1
|
付 玮, 宁 静, 付 伟, 张 静, 丁 士. [Effect of CMTM6 on PD-L1 in Helicobacter pylori infected gastric epithelial cells]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2025; 57:245-252. [PMID: 40219552 PMCID: PMC11992445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Indexed: 04/14/2025]
Abstract
OBJECTIVE To explore the changes of CKLF-like MARVEL transmembrane domain-containing 6 (CMTM6) and programmed death-ligand 1 (PD-L1) expression in gastric mucosal epithelial cells after Helicobacter pylori infection and the regulation of CMTM6 on PD-L1, and to analyze the mRNA expression differences before and after CMTM6 gene knock-out in helicobacter pylori infected gastric epithelial cells by microarray analysis. METHODS The standard Helicobacter pylori strain ATCC 26695 was co-cultured with human gastric epithelial cell GES-1 for 6, 24 and 48 hours, and the mRNA and protein levels of CMTM6 and PD-L1 were detected by real-time quantitative PCR and Western blot. Using CRISPR/Cas9 to construct CMTM6 gene knockout plasmid and knockout CMTM6 gene of GES-1 cells. Helicobacter pylori was co-cultured with CMTM6 gene knockout and wild type GES-1 cells for 48 hours to detect PD-L1 transcription and protein level changes, and CMTM6 gene knockout GES-1 cells were treated with the proteasome inhibitor MG-132 to detect the changes in PD-L1 protein levels. Agilent Human ceRNA Microarray 2019 was used to detect the differentially expressed genes in CMTM6 gene knockout and wild-type GES-1 cells co-cultured with Hp for 48 hours, and the signal pathway of differentially expressed genes enrichment was analyzed by Kyoto Encyclopedia of Genes and Genomes (KEGG) database. RESULTS The mRNA and protein levels of CMTM6 and PD-L1 in GES-1 cells were significantly up-regulated after Helicobacter pylori infection, and CMTM6 mRNA was most significantly up-regulated 48 hours after infection. After CMTM6 gene knockout, the CD274 gene transcription level of Helicobacter pylori infected GES-1 cells did not change significantly, but PD-L1 protein level was significantly down-regulated, and the PD-L1 level increased after the application of proteasome inhibitor MG-132. After CMTM6 gene knockout, 67 genes had more than two times of differential expression. The transcription levels of TMEM68, FERMT3, GPR142, ATP6V1FNB, NOV, UBE2S and other genes were significantly down-regulated. The transcription levels of PCDHGA6, CAMKMT, PDIA2, NTRK3, SPOCK1 and other genes were significantly up-regulated. After CMTM6 gene knockout, ubiquitin-conjugating enzyme E2S (UBE2S) gene expression was significantly down-regulated, which might affect protein ubiquitination degradation. After CMTM6 gene knockout, adrenoceptor alpha 1B (ADRA1B), cholinergic receptor muscarinic 1 (M1), CHRM1, platelet activating factor receptor (PTAFR) gene expression was significantly up-regulated. CONCLUSION Helicobacter pylori infection up-regulates the expression level of CMTM6 in gastric mucosa cells, and CMTM6 can stabilize PD-L1 and maintain the protein level of PD-L1. CMTM6 gene knockout may affect biological behaviors such as protein ubiquitination and cell surface receptor expression.
Collapse
Affiliation(s)
- 玮 付
- />北京大学第三医院消化科,幽门螺杆菌感染及上胃肠疾病防治研究北京市重点实验室,北京 100191Department of Gastroenterology, Peking University Third Hospital; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| | - 静 宁
- />北京大学第三医院消化科,幽门螺杆菌感染及上胃肠疾病防治研究北京市重点实验室,北京 100191Department of Gastroenterology, Peking University Third Hospital; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| | - 伟伟 付
- />北京大学第三医院消化科,幽门螺杆菌感染及上胃肠疾病防治研究北京市重点实验室,北京 100191Department of Gastroenterology, Peking University Third Hospital; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| | - 静 张
- />北京大学第三医院消化科,幽门螺杆菌感染及上胃肠疾病防治研究北京市重点实验室,北京 100191Department of Gastroenterology, Peking University Third Hospital; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| | - 士刚 丁
- />北京大学第三医院消化科,幽门螺杆菌感染及上胃肠疾病防治研究北京市重点实验室,北京 100191Department of Gastroenterology, Peking University Third Hospital; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| |
Collapse
|
2
|
Yang L, Miao Z, Li N, Meng L, Feng Q, Qiao D, Wang P, Wang Y, Bai Y, Li Z, Lian S. CMTM4 promotes the motility of colon cancer cells under radiation and is associated with an unfavorable neoadjuvant chemoradiotherapy response and patient survival in rectal cancer. Oncol Lett 2025; 29:138. [PMID: 39839608 PMCID: PMC11747855 DOI: 10.3892/ol.2025.14884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 12/02/2024] [Indexed: 01/23/2025] Open
Abstract
Neoadjuvant chemoradiotherapy (nCRT) is the standard treatment for locally advanced rectal cancer (LARC). Pathological complete regression is closely linked to disease outcomes. However, biomarkers predicting nCRT response and patient survival are lacking for LARC. In the present study, the clinical characteristics and follow-up information of 228 patients with LARC were retrospectively collected. Immunohistochemistry (IHC), reverse transcription-quantitative PCR (RT-qPCR), Kaplan-Meier and multivariate analyses were used to evaluate the expression and predict the role of CKLF-like MARVEL transmembrane domain member 4 (CMTM4) in LARC. Additionally, lentiviral short hairpin (sh)RNA was used to interfere with CMTM4 expression. The phenotype of CMTM4-knockdown LoVo cells was determined by colony formation, migration and invasion assays under irradiation (IR) treatment. RNA-sequencing (RNA-seq) analysis was also used to explore the CMTM4-regulated genes in LoVo-shCMTM4 cells compared with control cells. RT-qPCR was then used to confirm the expression of these CMTM4-regulated genes. CMTM4 expression in pre-nCRT tissues indicated an unfavorable response and a short disease-free survival (DFS) with LARC. The expression of CMTM4 significantly increased following nCRT treatment. Additionally, CMTM4 knockdown increased the proliferation, migration and invasion of colon cancer cells; however, IR disrupted the cell migration and invasion induced by CMTM4 knockdown. RNA-seq analysis, the Tumor Immune Estimation Resource database and RT-qPCR indicated that CMTM4 was involved in different signaling pathways and regulated immune-related genes such as cluster of differentiation 66b, chemokine (CXC motif) ligand 8 (CXCL8) and programmed cell death 1. Furthermore, CXCL8 expression was found to be negatively associated with CMTM4 expression in patients with LARC by IHC and RT-qPCR. CXCL8 expression on invasion margin regions in post-operative tissues was also an inferior predictor of DFS in patients with LARC. In conclusion, CMTM4 may predict the nCRT response and outcomes in patients with LARC.
Collapse
Affiliation(s)
- Lujing Yang
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Zhiting Miao
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Ningning Li
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Lin Meng
- Department of Biochemistry and Molecular Biology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Qin Feng
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Dongbo Qiao
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Ping Wang
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Yue Wang
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Yanhua Bai
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Zhongwu Li
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Shenyi Lian
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| |
Collapse
|
3
|
Liu Q, Wang J, Guo Z, Zhang H, Zhou Y, Wang P, Li T, Lu W, Liu F, Han W. CMTM6 promotes hepatocellular carcinoma progression through stabilizing β-catenin. Cancer Lett 2024; 583:216585. [PMID: 38101607 DOI: 10.1016/j.canlet.2023.216585] [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: 08/04/2023] [Revised: 11/10/2023] [Accepted: 12/04/2023] [Indexed: 12/17/2023]
Abstract
CMTM6, a regulator of PD-L1 stability, has been implicated in the development of various cancers. However, the expression and role of CMTM6 in hepatocellular carcinoma (HCC) remains controversial. Our study revealed a negative correlation between CMTM6 expression and HCC prognosis through bioinformatics analysis and immunofluorescence staining. CMTM6 expression was also positively associated with alpha-fetoprotein (AFP) levels, supporting its potential as a prognostic marker for HCC. Using Cmtm6 knockout mice, we found that Cmtm6 deficiency inhibited HCC formation and cell proliferation in primary liver cancer models induced by DEN and DEN/CCl4. In HCC cell lines, CMTM6 promoted cell proliferation and interacted with β-catenin, stabilizing it by preventing ubiquitination. In conclusion, our study suggested that CMTM6 upregulation promotes HCC cell proliferation through the β-catenin pathway, making it a potential therapeutic target for HCC treatment.
Collapse
Affiliation(s)
- Qiyao Liu
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China, Beijing, China
| | - Jiahui Wang
- Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Shandong, China
| | - Zixia Guo
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China, Beijing, China
| | - Hanxiao Zhang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China, Beijing, China
| | - Yifan Zhou
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
| | - Pingzhang Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China, Beijing, China
| | - Ting Li
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China, Beijing, China
| | - Wenping Lu
- Faculty of Hepato-Pancreato-Biliary Surgery, First Medical Center, Chinese PLA General Hospital, China.
| | - Fujun Liu
- Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Shandong, China.
| | - Wenling Han
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China, Beijing, China.
| |
Collapse
|
4
|
Duan SL, Jiang Y, Li GQ, Fu W, Song Z, Li LN, Li J. Research insights into the chemokine-like factor (CKLF)-like MARVEL transmembrane domain-containing family (CMTM): their roles in various tumors. PeerJ 2024; 12:e16757. [PMID: 38223763 PMCID: PMC10787544 DOI: 10.7717/peerj.16757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 12/13/2023] [Indexed: 01/16/2024] Open
Abstract
The chemokine-like factor (CKLF)-like MARVEL transmembrane domain-containing (CMTM) family includes CMTM1-8 and CKLF, and they play key roles in the hematopoietic, immune, cardiovascular, and male reproductive systems, participating in the physiological functions, cancer, and other diseases associated with these systems. CMTM family members activate and chemoattract immune cells to affect the proliferation and invasion of tumor cells through a similar mechanism, the structural characteristics typical of chemokines and transmembrane 4 superfamily (TM4SF). In this review, we discuss each CMTM family member's chromosomal location, involved signaling pathways, expression patterns, and potential roles, and mechanisms of action in pancreatic, breast, gastric and liver cancers. Furthermore, we discuss several clinically applied tumor therapies targeted at the CMTM family, indicating that CMTM family members could be novel immune checkpoints and potential targets effective in tumor treatment.
Collapse
Affiliation(s)
- Sai-Li Duan
- Department of General Surgery, Xiangya Hospital Central South University, Changsha Province, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha Province, Hunan, China
| | - Yingke Jiang
- Department of General Surgery, Xiangya Hospital Central South University, Changsha Province, Hunan, China
| | - Guo-Qing Li
- Xiangya School of Medicine, Central South University, Changsha Province, Hunan, China
| | - Weijie Fu
- Xiangya School of Medicine, Central South University, Changsha Province, Hunan, China
| | - Zewen Song
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha Province, Hunan, China
| | - Li-Nan Li
- Department of Oncology, The 1st Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jia Li
- Department of Oncology, The 1st Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
5
|
Pei Y, Zhang Z, Tan S. Current Opinions on the Relationship Between CMTM Family and Hepatocellular Carcinoma. J Hepatocell Carcinoma 2023; 10:1411-1422. [PMID: 37649636 PMCID: PMC10464892 DOI: 10.2147/jhc.s417202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 08/12/2023] [Indexed: 09/01/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a typically malignant tumor in the digestive system. The mortality of HCC ranks third place in the world, second only to lung cancer and colorectal cancer. For the characteristics of high invasiveness, high metastasis, high recurrence rate as well as short survival time, HCC treatment has always been difficult in clinical practice. Many causes have contributed to the appearance of these features, including insidious onset, high degree of malignancy, lack of effective early molecular diagnostic markers, and disease prediction models. The human chemokine-like factor superfamily (CMTMs) is a new gene family consisting of CKLF and CMTM1-CMTM8. CMTMs have a marvel domain which can activate and chemotaxis immune cells. Many studies have reported that CMTMs are involved in the regulation of cell growth and development, and play an important role in the malignant progression of the immune system and reproductive system, especially in the development of tumors. In this review, we summarized the structure and function of the human CMTMs, the relationship between its family members and HCC, the prognostic value, potential functions, and mechanisms in HCC. CMTMs could provide a new diagnostic and therapeutic target in clinical practice for patients with HCC.
Collapse
Affiliation(s)
- Yulin Pei
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, Guangxi, People’s Republic of China
- Public Health Department of Guilin Medical University, Guilin, Guangxi, People’s Republic of China
| | - Zhengbao Zhang
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, Guangxi, People’s Republic of China
- Public Health Department of Guilin Medical University, Guilin, Guangxi, People’s Republic of China
| | - Shengkui Tan
- Public Health Department of Youjiang Medical University For Nationalities, Baise, GuangxiPeople's Republic of China
| |
Collapse
|
6
|
Wang P, Wang Y, Wang Y. Ferroptosis patterns modulate immunocyte communication in tumor microenvironments: clinical value and therapeutic guidance of lung adenocarcinoma. Funct Integr Genomics 2023; 23:181. [PMID: 37231311 DOI: 10.1007/s10142-023-01100-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/12/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023]
Abstract
Lung adenocarcinoma (LUAD) emerges as one of the most aggressive tumor types with a poor prognosis. As a novel form of regulated cell death, ferroptosis promotes the clearance of tumor cells. However, few studies demonstrated whether ferroptosis-related genes can modify the behavior of tumor microenvironment (TME) cells. Resorting to non-negative matrix factorization (NMF) clustering based on the expression of ferroptosis-related genes, we identified multiple LUAD TME cell-type subpopulations. These subtypes of TME cells displayed extensive communication with tumor epithelial cells. ATF3+cancer-associated fibroblasts (CAFs), SLC40A1+CD8+T cells, and ALOX5+CD8+T cells showed distinct biological features compared to non-ferroptosis-related TME cells. Patients with a higher abundance of these ferroptosis-related TME cell subtypes showed a favorable clinical outcome. Our study depicted a detailed landscape of LUAD cell composition with a focus on ferroptosis-related genes, which, hopefully, may provide novel insight into further study of the LAUD immune microenvironment.
Collapse
Affiliation(s)
- Peng Wang
- Department of Oncology, First Hospital Affiliated to Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Ye Wang
- Department of Endocrinology, First Hospital Affiliated to Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yu Wang
- Physical Diagnostics Section, First Hospital Affiliated to Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| |
Collapse
|
7
|
Jia XM, Long YR, Yu XL, Chen RQ, Gong LK, Geng Y. Construction of stable membranal CMTM6-PD-L1 full-length complex to evaluate the PD-1/PD-L1-CMTM6 interaction and develop anti-tumor anti-CMTM6 nanobody. Acta Pharmacol Sin 2023; 44:1095-1104. [PMID: 36418428 PMCID: PMC10104848 DOI: 10.1038/s41401-022-01020-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/23/2022] [Indexed: 11/24/2022]
Abstract
CKLF (chemokine-like factor)-MARVEL transmembrane domain containing protein 6 (CMTM6) is a novel regulator to maintain the stability of PD-L1. CMTM6 can colocalize and interact with PD-L1 on the recycling endosomes and cell membrane, preventing PD-L1 from lysosome-mediated degradation and proteasome-mediated degradation thus increasing the half-life of PD-L1 on the cell membrane. The difficulties in obtaining stable full-length PD-L1 and CMTM6 proteins hinder the research on their structures, function as well as related drug development. Using lauryl maltose neopentyl glycol (LMNG) as the optimized detergent and a cell membrane mimetic strategy, we assembled a stable membrane-bound full-length CMTM6-PD-L1 complex with amphipol A8-35. When the PD-1/PD-L1-CMTM6 interactions were analyzed, we found that CMTM6 greatly enhanced the binding and delayed the dissociation of PD-1/PD-L1, thus affecting immunosuppressive signaling and anti-apoptotic signaling. We then used the CMTM6-PD-L1 complex as immunogens to generate immune repertoires in camels, and identified a functional anti-CMTM6 nanobody, called 1A5. We demonstrated that the anti-CMTM6 nanobody greatly decreased T-cell immunosuppression and promoted apoptotic susceptibility of tumor cells in vitro, and mainly relied on the cytotoxic effect of CD8+ T-cells to exert tumor growth inhibitory effects in CT26 tumor-bearing mice. In conclusion, the stable membrane-bound full-length CMTM6-PD-L1 complex has been successfully used in studying PD-1/PD-L1-CMTM6 interactions and CMTM6-targeting drug development, suggesting CMTM6 as a novel tumor immunotherapy target.
Collapse
Affiliation(s)
- Xiao-Min Jia
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yi-Ru Long
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiao-Lu Yu
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Run-Qiu Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Li-Kun Gong
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
| | - Yong Geng
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
8
|
Long Y, Chen R, Yu X, Tong Y, Peng X, Li F, Hu C, Sun J, Gong L. Suppression of Tumor or Host Intrinsic CMTM6 Drives Antitumor Cytotoxicity in a PD-L1-Independent Manner. Cancer Immunol Res 2023; 11:241-260. [PMID: 36484740 PMCID: PMC9896022 DOI: 10.1158/2326-6066.cir-22-0439] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/03/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022]
Abstract
CKLF-like MARVEL transmembrane domain-containing protein 6 (CMTM6) is known to be a regulator of membranal programmed death ligand 1 (PD-L1) stability and a factor associated with malignancy progression, but the effects and mechanisms of CMTM6 on tumor growth, as well as its potential as a target for therapy, are still largely unknown. Here, we show that CMTM6 expression increased with tumor progression in both patients and mice. Ablation of CMTM6 significantly reduced human and murine tumor growth in a manner dependent on T-cell immunity. Tumor CMTM6 suppression broke resistance to immune-checkpoint inhibitors and remodeled the tumor immune microenvironment, as specific antitumor cytotoxicity was enhanced and contributed primarily to tumor inhibition. Without the PD-1/PD-L1 axis, CMTM6 suppression still significantly dampened tumor growth dependent on cytotoxic cells. Furthermore, we identified that CMTM6 was widely expressed on immune cells. T-cell CMTM6 levels increased with sustained immune activation and intratumoral immune exhaustion and affected T cell-intrinsic PD-L1 levels. Host CMTM6 knockout significantly restrained tumor growth in a manner dependent on CD8+ T cells and not entirely dependent on PD-L1. Thus, we developed and evaluated the antitumor efficacy of CMTM6-targeting adeno-associated virus (AAV), which effectively mobilized antitumor immunity and could be combined with various antitumor drugs. Our findings reveal that both tumor and host CMTM6 are involved in antitumor immunity with or without the PD-1/PD-L1 axis and that gene therapy targeting CMTM6 is a promising strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Yiru Long
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Runqiu Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaolu Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yongliang Tong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xionghua Peng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Fanglin Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chao Hu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Jianhua Sun
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Likun Gong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
| |
Collapse
|
9
|
Meng H, Li S, Li Q, Wang Y, Wang G, Qu Y. Chemokine-like factor-like MARVEL transmembrane domain containing 6: Bioinformatics and experiments in vitro analyze in glioblastoma multiforme. Front Mol Neurosci 2023; 15:1026927. [PMID: 36698778 PMCID: PMC9869805 DOI: 10.3389/fnmol.2022.1026927] [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: 08/24/2022] [Accepted: 12/16/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction Chemokine-like factor (CKLF)-like MARVEL transmembrane domain containing 6 (CMTM6) is a protein localized to the cell membrane and is known for its ability to co-localize with PD-L1 on the plasma membrane, prevent PD-L1 degradation, and maintain PD-L1 expression on the cell membrane. CMTM6 is highly expressed and plays an important role in various tumors such as oral squamous cell carcinoma (OSCC) and colorectal cancer (CRC), however, its role in Glioblastoma multiforme (GBM) is unclear. Methods In this paper, to investigate the role of CMTM6 in GBM, we analyzed the expression of CMTM6 in GBM, the interaction with CMTM6 and the associated genes by bioinformatics. Importantly, we analyzed the expression of CMTM6 in GBM in relation to tumor-infiltrating lymphocytes (TILs), immunoinhibitors, immunostimulators, chemokines and chemokine receptors. We further analyzed the function of CMTM6 and performed in vitro experiments to verify it. Finally, the sensitivity of CMTM6 to drugs was also analyzed and the relationship between CMTM6 and the anticancer drug Piperlonguminine (PL) was verified in vitro. Results The results showed that CMTM6 was highly expressed in GBM and correlated with multiple genes. Furthermore, CMTM6 is closely related to the immune microenvironment and inflammatory response in GBM. Bioinformatic analysis of CMTM6 correlated with the function of GBM, and our experiments demonstrated that CMTM6 significantly promoted the migration of GBM cells and epithelial-mesenchymal transition (EMT), but had no significant effect on other functions. Interestingly, we found that in GBM, PL promotes the expression of CMTM6. Discussion In this paper, we have performed a detailed analysis and validation of the role of CMTM6 in GBM using bioinformatics analysis and in vitro experiments to demonstrate that CMTM6 may be a potential target for glioma therapy.
Collapse
Affiliation(s)
- Haining Meng
- Department of Emergency Medicine, Medical College of Qingdao University, Qingdao, Shandong, China,Department of Intensive Care Unit, Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shaohua Li
- Department of Laboratory Medicine, The Third People’s Hospital of Qingdao, Qingdao, Shandong, China
| | - Qingshu Li
- Department of Intensive Care Unit, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Yuqin Wang
- Department of Emergency, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Guoan Wang
- Qingdao Municipal Hospital, Qingdao, Shandong, China,*Correspondence: Guoan Wang, ✉
| | - Yan Qu
- Department of Intensive Care Unit, Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, Shandong, China,Yan Qu, ✉
| |
Collapse
|
10
|
Zhang T, Yu H, Dai X, Zhang X. CMTM6 and CMTM4 as two novel regulators of PD-L1 modulate the tumor microenvironment. Front Immunol 2022; 13:971428. [PMID: 35958549 PMCID: PMC9359082 DOI: 10.3389/fimmu.2022.971428] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
The tumor microenvironment (TME) plays crucial roles in regulating tumor occurrence, progress, metastasis and drug resistance. However, it remains largely elusive how the components of TME are regulated to govern its functions in tumor biology. Here, we discussed how the two novel functional proteins, chemokine-like factor (CKLF)-like MARVEL transmembrane domain-containing 6 (CMTM6) and CMTM4, which involved in the post-translational regulation of PD-L1, modulate the TME functions. The roles of CMTM6 and CMTM4 in regulating TME components, including immune cells and tumor cells themselves were discussed in this review. The potential clinical applications of CMTM6 and CMTM4 as biomarkers to predict therapy efficacy and as new or combined immunotherapy targets are also highlighted. Finally, the current hot topics for the biological function of CMTM6/4 and several significant research directions for CMTM6/4 are also briefly summarized in the review.
Collapse
Affiliation(s)
- Tong Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
| | - Haixiang Yu
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiangpeng Dai
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
- *Correspondence: Xiangpeng Dai, ; Xiaoling Zhang,
| | - Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
- *Correspondence: Xiangpeng Dai, ; Xiaoling Zhang,
| |
Collapse
|
11
|
Hoft SG, Pherson MD, DiPaolo RJ. Discovering Immune-Mediated Mechanisms of Gastric Carcinogenesis Through Single-Cell RNA Sequencing. Front Immunol 2022; 13:902017. [PMID: 35757757 PMCID: PMC9231461 DOI: 10.3389/fimmu.2022.902017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/27/2022] [Indexed: 12/17/2022] Open
Abstract
Single-cell RNA sequencing (scRNAseq) technology is still relatively new in the field of gastric cancer immunology but gaining significant traction. This technology now provides unprecedented insights into the intratumoral and intertumoral heterogeneities at the immunological, cellular, and molecular levels. Within the last few years, a volume of publications reported the usefulness of scRNAseq technology in identifying thus far elusive immunological mechanisms that may promote and impede gastric cancer development. These studies analyzed datasets generated from primary human gastric cancer tissues, metastatic ascites fluid from gastric cancer patients, and laboratory-generated data from in vitro and in vivo models of gastric diseases. In this review, we overview the exciting findings from scRNAseq datasets that uncovered the role of critical immune cells, including T cells, B cells, myeloid cells, mast cells, ILC2s, and other inflammatory stromal cells, like fibroblasts and endothelial cells. In addition, we also provide a synopsis of the initial scRNAseq findings on the interesting epithelial cell responses to inflammation. In summary, these new studies have implicated roles for T and B cells and subsets like NKT cells in tumor development and progression. The current studies identified diverse subsets of macrophages and mast cells in the tumor microenvironment, however, additional studies to determine their roles in promoting cancer growth are needed. Some groups specifically focus on the less prevalent ILC2 cell type that may contribute to early cancer development. ScRNAseq analysis also reveals that stromal cells, e.g., fibroblasts and endothelial cells, regulate inflammation and promote metastasis, making them key targets for future investigations. While evaluating the outcomes, we also highlight the gaps in the current findings and provide an assessment of what this technology holds for gastric cancer research in the coming years. With scRNAseq technology expanding rapidly, we stress the need for periodic review of the findings and assess the available scRNAseq analytical tools to guide future work on immunological mechanisms of gastric carcinogenesis.
Collapse
Affiliation(s)
- Stella G Hoft
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Michelle D Pherson
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, United States.,Genomics Core Facility, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Richard J DiPaolo
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, United States
| |
Collapse
|