department of Urology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
21
|
Zhan T, Cheng X, Zhu Q, Han Z, Zhu K, Tan J, Liu M, Chen W, Chen X, Chen X, Tian X, Huang X. LncRNA LOC105369504 inhibits tumor proliferation and metastasis in colorectal cancer by regulating PSPC1. Cell Death Discov 2023; 9:89. [PMID: 36894530 PMCID: PMC9998613 DOI: 10.1038/s41420-023-01384-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 02/21/2023] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
There is growing evidence that long non-coding RNAs (lncRNAs) are significant contributors to the epigenetic mechanisms implicated in the emergence, progression and metastasis of the colorectal cancer (CRC), but many remain underexplored. A novel lncRNA LOC105369504, was identified to be a potential functional lncRNA by microarray analysis. In CRC, the expression of LOC105369504 was markedly decreased and resulted in distinct variations in proliferation, invasion, migration and epithelial-mesenchymal transition (EMT) in vivo and in vitro. This study showed that LOC105369504 bound to the protein of paraspeckles compound 1 (PSPC1) directly and regulated its stability using the ubiquitin-proteasome pathway in CRC cells. The suppression of CRC by LOC105369504 could be reversed through PSPC1 overexpression.This study showed that in CRC, LOC105369504 was under-regulated and as a novel lncRNA, LOC105369504 exerted tumor suppressive activity to suppress the proliferation together with metastasis in CRC cells through the regulation of PSPC1. These results offer new perspectives on the lncRNA effect on the progression of CRC.
Collapse
Affiliation(s)
- Ting Zhan
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China
| | - Xueting Cheng
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China
| | - Qingxi Zhu
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China
| | - Zheng Han
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China
| | - Kejing Zhu
- Department of Pharmacy, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China
| | - Jie Tan
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China
| | - Men Liu
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China
| | - Wei Chen
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China
| | - Xiaoli Chen
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China
| | - Xiaohong Chen
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China
| | - Xia Tian
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China.
| | - Xiaodong Huang
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China.
| |
Collapse
|
22
|
Zhang XW, Wu YS, Xu TM, Cui MH. CAR-T Cells in the Treatment of Ovarian Cancer: A Promising Cell Therapy. Biomolecules 2023; 13:biom13030465. [PMID: 36979400 PMCID: PMC10046142 DOI: 10.3390/biom13030465] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/23/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Ovarian cancer (OC) is among the most common gynecologic malignancies with a poor prognosis and a high mortality rate. Most patients are diagnosed at an advanced stage (stage III or IV), with 5-year survival rates ranging from 25% to 47% worldwide. Surgical resection and first-line chemotherapy are the main treatment modalities for OC. However, patients usually relapse within a few years of initial treatment due to resistance to chemotherapy. Cell-based therapies, particularly adoptive T-cell therapy and chimeric antigen receptor T (CAR-T) cell therapy, represent an alternative immunotherapy approach with great potential for hematologic malignancies. However, the use of CAR-T-cell therapy for the treatment of OC is still associated with several difficulties. In this review, we comprehensively discuss recent innovations in CAR-T-cell engineering to improve clinical efficacy, as well as strategies to overcome the limitations of CAR-T-cell therapy in OC.
Collapse
|
23
|
C/EBPβ expression decreases in cervical cancer and leads to tumorigenesis. BMC Cancer 2023; 23:79. [PMID: 36694148 PMCID: PMC9872280 DOI: 10.1186/s12885-023-10543-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 01/12/2023] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Cervical cancer is currently estimated to be the fourth most common cancer among women worldwide and the leading cause of cancer-related deaths in some of the world's poorest countries. C/EBPβ has tumor suppressor effects because it is necessary for oncogene-induced senescence. However, C/EBPβ also has an oncogenic role. The specific role of C/EBPβ in cervical cancer as a tumor suppressor or oncoprotein is unclear. OBJECTIVE To explore the role of the C/EBPβ protein in cervical tumorigenesis and progression. METHODS Quantitative RT-PCR was used to analyze C/EBPβ (15 cervical cancer tissue samples and 15 corresponding normal cervical tissue samples), miR-661, and MTA1 mRNA expression in clinical samples (10 cervical cancer tissue samples and 10 corresponding normal cervical tissue samples). Immunohistochemistry was used to analyze C/EBPβ (381 clinical samples), Ki67 (80 clinical samples) and PCNA ( 60 clinical samples) protein expression. MALDI-TOF MassARRAY was used to analyze C/EBPβ gene methylation (13 cervical cancer tissues and 13 corresponding normal cervical tissues). Cell proliferation was analyzed by CCK-8 in cervical cancer cell lines. Western blotting and immunohistochemistry were performed to detect C/EBPβ protein expression levels, and mRNA expression was analyzed by quantitative RT-PCR analysis. Flow cytometry was performed to measure cell cycle distribution and cell apoptosis. Colony formation, Transwell, cell invasion, and wound healing assays were performed to detect cell migration and invasion. RESULTS C/EBPβ protein expression was significantly reduced in cervical cancer tissues compared with cervicitis tissues (P < 0.01). Ki67 protein and PCNA protein expression levels were significantly higher in cervical cancer tissues compared with cervicitis tissues. The rate of C/EBPβ gene promoter methylation of CpG12, 13, 14 and CpG19 in cervical cancer tissues was significantly increased compared with normal cervical tissue (P < 0.05). In addition, C/EBPβ was overexpressed in cervical cancer cells and this overexpression inhibited cell proliferation, migration, invasion, arrested cells in S phase, and promoted apoptosis. CONCLUSIONS We have demonstrated that C/EBPβ decreased in cervical cancer tissues and overexpression of the C/EBPβ gene in cervical cancer cells could inhibit proliferation, invasion and migration.
Collapse
|
24
|
LRRC superfamily expression in stromal cells predicts the clinical prognosis and platinum resistance of ovarian cancer. BMC Med Genomics 2023; 16:10. [PMID: 36653841 PMCID: PMC9850808 DOI: 10.1186/s12920-023-01435-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Leucine-rich repeat sequence domains are known to mediate protein‒protein interactions. Recently, some studies showed that members of the leucine rich repeat containing (LRRC) protein superfamily may become new targets for the diagnosis and treatment of tumours. However, it is not known whether any of the LRRC superfamily genes is expressed in the stroma of ovarian cancer (OC) and is associated with prognosis. METHODS The clinical data and transcriptional profiles of OC patients from the public databases TCGA (n = 427), GTEx (n = 88) and GEO (GSE40266 and GSE40595) were analysed by R software. A nomogram model was also generated through R. An online public database was used for auxiliary analysis of prognosis, immune infiltration and protein‒protein interaction (PPI) networks. Immunohistochemistry and qPCR were performed to determine the protein and mRNA levels of genes in high-grade serous ovarian cancer (HGSC) tissues of participants and the MRC-5 cell line induced by TGF-β. RESULTS LRRC15 and LRRC32 were identified as differentially expressed genes from the LRRC superfamily by GEO transcriptome analysis. PPI network analysis suggested that they were most enriched in TGF-β signalling. The TCGA-GTEx analysis results showed that only LRRC15 was highly expressed in both cancer-associated fibroblasts (CAFs) and the tumour stroma of OC and was related to clinical prognosis. Based on this, we developed a nomogram model to predict the incidence of adverse outcomes in OC. Moreover, LRRC15 was positively correlated with CAF infiltration and negatively correlated with CD8 + T-cell infiltration. As a single indicator, LRRC15 had the highest accuracy (AUC = 0.920) in predicting the outcome of primary platinum resistance. CONCLUSIONS The LRRC superfamily is related to the TGF-β pathway in the microenvironment of OC. LRRC15, as a stromal biomarker, can predict the clinical prognosis of HGSC and promote the immunosuppressive microenvironment. LRRC15 may be a potential therapeutic target for reversing primary resistance in OC.
Collapse
|
25
|
Villar VH, Subotički T, Đikić D, Mitrović-Ajtić O, Simon F, Santibanez JF. Transforming Growth Factor-β1 in Cancer Immunology: Opportunities for Immunotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1408:309-328. [PMID: 37093435 DOI: 10.1007/978-3-031-26163-3_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Transforming growth factor-beta1 (TGF-β) regulates a plethora of cell-intrinsic processes that modulate tumor progression in a context-dependent manner. Thus, although TGF-β acts as a tumor suppressor in the early stages of tumorigenesis, in late stages, this factor promotes tumor progression and metastasis. In addition, TGF-β also impinges on the tumor microenvironment by modulating the immune system. In this aspect, TGF-β exhibits a potent immunosuppressive effect, which allows both cancer cells to escape from immune surveillance and confers resistance to immunotherapy. While TGF-β inhibits the activation and antitumoral functions of T-cell lymphocytes, dendritic cells, and natural killer cells, it promotes the generation of T-regulatory cells and myeloid-derived suppressor cells, which hinder antitumoral T-cell activities. Moreover, TGF-β promotes tumor-associated macrophages and neutrophils polarization from M1 into M2 and N1 to N2, respectively. Altogether, these effects contribute to the generation of an immunosuppressive tumor microenvironment and support tumor promotion. This review aims to analyze the relevant evidence on the complex role of TGF-β in cancer immunology, the current outcomes of combined immunotherapies, and the anti-TGF-β therapies that may improve the success of current and new oncotherapies.
Collapse
Affiliation(s)
- Víctor H Villar
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Tijana Subotički
- Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Dragoslava Đikić
- Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Olivera Mitrović-Ajtić
- Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Felipe Simon
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute On Immunology and Immunotherapy, Santiago, Chile
- Millennium Nucleus of Ion Channel-Associated Diseases, Santiago, Chile
| | - Juan F Santibanez
- Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia.
- Integrative Center for Biology and Applied Chemistry (CIBQA), Bernardo O'Higgins University, Santiago, Chile.
- Molecular Oncology Group, Institute for Medical Research, University of Belgrade, Dr. Subotica 4, POB 102, 11129, Belgrade, Serbia.
| |
Collapse
|
26
|
Ding JT, Zhou HN, Huang YF, Peng J, Huang HY, Yi H, Zong Z, Ning ZK. TGF-β Pathways Stratify Colorectal Cancer into Two Subtypes with Distinct Cartilage Oligomeric Matrix Protein (COMP) Expression-Related Characteristics. Biomolecules 2022; 12:1877. [PMID: 36551305 PMCID: PMC9775768 DOI: 10.3390/biom12121877] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/11/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Colorectal cancers (CRCs) continue to be the leading cause of cancer-related deaths worldwide. The exact landscape of the molecular features of TGF-β pathway-inducing CRCs remains uncharacterized. METHODS Unsupervised hierarchical clustering was performed to stratify samples into two clusters based on the differences in TGF-β pathways. Weighted gene co-expression network analysis was applied to identify the key gene modules mediating the different characteristics between two subtypes. An algorithm integrating the least absolute shrinkage and selection operator (LASSO), XGBoost, and random forest regression was performed to narrow down the candidate genes. Further bioinformatic analyses were performed focusing on COMP-related immune infiltration and functions. RESULTS The integrated machine learning algorithm identified COMP as the hub gene, which exhibited a significant predictive value for two subtypes with an area under the curve (AUC) value equaling 0.91. Further bioinformatic analysis revealed that COMP was significantly upregulated in various cancers, especially in advanced CRCs, and regulated the immune infiltration, especially M2 macrophages and cancer-associated fibroblasts in CRCs. CONCLUSIONS Comprehensive immune analysis and experimental validation demonstrate that COMP is a reliable signature for subtype prediction. Our results could provide a new point for TGFβ-targeted anticancer drugs and contribute to guiding clinical decision making for CRC patients.
Collapse
Affiliation(s)
- Jia-Tong Ding
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medicine School, Nanchang University, Nanchang 330006, China
| | - Hao-Nan Zhou
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- Queen Mary School, Nanchang University, Nanchang 330006, China
| | - Ying-Feng Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jie Peng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medicine School, Nanchang University, Nanchang 330006, China
| | - Hao-Yu Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Hao Yi
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Zhen Zong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Zhi-Kun Ning
- Department of Day Ward, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
27
|
Toledo B, Picon-Ruiz M, Marchal JA, Perán M. Dual Role of Fibroblasts Educated by Tumour in Cancer Behavior and Therapeutic Perspectives. Int J Mol Sci 2022; 23:15576. [PMID: 36555218 PMCID: PMC9778751 DOI: 10.3390/ijms232415576] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/25/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022] Open
Abstract
Tumours are complex systems with dynamic interactions between tumour cells, non-tumour cells, and extracellular components that comprise the tumour microenvironment (TME). The majority of TME's cells are cancer-associated fibroblasts (CAFs), which are crucial in extracellular matrix (ECM) construction, tumour metabolism, immunology, adaptive chemoresistance, and tumour cell motility. CAF subtypes have been identified based on the expression of protein markers. CAFs may act as promoters or suppressors in tumour cells depending on a variety of factors, including cancer stage. Indeed, CAFs have been shown to promote tumour growth, survival and spread, and secretome changes, but they can also slow tumourigenesis at an early stage through mechanisms that are still poorly understood. Stromal-cancer interactions are governed by a variety of soluble factors that determine the outcome of the tumourigenic process. Cancer cells release factors that enhance the ability of fibroblasts to secrete multiple tumour-promoting chemokines, acting on malignant cells to promote proliferation, migration, and invasion. This crosstalk between CAFs and tumour cells has given new prominence to the stromal cells, from being considered as mere physical support to becoming key players in the tumour process. Here, we focus on the concept of cancer as a non-healing wound and the relevance of chronic inflammation to tumour initiation. In addition, we review CAFs heterogeneous origins and markers together with the potential therapeutic implications of CAFs "re-education" and/or targeting tumour progression inhibition.
Collapse
Affiliation(s)
- Belén Toledo
- Department of Health Sciences, University of Jaén, E-23071 Jaén, Spain
| | - Manuel Picon-Ruiz
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, E-18100 Granada, Spain
- Instituto de Investigación Sanitaria ibs. GRANADA, Hospitales Universitarios de Granada-Universidad de Granada, E-18071 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, E-18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, E-18016 Granada, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, E-18100 Granada, Spain
- Instituto de Investigación Sanitaria ibs. GRANADA, Hospitales Universitarios de Granada-Universidad de Granada, E-18071 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, E-18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, E-18016 Granada, Spain
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, E-23071 Jaén, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, E-18100 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, E-18016 Granada, Spain
| |
Collapse
|
28
|
Saito M. Novel Roles of Nanog in Cancer Cells and Their Extracellular Vesicles. Cells 2022; 11:cells11233881. [PMID: 36497144 PMCID: PMC9736053 DOI: 10.3390/cells11233881] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/22/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
The use of extracellular vesicle (EV)-based vaccines is a strategically promising way to prevent cancer metastasis. The effective roles of immune cell-derived EVs have been well understood in the literature. In the present paper, we focus on cancer cell-derived EVs to enforce, more thoroughly, the use of EV-based vaccines against unexpected malignant cells that might appear in poor prognostic patients. As a model of such a cancer cell with high malignancy, Nanog-overexpressing melanoma cell lines were developed. As expected, Nanog overexpression enhanced the metastatic potential of melanomas. Against our expectations, a fantastic finding was obtained that determined that EVs derived from Nanog-overexpressing melanomas exhibited a metastasis-suppressive effect. This is considered to be a novel role for Nanog in regulating the property of cancer cell-derived EVs. Stimulated by this result, the review of Nanog's roles in various cancer cells and their EVs has been updated once again. Although there was no other case presenting a similar contribution by Nanog, only one case suggested that NANOG and SOX might be better prognosis markers in head and neck squamous cell carcinomas. This review clarifies the varieties of Nanog-dependent phenomena and the relevant signaling factors. The information summarized in this study is, thus, suggestive enough to generate novel ideas for the construction of an EV-based versatile vaccine platform against cancer metastasis.
Collapse
Affiliation(s)
- Mikako Saito
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan
| |
Collapse
|
29
|
Huang T, Bao H, Meng YH, Zhu JL, Chu XD, Chu XL, Pan JH. Tumour budding is a novel marker in breast cancer: the clinical application and future prospects. Ann Med 2022; 54:1303-1312. [PMID: 35535687 PMCID: PMC9103277 DOI: 10.1080/07853890.2022.2070272] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Breast cancer (BC) is a group of markedly heterogeneous tumours. There are many subtypes with different biological behaviours and clinicopathological characteristics, leading to significantly different prognosis. Despite significant advances in the treatment of BC, early metastatic is a critical factor for poor prognosis in BC patients. Tumour budding (TB) is considered as the first step process of tumour metastasis and is related to the epithelial-mesenchymal transition (EMT). TB has been observed in a variety of cancers, such as colorectal and gastric cancer, and had been considered as a distinct clinicopathological characteristics for early metastasis. However, TB evaluation standards and clinical application are not uniform in BC, as well as its molecular mechanism is not fully understood. Here, we reviewed the interpretation criteria, mechanism, clinicopathological characteristics and clinical application prospects of TB in BC. Key messagesCurrently, tumour budding is a poor prognosis for various solid tumours, also in breast cancer.Tumour budding is based on epithelial-mesenchymal transition and tumour microenvironment factors and is presumed to be an early step in the metastatic process.Breast cancer tumour budding still needs multi-centre experiments. We summarize the current research on breast cancer tumour budding, analyse the method of discriminating breast cancer tumour budding and explore the prognostic role and mechanism in breast cancer.
Collapse
Affiliation(s)
- Ting Huang
- Department of Clinical Pathology, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Hui Bao
- Department of Plastic Surgery, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yu-Hua Meng
- Department of Clinical Pathology, the First People's Hospital of Shunde, Foshan, China
| | - Jian-Lin Zhu
- Department of General Surgery, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiao-Dong Chu
- Department of General Surgery, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiao-Li Chu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong, China.,Guangdong Provincial Key laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, Guangzhou, China
| | - Jing-Hua Pan
- Department of General Surgery, the First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
30
|
Cherkaoui S, Durot S, Bradley J, Critchlow S, Dubuis S, Masiero MM, Wegmann R, Snijder B, Othman A, Bendtsen C, Zamboni N. A functional analysis of 180 cancer cell lines reveals conserved intrinsic metabolic programs. Mol Syst Biol 2022; 18:e11033. [PMID: 36321552 PMCID: PMC9627673 DOI: 10.15252/msb.202211033] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 10/03/2022] [Accepted: 10/07/2022] [Indexed: 11/30/2022] Open
Abstract
Cancer cells reprogram their metabolism to support growth and invasion. While previous work has highlighted how single altered reactions and pathways can drive tumorigenesis, it remains unclear how individual changes propagate at the network level and eventually determine global metabolic activity. To characterize the metabolic lifestyle of cancer cells across pathways and genotypes, we profiled the intracellular metabolome of 180 pan-cancer cell lines grown in identical conditions. For each cell line, we estimated activity for 49 pathways spanning the entirety of the metabolic network. Upon clustering, we discovered a convergence into only two major metabolic types. These were functionally confirmed by 13 C-flux analysis, lipidomics, and analysis of sensitivity to perturbations. They revealed that the major differences in cancers are associated with lipid, TCA cycle, and carbohydrate metabolism. Thorough integration of these types with multiomics highlighted little association with genetic alterations but a strong association with markers of epithelial-mesenchymal transition. Our analysis indicates that in absence of variations imposed by the microenvironment, cancer cells adopt distinct metabolic programs which serve as vulnerabilities for therapy.
Collapse
Affiliation(s)
- Sarah Cherkaoui
- Institute of Molecular Systems BiologyETH ZürichZürichSwitzerland
- PhD Program in Systems BiologyLife Science ZürichZürichSwitzerland
| | - Stephan Durot
- Institute of Molecular Systems BiologyETH ZürichZürichSwitzerland
- PhD Program in Systems BiologyLife Science ZürichZürichSwitzerland
| | | | | | - Sebastien Dubuis
- Institute of Molecular Systems BiologyETH ZürichZürichSwitzerland
| | - Mauro Miguel Masiero
- Institute of Molecular Systems BiologyETH ZürichZürichSwitzerland
- PhD Program in Systems BiologyLife Science ZürichZürichSwitzerland
| | - Rebekka Wegmann
- Institute of Molecular Systems BiologyETH ZürichZürichSwitzerland
- PhD Program in Systems BiologyLife Science ZürichZürichSwitzerland
| | - Berend Snijder
- Institute of Molecular Systems BiologyETH ZürichZürichSwitzerland
| | - Alaa Othman
- Institute of Molecular Systems BiologyETH ZürichZürichSwitzerland
- PHRT Swiss Multi‐OMICS Center / smoc.ethz.chZürichSwitzerland
| | | | - Nicola Zamboni
- Institute of Molecular Systems BiologyETH ZürichZürichSwitzerland
- PHRT Swiss Multi‐OMICS Center / smoc.ethz.chZürichSwitzerland
| |
Collapse
|
31
|
Xie Q, Fan X, Han Y, Wu BX, Zhu B. Daphnoretin Arrests the Cell Cycle and Induces Apoptosis in Human Breast Cancer Cells. JOURNAL OF NATURAL PRODUCTS 2022; 85:2332-2339. [PMID: 36154031 DOI: 10.1021/acs.jnatprod.2c00504] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Emerging evidence has shown that daphnoretin, one of the main active ingredients of Daphne giraldii Nitsche, processes antitumor activities in several tumor cells (e.g., colon cancer, lung cancer, cervical cancer, and osteosarcoma). However, the antitumor effect and its mechanism in breast cancer are unexplored. In this study, our data indicated that daphnoretin obviously suppressed the proliferation of breast cancer MCF-7 and MDA-MB-231 cells. Further studies showed that daphnoretin remarkably increased the p21 level, decreased cyclin E and CDK2 levels, and then arrested the cell cycle at the S phase. Moreover, daphnoretin obviously lowered the BCL-2 level and raised the levels of BAX and cleaved caspase-9 and -3, leading to cell apoptosis. Furthermore, daphnoretin remarkably decreased the ratio of p-PI3K/PI3K and p-AKT/AKT in breast cancer cells. Collectively, these findings demonstrated that daphnoretin could suppress breast cancer cell proliferation through cell cycle arrest and inducing apoptosis, which is related to the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Qi Xie
- Jiangsu College of Nursing, 9 Science and Technology Avenue, Huaian, 223005, Jiangsu China
| | - Xinmei Fan
- Jiangsu Food & Pharmaceutical Science College, 4 Meicheng Road, Huaian, 223003, Jiangsu People's Republic of China
| | - Yonghong Han
- Jiangsu College of Nursing, 9 Science and Technology Avenue, Huaian, 223005, Jiangsu People's Republic of China
| | - Bao-Xiang Wu
- Key Laboratory of Coal Processing and Efficient Utilization, Ministry of Education, China University of Mining & Technology, Xuzhou221116, Jiangsu People's Republic of China
| | - Bao Zhu
- Jiangsu College of Nursing, 9 Science and Technology Avenue, Huaian, 223005, Jiangsu People's Republic of China
| |
Collapse
|
32
|
Shi X, Yang J, Deng S, Xu H, Wu D, Zeng Q, Wang S, Hu T, Wu F, Zhou H. TGF-β signaling in the tumor metabolic microenvironment and targeted therapies. J Hematol Oncol 2022; 15:135. [PMID: 36115986 PMCID: PMC9482317 DOI: 10.1186/s13045-022-01349-6] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/24/2022] [Indexed: 12/30/2022] Open
Abstract
AbstractTransforming growth factor-β (TGF-β) signaling has a paradoxical role in cancer progression, and it acts as a tumor suppressor in the early stages but a tumor promoter in the late stages of cancer. Once cancer cells are generated, TGF-β signaling is responsible for the orchestration of the immunosuppressive tumor microenvironment (TME) and supports cancer growth, invasion, metastasis, recurrence, and therapy resistance. These progressive behaviors are driven by an “engine” of the metabolic reprogramming in cancer. Recent studies have revealed that TGF-β signaling regulates cancer metabolic reprogramming and is a metabolic driver in the tumor metabolic microenvironment (TMME). Intriguingly, TGF-β ligands act as an “endocrine” cytokine and influence host metabolism. Therefore, having insight into the role of TGF-β signaling in the TMME is instrumental for acknowledging its wide range of effects and designing new cancer treatment strategies. Herein, we try to illustrate the concise definition of TMME based on the published literature. Then, we review the metabolic reprogramming in the TMME and elaborate on the contribution of TGF-β to metabolic rewiring at the cellular (intracellular), tissular (intercellular), and organismal (cancer-host) levels. Furthermore, we propose three potential applications of targeting TGF-β-dependent mechanism reprogramming, paving the way for TGF-β-related antitumor therapy from the perspective of metabolism.
Collapse
|
33
|
Hou CP, Tsui KH, Chen ST, Chang KS, Sung HC, Hsu SY, Lin YH, Feng TH, Juang HH. The Upregulation of Caffeic Acid Phenethyl Ester on Growth Differentiation Factor 15 Inhibits Transforming Growth Factor β/Smad Signaling in Bladder Carcinoma Cells. Biomedicines 2022; 10:biomedicines10071625. [PMID: 35884930 PMCID: PMC9312961 DOI: 10.3390/biomedicines10071625] [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] [Received: 06/14/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 11/16/2022] Open
Abstract
Growth differentiation factor 15 (GDF15) is known as a TGFβ-like cytokine acting on the TGFβ receptor to modulate target genes. GDF15 is regarded as a tumor suppressor gene in the human bladder and the caffeic acid phenethyl ester (CAPE) induces GDF15 expression to inhibit the tumor growth in vitro and in vivo. However, the interactions among GDF15, CAPE, and TGFβ/Smads signaling in the human bladder carcinoma cells remain unexplored. Results revealed that TGFβ downregulated the expression of GDF15 via the activation of Smad 2/3 and Smad 1/5. Induction of GDF15 on its downstream genes, NDRG1 and maspin, is dependent on the TGFβ/Smad pathways. Moreover, TGFβ blocked the CAPE-inducing expressions of GDF15, maspin, and NDRG1. Pretreatment of TGF receptor kinase inhibitor not only blocked the activation of TGFβ but also attenuated the activation of GDF15 on the expressions of maspin and NDRG1. The CAPE treatment attenuated the activation of TGFβ on cell proliferation and invasion. Our findings indicate that TGFβ downregulated the expressions of GDF15, maspin, and NDRG1 via TGFβ/Smad signaling. Whereas, CAPE acts as an antagonist on TGFβ/Smad signaling to block the effect of TGFβ on the GDF15 expression and cell proliferation and invasion in bladder carcinoma cells.
Collapse
Affiliation(s)
- Chen-Pang Hou
- Graduate Institute of Clinical Medical Science, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan;
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan;
- Department of Healthcare Management, Yuanpei University of Medical Technology, Hsinchu 300, Taiwan
| | - Ke-Hung Tsui
- Department of Urology, Shuang Ho Hospital, New Taipei City 235041, Taiwan;
- TMU Research Center of Urology and Kindey, Department of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Syue-Ting Chen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (S.-T.C.); (K.-S.C.); (H.-C.S.); (S.-Y.H.)
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Kang-Shuo Chang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (S.-T.C.); (K.-S.C.); (H.-C.S.); (S.-Y.H.)
| | - Hsin-Ching Sung
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (S.-T.C.); (K.-S.C.); (H.-C.S.); (S.-Y.H.)
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Shu-Yuan Hsu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (S.-T.C.); (K.-S.C.); (H.-C.S.); (S.-Y.H.)
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Yu-Hsiang Lin
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan;
| | - Tsui-Hsia Feng
- School of Nursing, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan;
| | - Horng-Heng Juang
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan;
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (S.-T.C.); (K.-S.C.); (H.-C.S.); (S.-Y.H.)
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Correspondence: ; Tel.: +886-3-2118800; Fax: +886-3-2118112
| |
Collapse
|
34
|
Irshad K, Srivastava C, Malik N, Arora M, Gupta Y, Goswami S, Sarkar C, Suri V, Mahajan S, Gupta DK, Suri A, Chattopadhyay P, Sinha S, Chosdol K. Upregulation of Atypical Cadherin FAT1 Promotes an Immunosuppressive Tumor Microenvironment via TGF-β. Front Immunol 2022; 13:813888. [PMID: 35720420 PMCID: PMC9205206 DOI: 10.3389/fimmu.2022.813888] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 04/11/2022] [Indexed: 12/15/2022] Open
Abstract
FAT atypical cadherin 1 (FAT1) promotes glioblastoma (GBM) by promoting protumorigenic inflammatory cytokine expression in tumor cells. However, tumors also have an immunosuppressive microenvironment maintained by mediators such as transforming growth factor (TGF)-β cytokines. Here, we have studied the role of FAT1 in tumor immune suppression. Our preliminary TIMER2.0 analysis of The Cancer Genome Atlas (TCGA) database revealed an inverse correlation of FAT1 expression with infiltration of tumor-inhibiting immune cells (such as monocytes and T cells) and a positive correlation with tumor-promoting immune cells [such as myeloid-derived suppressor cells (MDSCs)] in various cancers. We have analyzed the role of FAT1 in modulating the expression of TGF-β1/2 in resected human gliomas, primary glioma cultures, and other cancer cell lines (U87MG, HepG2, Panc-1, and HeLa). Positive correlations of gene expression of FAT1 and TGF-β1/2 were observed in various cancers in TCGA, Glioma Longitudinal Analysis Consortium (GLASS), and Chinese Glioma Genome Atlas (CGGA) databases. Positive expression correlations of FAT1 were also found with TGF-β1/2 and Serpine1 (downstream target) in fresh-frozen GBM samples using q-PCR. siRNA-mediated FAT1 knockdown in cancer cell lines and in primary cultures led to decreased TGF-β1/2 expression/secretion as assessed by q-PCR, Western blotting, and ELISA. There was increased chemotaxis (transmigration) of THP-1 monocytes toward siFAT1-transfected tumor cell supernatant as a consequence of decreased TGF-β1/2 secretion. Reduced TGF-β1 expression was also observed in THP-1 cultured in conditioned media from FAT1-depleted glioma cells, thus contributing to immune suppression. In U87MG cells, decreased TGF-β1 upon FAT1 knockdown was mediated by miR-663a, a known modulator. FAT1 expression was also observed to correlate positively with the expression of surrogate markers of MDSCs [programmed death ligand-1 (PD-L1), PD-L2, and interleukin (IL)-10] in glioma tumors, suggesting a potential role of FAT1 in MDSC-mediated immunosuppression. Hence, our findings elaborate contributions of FAT1 to immune evasion, where FAT1 enables an immunosuppressive microenvironment in GBM and other cancers via TGF-β1/2.
Collapse
Affiliation(s)
- Khushboo Irshad
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Chitrangda Srivastava
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Nargis Malik
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Manvi Arora
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Yakhlesh Gupta
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Sanjeev Goswami
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Chitra Sarkar
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Vaishali Suri
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Swati Mahajan
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Deepak Kumar Gupta
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | - Ashish Suri
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | | | - Subrata Sinha
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Kunzang Chosdol
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
35
|
Lin HY, Ko CJ, Lo TY, Wu SR, Lan SW, Huang CA, Lin YC, Lin HH, Tu HF, Lee CF, Hsiao PW, Huang HP, Chen MJ, Chang KH, Lee MS. Matriptase-2/NR4A3 axis switches TGF-β action toward suppression of prostate cancer cell invasion, tumor growth, and metastasis. Oncogene 2022; 41:2833-2845. [PMID: 35418692 DOI: 10.1038/s41388-022-02303-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/21/2022] [Accepted: 03/29/2022] [Indexed: 11/09/2022]
Abstract
Dysregulation of pericellular proteolysis is strongly implicated in cancer metastasis through alteration of cell invasion and the microenvironment. Matriptase-2 (MT-2) is a membrane-anchored serine protease which can suppress prostate cancer (PCa) cell invasion. In this study, we showed that MT-2 was down-regulated in PCa and could suppress PCa cell motility, tumor growth, and metastasis. Using microarray and biochemical analysis, we found that MT-2 shifted TGF-β action towards its tumor suppressor function by repressing epithelial-to-mesenchymal transition (EMT) and promoting Smad2 phosphorylation and nuclear accumulation to upregulate two TGF-β1 downstream effectors (p21 and PAI-1), culminating in hindrance of PCa cell motility and malignant growth. Mechanistically, MT-2 could dramatically up-regulate the expression of nuclear receptor NR4A3 via iron metabolism in PCa cells. MT-2-induced NR4A3 further coactivated Smad2 to activate p21 and PAI-1 expression. In addition, NR4A3 functioned as a suppressor of PCa and mediated MT-2 signaling to inhibit PCa tumorigenesis and metastasis. These results together indicate that NR4A3 sustains MT-2 signaling to suppress PCa cell invasion, tumor growth, and metastasis, and serves as a contextual factor for the TGF-β/Smad2 signaling pathway in favor of tumor suppression via promoting p21 and PAI-1 expression.
Collapse
Affiliation(s)
- Hsin-Ying Lin
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Chun-Jung Ko
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Tzu-Yu Lo
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Shang-Ru Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Shao-Wei Lan
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Chen-An Huang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Yi-Chin Lin
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Hsin-Hsien Lin
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Hsin-Fang Tu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Cheng-Fan Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Pei-Wen Hsiao
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan, ROC
| | - Hsiang-Po Huang
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Mei-Jou Chen
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Kai-Hsiung Chang
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan, ROC
| | - Ming-Shyue Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC.
| |
Collapse
|
36
|
Polygonum cuspidatum Extract (Pc-Ex) Containing Emodin Suppresses Lung Cancer-Induced Cachexia by Suppressing TCF4/TWIST1 Complex-Induced PTHrP Expression. Nutrients 2022; 14:nu14071508. [PMID: 35406121 PMCID: PMC9002362 DOI: 10.3390/nu14071508] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/01/2022] [Accepted: 04/01/2022] [Indexed: 12/18/2022] Open
Abstract
Cachexia, which is characterised by the wasting of fat and skeletal muscles, is the most common risk factor for increased mortality rates among patients with advanced lung cancer. PTHLH (parathyroid hormone-like hormone) is reported to be involved in the pathogenesis of cancer cachexia. However, the molecular mechanisms underlying the regulation of PTHLH expression and the inhibitors of PTHLH have not yet been identified. The PTHLH mRNA levels were measured using quantitative real-time polymerase chain reaction, while the PTHrP (parathyroid hormone-related protein) expression levels were measured using Western blotting and enzyme-linked immunosorbent assay. The interaction between TCF4 (Transcription Factor 4) and TWIST1 and the binding of the TCF4–TWIST1 complex to the PTHLH promoter were analysed using co-immunoprecipitation and chromatin immunoprecipitation. The results of the mammalian two-hybrid luciferase assay revealed that emodin inhibited TCF4–TWIST1 interaction. The effects of Polygonum cuspidatum extract (Pc-Ex), which contains emodin, on cachexia were investigated in vivo using A549 tumour-bearing mice. Ectopic expression of TCF4 upregulated PTHLH expression. Conversely, TCF4 knockdown downregulated PTHLH expression in lung cancer cells. The expression of PTHLH was upregulated in cells ectopically co-expressing TCF4 and TWIST1 when compared with that in cells expressing TCF4 or TWIST1 alone. Emodin inhibited the interaction between TCF4 and TWIST1 and consequently suppressed the TCF4/TWIST1 complex-induced upregulated mRNA and protein levels of PTHLH and PTHrP. Meanwhile, emodin-containing Pc-Ex significantly alleviated skeletal muscle atrophy and downregulated fat browning-related genes in A549 tumour-bearing mice. Emodin-containing Pc-Ex exerted therapeutic effects on lung cancer-associated cachexia by inhibiting TCF4/TWIST1 complex-induced PTHrP expression.
Collapse
|
37
|
Xiao JF, Kua LF, Ding LW, Sun QY, Myint KN, Chia XR, Venkatachalam N, Loh X, Duex JE, Neang V, Zhou S, Li Y, Yang H, Koeffler HP, Theodorescu D. KDM6A Depletion in Breast Epithelial Cells Leads to Reduced Sensitivity to Anticancer Agents and Increased TGFβ Activity. Mol Cancer Res 2022; 20:637-649. [PMID: 35022315 PMCID: PMC10030164 DOI: 10.1158/1541-7786.mcr-21-0402] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/29/2021] [Accepted: 01/05/2022] [Indexed: 11/16/2022]
Abstract
KDM6A, an X chromosome-linked histone lysine demethylase, was reported to be frequently mutated in many tumor types including breast and bladder cancer. However, the functional role of KDM6A is not fully understood. Using MCF10A as a model of non-tumorigenic epithelial breast cells, we found that silencing KDM6A promoted cell migration and transformation demonstrated by the formation of tumor-like acini in three-dimensional culture. KDM6A loss reduced the sensitivity of MCF10A cells to therapeutic agents commonly used to treat patients with triple-negative breast cancer and also induced TGFβ extracellular secretion leading to suppressed expression of cytotoxic genes in normal human CD8+ T cells in vitro. Interestingly, when cells were treated with TGFβ, de novo synthesis of KDM6A protein was suppressed while TGFB1 transcription was enhanced, indicating a TGFβ/KDM6A-negative regulatory axis. Furthermore, both KDM6A deficiency and TGFβ treatment promoted disorganized acinar structures in three-dimensional culture, as well as transcriptional profiles associated with epithelial-to-mesenchymal transition and metastasis, suggesting KDM6A depletion and TGFβ drive tumor progression. IMPLICATIONS Our study provides the preclinical rationale for evaluating KDM6A and TGFβ in breast tumor samples as predictors for response to chemo and immunotherapy, informing personalized therapy based on these findings.
Collapse
Affiliation(s)
- Jin-Fen Xiao
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Division of Medical Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Surgery (Urology), Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Corresponding authors: Dan Theodorescu, Address: 8700 Beverly Blvd, NT-Plaza Level 2429C, Los Angeles, CA 90048; , Phone: +1(310)-423-8431; Jin-Fen Xiao, Address: Davis Research Building RM3057, 110 N George Burns Rd, Los Angeles, CA 90048; ; Phone: 1(310)423-1326
| | - Ley-Fang Kua
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Ling-Wen Ding
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Qiao-Yang Sun
- Department of Hematology, Singapore General Hospital, Singapore
| | - Khine Nyein Myint
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Xiu-Rong Chia
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | | | - Xinyi Loh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Jason E. Duex
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
| | - Vanessa Neang
- Division of Medical Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Siqin Zhou
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Ying Li
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - H. Phillip Koeffler
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Division of Medical Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dan Theodorescu
- Department of Surgery (Urology), Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Corresponding authors: Dan Theodorescu, Address: 8700 Beverly Blvd, NT-Plaza Level 2429C, Los Angeles, CA 90048; , Phone: +1(310)-423-8431; Jin-Fen Xiao, Address: Davis Research Building RM3057, 110 N George Burns Rd, Los Angeles, CA 90048; ; Phone: 1(310)423-1326
| |
Collapse
|
38
|
C/EBPβ isoform-specific regulation of migration and invasion in triple-negative breast cancer cells. NPJ Breast Cancer 2022; 8:11. [PMID: 35042889 PMCID: PMC8766495 DOI: 10.1038/s41523-021-00372-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 12/06/2021] [Indexed: 12/29/2022] Open
Abstract
The transcription factor C/EBPβ is a master regulator of mammary gland development and tissue remodelling during lactation. The CEBPB-mRNA is translated into three distinct protein isoforms named C/EBPβ-LAP1, -LAP2 and -LIP that are functionally different. The smaller isoform LIP lacks the N-terminal transactivation domains and is considered to act as an inhibitor of the transactivating LAP1/2 isoforms by competitive binding for the same DNA recognition sequences. Aberrantly high expression of LIP is associated with mammary epithelial proliferation and is found in grade III, estrogen receptor (ER) and progesterone (PR) receptor-negative human breast cancer. Here, we show that reverting the high LIP/LAP ratios in triple-negative breast cancer (TNBC) cell lines into low LIP/LAP ratios by overexpression of LAP reduces migration and matrix invasion of these TNBC cells. In addition, in untransformed MCF10A human mammary epithelial cells overexpression of LIP stimulates migration. Knockout of CEBPB in TNBC cells where LIP expression prevails, resulted in strongly reduced migration that was accompanied by a downregulation of genes involved in cell migration, extracellular matrix production and cytoskeletal remodelling, many of which are epithelial to mesenchymal transition (EMT) marker genes. Together, this study suggests that the LIP/LAP ratio is involved in regulating breast cancer cell migration and invasion. This study together with studies from others shows that understanding the functions the C/EBPβ-isoforms in breast cancer development may reveal new avenues of treatment.
Collapse
|
39
|
Zhang G, Li Z, Dong J, Zhou W, Zhang Z, Que Z, Zhu X, Xu Y, Cao N, Zhao A. Acacetin inhibits invasion, migration and TGF-β1-induced EMT of gastric cancer cells through the PI3K/Akt/Snail pathway. BMC Complement Med Ther 2022; 22:10. [PMID: 35000605 PMCID: PMC8744305 DOI: 10.1186/s12906-021-03494-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 12/21/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Epithelial-to-mesenchymal transition (EMT) is a pivotal cellular phenomenon involved in tumour metastasis and progression. In gastric cancer (GC), EMT is the main reason for recurrence and metastasis in postoperative patients. Acacetin exhibits various biological activities. However, the inhibitory effect of acacetin on EMT in GC is still unknown. Herein, we explored the possible mechanism of acacetin on EMT in GC in vitro and in vivo. METHODS In vitro, MKN45 and MGC803 cells were treated with acacetin, after which cell viability was detected by CCK-8 assays, cell migration and invasion were detected by using Transwell and wound healing assays, and protein expression was analysed by western blots and immunofluorescence staining. In vivo, a peritoneal metastasis model of MKN45 GC cells was used to investigate the effects of acacetin. RESULTS Acacetin inhibited the proliferation, invasion and migration of MKN45 and MGC803 human GC cells by regulating the expression of EMT-related proteins. In TGF-β1-induced EMT models, acacetin reversed the morphological changes from epithelial to mesenchymal cells, and invasion and migration were limited by regulating EMT. In addition, acacetin suppressed the activation of PI3K/Akt signalling and decreased the phosphorylation levels of TGF-β1-treated GC cells. The in vivo experiments demonstrated that acacetin delayed the development of peritoneal metastasis of GC in nude mice. Liver metastasis was restricted by altering the expression of EMT-related proteins. CONCLUSION Our study showed that the invasion, metastasis and TGF-β1-induced EMT of GC are inhibited by acacetin, and the mechanism may involve the suppression of the PI3K/Akt/Snail signalling pathway. Therefore, acacetin is a potential therapeutic reagent for the treatment of GC patients with recurrence and metastasis.
Collapse
Affiliation(s)
- Guangtao Zhang
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Zhaoyan Li
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.,Department of Oncology, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiahuan Dong
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Weili Zhou
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Zhanxia Zhang
- Institute of Traditional Chinese Medicine Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zujun Que
- Institute of Traditional Chinese Medicine Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,School of Oncology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaohong Zhu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yan Xu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Nida Cao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Aiguang Zhao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
40
|
Trebska-McGowan K, Chaib M, Alvarez MA, Kansal R, Pingili AK, Shibata D, Makowski L, Glazer ES. TGF-β Alters the Proportion of Infiltrating Immune Cells in a Pancreatic Ductal Adenocarcinoma. J Gastrointest Surg 2022; 26:113-121. [PMID: 34260016 DOI: 10.1007/s11605-021-05087-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 06/27/2021] [Indexed: 01/31/2023]
Abstract
PURPOSE Immunotherapy, such as checkpoint inhibitors against anti-programmed death-ligand 1 (PD-L1), has not been successful in treating patients with pancreatic ductal adenocarcinoma (PDAC). Tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), dendritic cells (DCs), and the TGF-β cytokine are critical in anti-cancer immunity. We hypothesized that TGF-β enhances the immunosuppressive effects of TAM, MDSC, and DC presence in tumors. METHODS Using a murine PDAC cell line derived from a genetically engineered mouse model, we orthotopically implanted treated cells plus drug embedded in Matrigel into immunocompetent mice. Treatments included saline control, TGF-β1, or a TGF-β receptor 1 small molecule inhibitor, galunisertib. We investigated TAM, MDSC, DC, and TAM PD-L1 expression with flow cytometry in tumors. Separately, we used the TIMER2.0 database to analyze TAM and PD-L1 gene expression in human PDAC tumors in TCGA database. RESULTS TGF-β did not alter MDSC or DC frequencies in the primary tumors. However, in PDAC metastases to the liver, TGF-β decreased the proportion of MDSCs (P=0.022) and DCs (P=0.005). TGF-β significantly increased the percent of high PD-L1 expressing TAMs (32 ± 6 % vs. 12 ± 5%, P=0.013) but not the proportion of TAMs in primary and metastatic tumors. TAM PD-L1 gene expression in TCGA PDAC database was significantly correlated with tgb1 and tgfbr1 gene expression (P<0.01). CONCLUSIONS TGF-β is important in PDAC anti-tumor immunity, demonstrating context-dependent impact on immune cells. TGF-β has an overall immunosuppressive effect mediated by TAM PD-L1 expression and decreased presence of DCs. Future investigations will focus on enhancing anti-cancer immune effects of TGF-β receptor inhibition.
Collapse
Affiliation(s)
- Kasia Trebska-McGowan
- Divisiion of Surgical Oncology, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, 910 Madison Ave, Suite 325, Memphis, TN, 38163, USA
| | - Mehdi Chaib
- Division of Hematology Oncology, Department of Medicine, College of Medicine, The University of Tennessee Health Science Center, Memphis, USA
| | - Marcus A Alvarez
- Divisiion of Surgical Oncology, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, 910 Madison Ave, Suite 325, Memphis, TN, 38163, USA
| | - Rita Kansal
- Divisiion of Surgical Oncology, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, 910 Madison Ave, Suite 325, Memphis, TN, 38163, USA
| | - Ajeeth K Pingili
- Division of Hematology Oncology, Department of Medicine, College of Medicine, The University of Tennessee Health Science Center, Memphis, USA
| | - David Shibata
- Divisiion of Surgical Oncology, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, 910 Madison Ave, Suite 325, Memphis, TN, 38163, USA
- Center for Cancer Research, The University of Tennessee Health Science Center, Memphis, USA
| | - Liza Makowski
- Division of Hematology Oncology, Department of Medicine, College of Medicine, The University of Tennessee Health Science Center, Memphis, USA
- Center for Cancer Research, The University of Tennessee Health Science Center, Memphis, USA
| | - Evan S Glazer
- Divisiion of Surgical Oncology, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, 910 Madison Ave, Suite 325, Memphis, TN, 38163, USA.
- Center for Cancer Research, The University of Tennessee Health Science Center, Memphis, USA.
| |
Collapse
|
41
|
Rohini M, Vairamani M, Selvamurugan N. TGF-β1-stimulation of NFATC2 and ATF3 proteins and their interaction for matrix metalloproteinase 13 expression in human breast cancer cells. Int J Biol Macromol 2021; 192:1325-1330. [PMID: 34687766 DOI: 10.1016/j.ijbiomac.2021.10.099] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 09/30/2021] [Accepted: 10/14/2021] [Indexed: 01/25/2023]
Abstract
Activating transcription factor 3 (ATF3), an inducible stress gene, is stimulated by transforming growth factor-beta1 (TGF-β1) in a protracted and relentless manner in human mammary cancer cells (hBC cells; MDA-MB231). The molecular mechanism behind this stable expression of ATF3 via TGF-β1 in MDA-MB231 cells is unknown. This study found that TGF-β1 stimulated the expression of the nuclear factor of activated T Cells 2 (NFATC2) in MDA-MB231 cells and provided evidence of its interaction with ATF3. The functional characterization of NFATC2 in association with ATF3 was determined by silencing of NFATC2 using siRNA. Knock-down of NFATC2 decreased the expression of both ATF3 and its target gene MMP13 (matrix metalloproteinase 13, a critical invasive gene) in hBC cells. Chromatin immunoprecipitation revealed that TGF-β1 promoted NFATC2 binding and NFATC2-ATF3 complex binding at the MMP13 promoter region, whereas silencing of NFATC2 decreased their binding in hBC cells. Thus, we uncovered the mechanism of interaction between NFATC2 and ATF3 regulated by TGF-β1, and NFATC2 acted as a pivotal factor in providing ATF3 stability and further drove MMP13 transcription. Targeting NFATC2 and blocking its association with ATF3 could therefore help to slow the progression of breast cancer.
Collapse
Affiliation(s)
- M Rohini
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - M Vairamani
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - N Selvamurugan
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India.
| |
Collapse
|
42
|
Luo F, Huang Y, Li Y, Zhao X, Xie Y, Zhang Q, Mei J, Liu X. A narrative review of the relationship between TGF-β signaling and gynecological malignant tumor. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1601. [PMID: 34790807 PMCID: PMC8576662 DOI: 10.21037/atm-21-4879] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 10/14/2021] [Indexed: 12/24/2022]
Abstract
Objective This paper reviews the association between transforming growth factor-β (TGF-β) and its receptor and tumor, focusing on gynecological malignant tumors. we hope to provide more methods to help increase the potential of TGF-β signaling targeted treatment of specific cancers. Background The occurrence of a malignant tumor is a complex process of multi-step, multi-gene regulation, and its progression is affected by various components of the tumor cells and/or tumor microenvironment. The occurrence of gynecological diseases not only affect women's health, but also bring some troubles to their normal life. Especially when gynecological malignant tumors occur, the situation is more serious, which will endanger the lives of patients. Due to differences in environmental and economic conditions, not all women have access to assistance and treatment specifically meeting their needs. TGF-β is a multi-potent growth factor that maintains homeostasis in mammals by inhibiting cell growth and promoting apoptosis in vivo. TGF-β signaling is fundamental to inflammatory disease and favors the emergence of tumors, and it also plays an important role in immunosuppression in the tumor microenvironment. In the early stages of the tumor, TGF-β acts as a tumor inhibitor, whereas in advanced tumors, mutations or deletion of the TGF-β signaling core component initiate neogenesis. Methods Literatures about TGF-β and gynecological malignant tumor were extensively reviewed to analyze and discuss. Conclusions We discussed the role of TGF-β signaling in different types of gynecological tumor cells, thus demonstrating that targeted TGF-β signaling may be an effective tumor treatment strategy.
Collapse
Affiliation(s)
- Fangyuan Luo
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, China.,Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Yu Huang
- Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Yilin Li
- Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Xiaolan Zhao
- Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Yao Xie
- Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Qianwen Zhang
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Jie Mei
- Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Xinghui Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
43
|
Li J, Cui P, Sun Q, Du Z, Chen Z, Li Z, Liu C, Cao Y, Yang Z, Liu R, Luo M. PSPC1 regulates CHK1 phosphorylation through phase separation and participates in mouse oocyte maturation. Acta Biochim Biophys Sin (Shanghai) 2021; 53:1527-1537. [PMID: 34490876 DOI: 10.1093/abbs/gmab123] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Indexed: 12/21/2022] Open
Abstract
Liquid-liquid phase separation (LLPS) underlies the formation of membraneless compartments in mammal cells. However, there are few reports that focus on the correlation of mouse oocyte maturation with LLPS. Previous studies have reported that paraspeckle component 1 (PSPC1) is related to the occurrence and development of tumors, but whether PSPC1 functions in mouse oocyte maturation is still unclear. Sequence analysis of PSPC1 protein showed that it contains a prion-like domain (PrLD) that is required for phase separation of proteins. In this study, we found that PSPC1 could undergo phase separation. Moreover, the loss of PrLD domain of PSPC1 could greatly weaken its phase separation ability. The immunofluorescence assays showed that PSPC1 is present in mouse oocytes in the germinal vesicle (GV) stage. Knockdown of PSPC1 significantly impeded the maturation of mouse oocytes in vitro. CHK1 has been reported to play important roles in the GV stage of mouse oocytes. Co-IP experiment revealed that PSPC1 could interact with phosphatase serine/threonine-protein phosphatase 5 (PPP5C), which regulates CHK1 phosphorylation. Western blot analysis revealed that PSPC1 could regulate the phosphorylation of CHK1 through PPP5C; however, PSPC1 without PrLD domain was inactive, suggesting that the lack of phase separation ability led to the abnormal function of PSPC1 in regulating CHK1 phosphorylation. Thus, we conclude that PSPC1 may undergo phase separation to regulate the phosphorylation level of CHK1 via PPP5C and participate in mouse oocyte maturation. Our study provides new insights into the mechanism of mouse oocyte maturation.
Collapse
Affiliation(s)
- Jiong Li
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Tissue and Embryology, School of Basic Medical Sciences, Wuhan University, Wuhan 430062, China
| | - Peng Cui
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Tissue and Embryology, School of Basic Medical Sciences, Wuhan University, Wuhan 430062, China
| | - Qi Sun
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Tissue and Embryology, School of Basic Medical Sciences, Wuhan University, Wuhan 430062, China
| | - Ziye Du
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Tissue and Embryology, School of Basic Medical Sciences, Wuhan University, Wuhan 430062, China
| | - Zhen Chen
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Tissue and Embryology, School of Basic Medical Sciences, Wuhan University, Wuhan 430062, China
| | - Zejia Li
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Tissue and Embryology, School of Basic Medical Sciences, Wuhan University, Wuhan 430062, China
| | - Cong Liu
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Tissue and Embryology, School of Basic Medical Sciences, Wuhan University, Wuhan 430062, China
| | - Yuming Cao
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Tissue and Embryology, School of Basic Medical Sciences, Wuhan University, Wuhan 430062, China
| | - Zhe Yang
- College of Life Science, Liaoning University, Shenyang 110036, China
| | - Rong Liu
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Tissue and Embryology, School of Basic Medical Sciences, Wuhan University, Wuhan 430062, China
| | - Mengcheng Luo
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Tissue and Embryology, School of Basic Medical Sciences, Wuhan University, Wuhan 430062, China
| |
Collapse
|
44
|
Trivedi T, Pagnotti GM, Guise TA, Mohammad KS. The Role of TGF-β in Bone Metastases. Biomolecules 2021; 11:1643. [PMID: 34827641 PMCID: PMC8615596 DOI: 10.3390/biom11111643] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/29/2021] [Accepted: 10/29/2021] [Indexed: 02/07/2023] Open
Abstract
Complications associated with advanced cancer are a major clinical challenge and, if associated with bone metastases, worsen the prognosis and compromise the survival of the patients. Breast and prostate cancer cells exhibit a high propensity to metastasize to bone. The bone microenvironment is unique, providing fertile soil for cancer cell propagation, while mineralized bone matrices store potent growth factors and cytokines. Biologically active transforming growth factor β (TGF-β), one of the most abundant growth factors, is released following tumor-induced osteoclastic bone resorption. TGF-β promotes tumor cell secretion of factors that accelerate bone loss and fuel tumor cells to colonize. Thus, TGF-β is critical for driving the feed-forward vicious cycle of tumor growth in bone. Further, TGF-β promotes epithelial-mesenchymal transition (EMT), increasing cell invasiveness, angiogenesis, and metastatic progression. Emerging evidence shows TGF-β suppresses immune responses, enabling opportunistic cancer cells to escape immune checkpoints and promote bone metastases. Blocking TGF-β signaling pathways could disrupt the vicious cycle, revert EMT, and enhance immune response. However, TGF-β's dual role as both tumor suppressor and enhancer presents a significant challenge in developing therapeutics that target TGF-β signaling. This review presents TGF-β's role in cancer progression and bone metastases, while highlighting current perspectives on the therapeutic potential of targeting TGF-β pathways.
Collapse
Affiliation(s)
- Trupti Trivedi
- Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.T.); (G.M.P.); (T.A.G.)
| | - Gabriel M. Pagnotti
- Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.T.); (G.M.P.); (T.A.G.)
| | - Theresa A. Guise
- Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.T.); (G.M.P.); (T.A.G.)
| | - Khalid S. Mohammad
- Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.T.); (G.M.P.); (T.A.G.)
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
45
|
Babyshkina N, Dronova T, Erdyneeva D, Gervas P, Cherdyntseva N. Role of TGF-β signaling in the mechanisms of tamoxifen resistance. Cytokine Growth Factor Rev 2021; 62:62-69. [PMID: 34635390 DOI: 10.1016/j.cytogfr.2021.09.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/18/2021] [Accepted: 09/19/2021] [Indexed: 12/24/2022]
Abstract
The transforming growth factor beta (TGF-β) signaling pathway plays complex role in the regulation of cell proliferation, apoptosis and differentiation in breast cancer. TGF-β activation can lead to multiple cellular responses mediating the drug resistance evolution, including the resistance to antiestrogens. Tamoxifen is the most commonly prescribed antiestrogen that functionally involved in regulation of TGF-β activity. In this review, we focus on the role of TGF-β signaling in the mechanisms of tamoxifen resistance, including its interaction with estrogen receptors alfa (ERα) pathway and breast cancer stem cells (BCSCs). We summarize the current reported data regarding TGF-β signaling components as markers of tamoxifen resistance and review current approaches to overcoming tamoxifen resistance based on studies of TGF-β signaling.
Collapse
Affiliation(s)
- Nataliya Babyshkina
- Department of Molecular Oncology and Immunology, Саncеr Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634050, Russian Federation; Siberian State Medical University, Tomsk 634050, Russian Federation.
| | - Tatyana Dronova
- Department of Biology of Tumor Progression, Саncеr Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634050, Russian Federation
| | - Daiana Erdyneeva
- Department of Molecular Oncology and Immunology, Саncеr Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634050, Russian Federation
| | - Polina Gervas
- Department of Molecular Oncology and Immunology, Саncеr Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634050, Russian Federation
| | - Nadejda Cherdyntseva
- Department of Molecular Oncology and Immunology, Саncеr Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634050, Russian Federation
| |
Collapse
|
46
|
Ye Z, Li W, Jiang Z, Wang E, Wang J. An intermediate state in trans-differentiation with proliferation, metabolic, and epigenetic switching. iScience 2021; 24:103057. [PMID: 34541470 PMCID: PMC8441076 DOI: 10.1016/j.isci.2021.103057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/23/2021] [Accepted: 08/25/2021] [Indexed: 01/03/2023] Open
Abstract
Although TGF-β signaling can effectively activate fibroblasts to transform to myofibroblasts, the underlying mechanisms involved in the cell fate switching for trans-differentiation have not been fully elucidated. In this study, we found the evidence of an intermediate state in the process of trans-differentiation. In the early stage of trans-differentiation, cells enter the intermediate state first with multiple characteristics such as accelerating cell cycle, metabolic switching, enhanced anti-apoptotic ability, and pluripotency, which is very similar to the early stage of reprogramming. As the trans-differentiation continues, these characteristics get switched. Therefore, trans-differentiation appears to require the switching of cell proliferation ability, metabolic pathway, and “stemness” to complete the process. In this study, we can conclude that an intermediate state may be necessary with high pluripotency in trans-differentiation from fibroblasts to myofibroblasts. Only after passing the intermediate state, the trans-differentiation is finally completed and will not easily return to the original state. Smads and Akt/p38MAPK pathways play the key role in fibroblast transition There is a cell proliferation capability switching induced by TGF-β1 Metabolic reprogramming is required during trans-differentiation An intermediate state appeared with high pluripotency in trans-differentiation
Collapse
Affiliation(s)
- Zhikai Ye
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.,University of Science and Technology of China, Hefei, Anhui 230029, P. R. China
| | - Wenbo Li
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Zhenlong Jiang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Erkang Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.,University of Science and Technology of China, Hefei, Anhui 230029, P. R. China
| | - Jin Wang
- Department of Chemistry, Physics and Applied Mathematics, State University of New York at Stony Brook., Stony Brook, NY 11794-3400, USA
| |
Collapse
|
47
|
He D, Gao J, Zheng L, Liu S, Ye L, Lai H, Pan B, Pan W, Lou C, Chen Z, Fan S. TGF‑β inhibitor RepSox suppresses osteosarcoma via the JNK/Smad3 signaling pathway. Int J Oncol 2021; 59:84. [PMID: 34533199 PMCID: PMC8460063 DOI: 10.3892/ijo.2021.5264] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 07/29/2021] [Indexed: 02/06/2023] Open
Abstract
Osteosarcoma (OS) is the most common malignant bone tumor and the long-term survival rates remain unsatisfactory. Transforming growth factor-β (TGF-β) has been revealed to play a crucial role in OS progression, and RepSox is an effective TGF-β inhibitor. In the present study, the effect of RepSox on the proliferation of the OS cell lines (HOS and 143B) was detected. The results revealed that RepSox effectively inhibited the proliferation of OS cells by inducing S-phase arrest and apoptosis. Moreover, the inhibitory effect of RepSox on cell migration and invasion was confirmed by wound-healing and Transwell assays. Furthermore, western blotting revealed that the protein levels of molecules associated with the epithelial-mesenchymal transition (EMT) phenotype, including E-cadherin, N-cadherin, Vimentin, matrix metalloproteinase (MMP)-2 and MMP-9, were reduced by RepSox treatment. Concurrently, it was also revealed that the JNK and Smad3 signaling pathway was inhibited. Our in vivo findings using a xenograft model also revealed that RepSox markedly inhibited the growth of tumors. In general, our data demonstrated that RepSox suppressed OS proliferation, EMT and promoted apoptosis by inhibiting the JNK/Smad3 signaling pathway. Thus, RepSox may be a potential anti-OS drug.
Collapse
Affiliation(s)
- Dengwei He
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, P.R. China
| | - Jiawei Gao
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, P.R. China
| | - Lin Zheng
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, P.R. China
| | - Shijie Liu
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, P.R. China
| | - Lin Ye
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, P.R. China
| | - Hehuan Lai
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, P.R. China
| | - Bin Pan
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, P.R. China
| | - Wenzheng Pan
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, P.R. China
| | - Chao Lou
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, P.R. China
| | - Zhenzhong Chen
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, P.R. China
| | - Shunwu Fan
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| |
Collapse
|
48
|
Robbins Y, Friedman J, Clavijo PE, Sievers C, Bai K, Donahue RN, Schlom J, Sinkoe A, Hinrichs CS, Allen C, Abdul Sater H, Gulley JL, Norberg S. Dual PD-L1 and TGF-b blockade in patients with recurrent respiratory papillomatosis. J Immunother Cancer 2021; 9:jitc-2021-003113. [PMID: 34462327 PMCID: PMC8407210 DOI: 10.1136/jitc-2021-003113] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2021] [Indexed: 12/26/2022] Open
Abstract
Background Recurrent respiratory papillomatosis (RRP) is a human papillomavirus (HPV) driven neoplastic disorder of the upper aerodigestive tract that causes significant morbidity and can lead to fatal airway obstruction. Prior clinical study demonstrated clinical benefit with the programmed death-ligand 1 (PD-L1) monoclonal antibody avelumab. Bintrafusp alfa is a bifunctional inhibitor of PD-L1 and transforming growth factor-beta (TGF-b) that has shown clinical activity in several cancer types. Methods We conducted a phase II clinical trial evaluating bintrafusp alfa in adults with RRP. Papilloma samples before and after treatment with bintrafusp alfa were assessed for correlates of response with multiplex immunofluorescence as well as immunological and genomic analyses. Post hoc analyses of papilloma samples before and after treatment with avelumab were assessed for comparison. Results Dual PD-L1/TGF-b inhibition failed to abrogate papilloma growth in most subjects and increased the frequency of clinically indicated interventions after treatment in four of eight subjects based on each subject’s own historical control. TGF-b neutralization consistently decreased pSMAD3 and p21 and increased Ki67 expression within the basal layers of papillomas, indicating that TGF-b restrained proliferation. These alterations were not observed in papillomas treated with PD-L1 blockade alone. Dual PD-L1/TGF-b inhibition did not enhance anti-HPV immunity within papillomas beyond that observed with PD-L1 blockade. Genomic alterations in TGF-b superfamily genes were infrequent in papillomas and normal mucosa but present in a significant fraction of head and neck carcinomas. Conclusions Intact TGF-b signaling restrains proliferation within papillomas, and the use of clinical agents that abrogate this pathway should be avoided in patients with RRP. Trial registration numbers NCT03707587 and NCT02859454.
Collapse
Affiliation(s)
- Yvette Robbins
- Section on Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Jay Friedman
- Section on Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Paul E Clavijo
- Section on Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Cem Sievers
- Section on Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Ke Bai
- Section on Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Renee N Donahue
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, Bethesda, Maryland, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, Bethesda, Maryland, USA
| | - Andrew Sinkoe
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | | | - Clint Allen
- Section on Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Houssein Abdul Sater
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - James L Gulley
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Scott Norberg
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
49
|
Zhang N, Ng AS, Cai S, Li Q, Yang L, Kerr D. Novel therapeutic strategies: targeting epithelial-mesenchymal transition in colorectal cancer. Lancet Oncol 2021; 22:e358-e368. [PMID: 34339656 DOI: 10.1016/s1470-2045(21)00343-0] [Citation(s) in RCA: 171] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/30/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is a process during which cells lose their epithelial characteristics, for instance apical-basal cell polarity and cell-cell contact, and gain mesenchymal properties, such as increased motility. In colorectal cancer, EMT has an important role in tumour progression, metastasis, and drug resistance. There has been accumulating evidence from preclinical and early clinical studies that show that EMT markers might serve as outcome predictors and potential therapeutic targets in colorectal cancer. This Review describes the fundamentals of EMT, including biology, newly partial EMT, and associated changes. We also provide a comprehensive summary of therapeutic compounds capable of targeting EMT markers, including drugs in preclinical and clinical trials and those with repurpose potential. Lastly, we explore the obstacles of EMT bench-to-bedside drug development.
Collapse
Affiliation(s)
- Nan Zhang
- West China School of Medicine, Sichuan University, Chengdu, China; Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK; University of Oxford-Sichuan University Huaxi Joint Centre for Gastrointestinal Cancer, Oxford, UK; Department of Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Aik Seng Ng
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK; University of Oxford-Sichuan University Huaxi Joint Centre for Gastrointestinal Cancer, Oxford, UK
| | - Shijie Cai
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK; University of Oxford-Sichuan University Huaxi Joint Centre for Gastrointestinal Cancer, Oxford, UK
| | - Qiu Li
- West China School of Medicine, Sichuan University, Chengdu, China; Department of Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Li Yang
- West China School of Medicine, Sichuan University, Chengdu, China; University of Oxford-Sichuan University Huaxi Joint Centre for Gastrointestinal Cancer, Oxford, UK; Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China.
| | - David Kerr
- West China School of Medicine, Sichuan University, Chengdu, China; Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK; University of Oxford-Sichuan University Huaxi Joint Centre for Gastrointestinal Cancer, Oxford, UK
| |
Collapse
|
50
|
Chandra Jena B, Sarkar S, Rout L, Mandal M. The transformation of cancer-associated fibroblasts: Current perspectives on the role of TGF-β in CAF mediated tumor progression and therapeutic resistance. Cancer Lett 2021; 520:222-232. [PMID: 34363903 DOI: 10.1016/j.canlet.2021.08.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/16/2021] [Accepted: 08/01/2021] [Indexed: 12/12/2022]
Abstract
Over the last few years, the Transforming growth factor- β (TGF-β) has been significantly considered as an effective and ubiquitous mediator of cell growth. The cytokine, TGF-β is being increasingly recognized as the most potent inducer of cancer cell initiation, differentiation, migration as well as progression through both the SMAD-dependent and independent pathways. There is growing evidence that supports the role of secretory cytokine TGF-β as a crucial mediator of tumor-stroma crosstalk. Contextually, the CAFs are the prominent component of tumor stroma that helps in tumor progression and onset of chemoresistance. The interplay between the CAFs and the tumor cells through the paracrine signals is facilitated by cytokine TGF-β to induce the malignant progression. Here in this review, we have dissected the most recent advancements in understanding the mechanisms of TGF-β induced CAF activation, their multiple origins, and most importantly their role in conferring chemoresistance. Considering the pivotal role of TGF-β in tumor perogression and associated stemness, it is one the proven clinical targets We have also included the clinical trials going on, targeting the TGF-β and CAFs crosstalk with the tumor cells. Ultimately, we have underscored some of the outstanding issues that must be deciphered with utmost importance to unravel the successful strategies of anti-cancer therapies.
Collapse
Affiliation(s)
- Bikash Chandra Jena
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| | - Siddik Sarkar
- CSIR-Indian Institue of Chemical Biology, Translational Research Unit of Excellence, Kolkata, West Bengal, India
| | - Lipsa Rout
- Department of Chemistry, Institute of Technical Education and Research, Siksha'O'Anusandhan Deemed to be University, Bhubaneswar, Odisha, India
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India.
| |
Collapse
|