1
|
Nakamura K, Ida N, Hirasawa A, Okamoto K, Vu TH, Hai Ly DT, Masuyama H. CD63 as a potential biomarker for patients with ovarian cancer. Eur J Obstet Gynecol Reprod Biol 2025; 306:87-93. [PMID: 39799740 DOI: 10.1016/j.ejogrb.2025.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/12/2024] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
INTRODUCTION Exosomes play an important role in regulating physiological processes and mediating the systemic dissemination of various types of cancer. We investigated the association of exosomal tetraspanins CD9, CD63, and CD81 in patients with ovarian cancer (OC). MATERIAL AND METHODS We measured the plasma tetraspanins CD9, CD63, and CD81 by enzyme-linked immunosorbent assay in 91 patients who underwent treatment for OC between April 2018 and March 2024. Additionally, we analyzed clinical pathologic factors, chemotherapy response, and prognosis. RESULTS In terms of stages, CD63 expression was significantly higher in patients with stage IV compared to those with stage I OC (p = 0.003). In terms of histological type, CD63 expression was significantly higher in high-grade serous carcinoma (HGSC) than in clear cell carcinoma (CCC) with OC (p = 0.009). Furthermore, CD63 levels were significantly higher in advanced-stage, HGSC than in patients with early-stage, non-HGSC and early-stage, HGSC OC (p = 0.045 and p = 0.002, respectively). In the Neoadjuvant chemotherapy (NAC) of 12 patients with OC assessed as having either a partial response (PR) or complete response (CR), CD63 was significantly decreased (p = 0.043), whereas perforin was significantly increased (p = 0.001). In the NAC of 16 patients with OC, CD63 of the response rate to chemotherapy tended to differ between the progressive disease (PD) and PR/CR groups (p = 0.056). A moderate inverse correlation was observed between CD63 and perforin levels (R = 0.638, R2 = 0.428, p = 0.008). CONCLUSIONS CD63 could be a potential biomarker for all types of OC patients.
Collapse
Affiliation(s)
- Keiichiro Nakamura
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho kitaku, Okayama 700-8558, Japan.
| | - Naoyuki Ida
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho kitaku, Okayama 700-8558, Japan
| | - Akira Hirasawa
- Department of Clinical Genomic Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho kitaku, Okayama 700-8558, Japan.
| | - Kazuhiro Okamoto
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho kitaku, Okayama 700-8558, Japan
| | - Thuy Ha Vu
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho kitaku, Okayama 700-8558, Japan
| | - Dao Thi Hai Ly
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho kitaku, Okayama 700-8558, Japan.
| | - Hisashi Masuyama
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho kitaku, Okayama 700-8558, Japan.
| |
Collapse
|
2
|
Duke LC, Cone AS, Sun L, Dittmer DP, Meckes DG, Tomko RJ. Tetraspanin CD9 alters cellular trafficking and endocytosis of tetraspanin CD63, affecting CD63 packaging into small extracellular vesicles. J Biol Chem 2025; 301:108255. [PMID: 39909378 PMCID: PMC11919600 DOI: 10.1016/j.jbc.2025.108255] [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: 10/18/2024] [Revised: 01/13/2025] [Accepted: 01/27/2025] [Indexed: 02/07/2025] Open
Abstract
Small extracellular vesicles (sEVs) are particles secreted from cells that play vital roles both in normal physiology and in human disease. sEVs are highly enriched in tetraspanin proteins, such as CD9 and CD63, and contain tetraspanin-enriched membrane microdomains involved in loading of sEVs with macromolecule cargoes and in sEV biogenesis. However, the precise roles of individual tetraspanins in sEV biogenesis and cargo loading remain poorly understood. Here, we report that CD9 negatively regulated CD63 trafficking to tetraspanin-enriched microdomains and its subsequent packaging into sEVs, whereas CD63 had no discernable effect on CD9 localization or packaging. Using super resolution microscopy of individual vesicles, we showed that CD9 governs the fraction of sEVs that are loaded with CD63. Interestingly, CD9-dependent suppression of CD63 packaging was rescued by pharmacological blockade of endocytosis. Together, our data support a model where CD9 contributes to the regulation and secretion of CD63 in an endocytosis-dependent manner to reprogram the contents of sEVs and tetraspanin-enriched microdomains.
Collapse
Affiliation(s)
- Leanne C Duke
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA.
| | - Allaura S Cone
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Li Sun
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA
| | - Dirk P Dittmer
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Robert J Tomko
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA
| |
Collapse
|
3
|
Shao S, Bu Z, Xiang J, Liu J, Tan R, Sun H, Hu Y, Wang Y. The role of Tetraspanins in digestive system tumor development: update and emerging evidence. Front Cell Dev Biol 2024; 12:1343894. [PMID: 38389703 PMCID: PMC10882080 DOI: 10.3389/fcell.2024.1343894] [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: 11/24/2023] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
Digestive system malignancies, including cancers of the esophagus, pancreas, stomach, liver, and colorectum, are the leading causes of cancer-related deaths worldwide due to their high morbidity and poor prognosis. The lack of effective early diagnosis methods is a significant factor contributing to the poor prognosis for these malignancies. Tetraspanins (Tspans) are a superfamily of 4-transmembrane proteins (TM4SF), classified as low-molecular-weight glycoproteins, with 33 Tspan family members identified in humans to date. They interact with other membrane proteins or TM4SF members to form a functional platform on the cytoplasmic membrane called Tspan-enriched microdomain and serve multiple functions including cell adhesion, migration, propagation and signal transduction. In this review, we summarize the various roles of Tspans in the progression of digestive system tumors and the underlying molecular mechanisms in recent years. Generally, the expression of CD9, CD151, Tspan1, Tspan5, Tspan8, Tspan12, Tspan15, and Tspan31 are upregulated, facilitating the migration and invasion of digestive system cancer cells. Conversely, Tspan7, CD82, CD63, Tspan7, and Tspan9 are downregulated, suppressing digestive system tumor cell metastasis. Furthermore, the connection between Tspans and the metastasis of malignant bone tumors is reviewed. We also summarize the potential role of Tspans as novel immunotherapy targets and as an approach to overcome drug resistance. Finally, we discuss the potential clinical value and therapeutic targets of Tspans in the treatments of digestive system malignancies and provide some guidance for future research.
Collapse
Affiliation(s)
- Shijie Shao
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Zhen Bu
- Department of General Surgery, Xinyi People's Hospital, Xinyi, China
| | - Jinghua Xiang
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jiachen Liu
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Rui Tan
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Han Sun
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yuanwen Hu
- Department of Gastroenterology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Yimin Wang
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
4
|
P C S, Shetty SS, Kumari N S, Shetty VV, Shetty P, Rao C, Shetty PK. Prognostic significance of tetraspanin CD9 and oncogenic epidermal growth factor receptor in tongue squamous cell carcinoma survival. Pathol Res Pract 2023; 248:154651. [PMID: 37390757 DOI: 10.1016/j.prp.2023.154651] [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: 05/29/2023] [Revised: 06/23/2023] [Accepted: 06/25/2023] [Indexed: 07/02/2023]
Abstract
The most prevalent locations for head and neck cancer is the tongue. The surviving patients who are receiving therapy have considerably compromised speech, taste, chewing, and swallowing. CD9 is a cell surface protein that has contradictory role in cancer progression. The objective of the study is to analyze the Cluster of Differentiation 9(CD9), Epidermal Growth Factor Receptor (EGFR) and Phosphorylated Akt (p-Akt) expression in tongue cancer specimens and its clinical significance.50 tongue cancer sections were used to analyze the expression of CD9,EGFR and p-Akt by immunohistochemistry. Data regarding the histological grade of the tumor, age, sex, and habits were recorded, and relation with CD9,EGFR and p-Akt expression was assessed. Data were expressed as mean ± SEM. Categorical data was analyzed by Chi-square test. Student t-test was used to check the significance of data between two groups.A significant increase in the CD9,EGFR and p-Akt expression (1.8 ± 0.11, 2.06 ± 0.18 and 2.3 ± 0.15 respectively) was seen in the tongue cancer specimens. CD9 and p-Akt expression had a significant association with the histological grade (p < 0.004 and p < 0.006 respectively). CD9 expression was higher in patients with the combination of addiction/habit compared to patients with single addictions(1.08 ± 0.11 and 0.75 ± 0.47). Overall a poor rate of survival was observed in CD9 positive patients(p < 0.039). EGFR and p-Akt expression increased with increasing expression of CD9, suggesting its use as a biomarker to track the development of TSCC.
Collapse
Affiliation(s)
- Suhasini P C
- Central Research Laboratory, KS Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangalore, India.
| | - Shilpa S Shetty
- Central Research Laboratory, KS Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangalore, India.
| | - Suchetha Kumari N
- Central Research Laboratory, KS Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangalore, India.
| | - Vijith Vittal Shetty
- Department of Oncology, KS Hegde Medical Academy, Deralakatte, Mangalore, Karnataka, India.
| | - Pushparaj Shetty
- Department of Oral and Maxillofacial Pathology and Microbiology, AB Shetty Memorial Institute of Dental Sciences,Nitte (Deemed to be University), Deralakatte, Mangalore, India.
| | - Chandrika Rao
- Department of Pathology, KS Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangalore, India.
| | - Praveen Kumar Shetty
- Department of Biochemistry, KS Hegde Medical Academy, Nitte (Deemed to be University), India.
| |
Collapse
|
5
|
Zhou Z, Yang Z, Zhou L, Yang M, He S. The versatile roles of testrapanins in cancer from intracellular signaling to cell-cell communication: cell membrane proteins without ligands. Cell Biosci 2023; 13:59. [PMID: 36941633 PMCID: PMC10025802 DOI: 10.1186/s13578-023-00995-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/21/2023] [Indexed: 03/23/2023] Open
Abstract
The tetraspanins (TSPANs) are a family of four-transmembrane proteins with 33 members in mammals. They are variably expressed on the cell surface, various intracellular organelles and vesicles in nearly all cell types. Different from the majority of cell membrane proteins, TSPANs do not have natural ligands. TSPANs typically organize laterally with other membrane proteins to form tetraspanin-enriched microdomains (TEMs) to influence cell adhesion, migration, invasion, survival and induce downstream signaling. Emerging evidence shows that TSPANs can regulate not only cancer cell growth, metastasis, stemness, drug resistance, but also biogenesis of extracellular vesicles (exosomes and migrasomes), and immunomicroenvironment. This review summarizes recent studies that have shown the versatile function of TSPANs in cancer development and progression, or the molecular mechanism of TSPANs. These findings support the potential of TSPANs as novel therapeutic targets against cancer.
Collapse
Affiliation(s)
- Zhihang Zhou
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, SAR, People's Republic of China.
| | - Zihan Yang
- Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, SAR, People's Republic of China
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong, China
| | - Li Zhou
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, SAR, People's Republic of China
| | - Mengsu Yang
- Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, SAR, People's Republic of China
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong, China
| | - Song He
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
6
|
Targeting receptor tyrosine kinases in ovarian cancer: Genomic dysregulation, clinical evaluation of inhibitors, and potential for combinatorial therapies. Mol Ther Oncolytics 2023; 28:293-306. [PMID: 36911068 PMCID: PMC9999170 DOI: 10.1016/j.omto.2023.02.006] [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] [Indexed: 02/22/2023] Open
Abstract
Epithelial ovarian cancer (EOC) remains one of the leading causes of cancer-related deaths among women worldwide. Receptor tyrosine kinases (RTKs) have long been sought as therapeutic targets for EOC, as they are frequently hyperactivated in primary tumors and drive disease relapse, progression, and metastasis. More recently, these oncogenic drivers have been implicated in EOC response to poly(ADP-ribose) polymerase (PARP) inhibitors and epigenome-interfering agents. This evidence revives RTKs as promising targets for therapeutic intervention of EOC. This review summarizes recent studies on the role of RTKs in EOC malignancy and the use of their inhibitors for clinical treatment. Our focus is on the ERBB family, c-Met, and VEGFR, as they are linked to drug resistance and targetable using commercially available drugs. The importance of these RTKs and their inhibitors is highlighted by their impact on signal transduction and intratumoral heterogeneity in EOC and successful use as maintenance therapy in the clinic through suppression of the VEGF/VEGFR axis. Finally, the therapeutic potential of RTK inhibitors is discussed in the context of combinatorial targeting via co-inhibiting proliferative and anti-apoptotic pathways, epigenomic/transcriptional programs, and harnessing the efficacy of PARP inhibitors and programmed cell death 1/ligand 1 immune checkpoint therapies.
Collapse
|
7
|
Huang Y, Zhao X, Zhang Q, Yang X, Hou G, Peng C, Jia M, Zhou L, Yamamoto T, Zheng J. Novel therapeutic perspectives for crescentic glomerulonephritis through targeting parietal epithelial cell activation and proliferation. Expert Opin Ther Targets 2023; 27:55-69. [PMID: 36738160 DOI: 10.1080/14728222.2023.2177534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Kidney injury is clinically classified as crescentic glomerulonephritis (CrGN) when ≥50% of the glomeruli in a biopsy sample contain crescentic lesions. However, current strategies, such as systemic immunosuppressive therapy and plasmapheresis for CrGN, are partially effective, and these drugs have considerable systemic side effects. Hence, targeted therapy to prevent glomerular crescent formation and expansion remains an unmet clinical need. AREAS COVERED Hyperproliferative parietal epithelial cells (PECs) are the main constituent cells of the glomerular crescent with cell-tracing evidence. Crescents obstruct the flow of primary urine, pressure the capillaries, and degenerate the affected nephrons. We reviewed the markers of PEC activation and proliferation, potential therapeutic effects of thrombin and thrombin receptor inhibitors, and how podocytes cross-talk with PECs. These experiments may help identify potential early specific targets for the prevention and treatment of glomerular crescentic injury. EXPERT OPINION Inhibiting PEC activation and proliferation in CrGN can alleviate glomerular crescent progression, which has been supported by preclinical studies with evidence of genetic deletion. Clarifying the outcome of PEC transformation to the podocyte phenotype and suppressing thrombin, thrombin receptors, and PEC hyperproliferation in early therapeutic strategies will be the research goals in the next ten years.
Collapse
Affiliation(s)
- Yanjie Huang
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China.,Department of Pediatrics, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xueru Zhao
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Qiushuang Zhang
- Department of Pediatrics, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xiaoqing Yang
- Department of Pediatrics, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Gailing Hou
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Chaoqun Peng
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Mengzhen Jia
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Li Zhou
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Tatsuo Yamamoto
- Department of Nephrology, Fujieda Municipal General Hospital, 4-1-11 Surugadai, Fujieda, Japan
| | - Jian Zheng
- Institute of Pediatrics of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| |
Collapse
|
8
|
P C S, Shetty SS, Nalilu SK, Shetty PK, Patil P. Tetraspanin CD9: A friend or foe of head and neck cancer (Review). Oncol Rep 2022; 47:88. [PMID: 35266009 PMCID: PMC8931833 DOI: 10.3892/or.2022.8299] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/15/2021] [Indexed: 12/02/2022] Open
Abstract
Head and neck cancers are diverse and complex diseases characterised by unregulated growth of tumour cells in various parts of the head and neck region, such as in the buccal mucosa, floor of the mouth, tongue, oropharynx, hypopharynx, oesophagus, nasopharynx and salivary glands. Partial or total glossectomy, radiation or chemotherapy greatly affect patient quality of life. However, even following treatment, patients may relapse. Nicotine-derived nitrosamines and alcohol are the major etiological factors underlying this deadly disease. These compounds induce DNA damage that may lead to mutation in crucial genes, such as p53 and p21, which are important to regulate cell proliferation, thus leading to cancer. CD9 is a tetraspanin, which are a group of transmembrane proteins that have a role in cell motility and adhesion. The present review aimed to explore the role of CD9 in head and neck cancer. Epidermal growth factor receptor activity and cell proliferation are regulated by the CD9-integrin/CD9-transforming growth factor interaction. Hence, CD9 can play a dual role in various types of cancer.
Collapse
Affiliation(s)
- Suhasini P C
- Central Research Laboratory, K.S. Hegde Medical Academy, Nitte (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Shilpa S Shetty
- Central Research Laboratory, K.S. Hegde Medical Academy, Nitte (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Suchetha Kumari Nalilu
- Department of Biochemistry, K.S. Hegde Medical Academy, Nitte (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Praveen Kumar Shetty
- Department of Biochemistry, K.S. Hegde Medical Academy, Nitte (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Prakash Patil
- Central Research Laboratory, K.S. Hegde Medical Academy, Nitte (Deemed to be University), Mangalore, Karnataka 575018, India
| |
Collapse
|
9
|
Saad MA, Xavierselvan M, Sharif HA, Selfridge S, Pawle R, Varvares M, Mallidi S, Hasan T. Dual Function Antibody Conjugates for Multimodal Imaging and Photoimmunotherapy of Cancer Cells. Photochem Photobiol 2022; 98:220-231. [PMID: 34379796 PMCID: PMC10038131 DOI: 10.1111/php.13501] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/08/2021] [Indexed: 11/29/2022]
Abstract
Precision imaging, utilizing molecular targeted agents, is an important tool in cancer diagnostics and guiding therapies. While there are limitations associated with single mode imaging probes, multimodal molecular imaging probes enabling target visualization through complementary imaging technologies provides an attractive alternative. However, there are several challenges associated with designing molecular probes carrying contrast agents for complementary multimodal imaging. Here, we propose a dual function antibody conjugate (DFAC) comprising an FDA approved photosensitizer Benzoporphyrin derivative (BPD) and a naphthalocyanine-based photoacoustic dye (SiNc(OH)) for multimodal infrared (IR) imaging. While fluorescence imaging, through BPD, provides sensitivity, complementing it with photoacoustic imaging, through SiNc(OH), provides a depth-resolved spatial resolution much beyond the optical diffusion limits of fluorescence measurements. Through a series of in vitro experiments, we demonstrate the development and utilization of DFACs for multimodal imaging and photodynamic treatment of squamous cell carcinoma (A431) cell line. The proposed DFACs have potential use in precision imaging applications such as guiding tumor resection surgeries and photodynamic treatment of residual microscopic disease thereby minimizing local recurrence. The data demonstrated in this study merits further investigation for its preclinical and clinical translation.
Collapse
Affiliation(s)
- Mohammad A. Saad
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Marvin Xavierselvan
- Department of Biomedical Engineering, Science and Technology Center, Tufts University, Medford, MA
| | | | | | | | - Mark Varvares
- Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, The Massachusetts Eye and Ear, Boston, MA
| | - Srivalleesha Mallidi
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Department of Biomedical Engineering, Science and Technology Center, Tufts University, Medford, MA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA
| |
Collapse
|
10
|
Garcia-Mayea Y, Mir C, Carballo L, Sánchez-García A, Bataller M, LLeonart ME. TSPAN1, a novel tetraspanin member highly involved in carcinogenesis and chemoresistance. Biochim Biophys Acta Rev Cancer 2021; 1877:188674. [PMID: 34979155 DOI: 10.1016/j.bbcan.2021.188674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 12/11/2022]
Abstract
The tetraspanin (TSPAN) family constitutes a poorly explored family of membrane receptors involved in various physiological processes, with relevant roles in anchoring multiple proteins, acting as scaffolding proteins, and cell signaling. Recent studies have increasingly demonstrated the involvement of TSPANs in cancer. In particular, tetraspanin 1 (also known as TSPAN1, NET-1, TM4C, C4.8 or GEF) has been implicated in cell survival, proliferation and invasion. Recently, our laboratory revealed a key role of TSPAN1 in the acquired resistance of tumor cells to conventional chemotherapy (e.g., cisplatin). In this review, we summarize and discuss the latest research on the physiological mechanisms of TSPANs in cancer and, in particular, on TSPAN1 regulating resistance to chemotherapy. A model of TSPAN1 action is proposed, and the potential of targeting TSPAN1 in anticancer therapeutic strategies is discussed.
Collapse
Affiliation(s)
- Yoelsis Garcia-Mayea
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Cristina Mir
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Laia Carballo
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Almudena Sánchez-García
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Marina Bataller
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Matilde E LLeonart
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain; Spanish Biomedical Research Network Center in Oncology, CIBERONC, Spain.
| |
Collapse
|
11
|
Bui S, Mejia I, Díaz B, Wang Y. Adaptation of the Golgi Apparatus in Cancer Cell Invasion and Metastasis. Front Cell Dev Biol 2021; 9:806482. [PMID: 34957124 PMCID: PMC8703019 DOI: 10.3389/fcell.2021.806482] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
The Golgi apparatus plays a central role in normal cell physiology by promoting cell survival, facilitating proliferation, and enabling cell-cell communication and migration. These roles are partially mediated by well-known Golgi functions, including post-translational modifications, lipid biosynthesis, intracellular trafficking, and protein secretion. In addition, accumulating evidence indicates that the Golgi plays a critical role in sensing and integrating external and internal cues to promote cellular homeostasis. Indeed, the unique structure of the mammalian Golgi can be fine-tuned to adapt different Golgi functions to specific cellular needs. This is particularly relevant in the context of cancer, where unrestrained proliferation and aberrant survival and migration increase the demands in Golgi functions, as well as the need for Golgi-dependent sensing and adaptation to intrinsic and extrinsic stressors. Here, we review and discuss current understanding of how the structure and function of the Golgi apparatus is influenced by oncogenic transformation, and how this adaptation may facilitate cancer cell invasion and metastasis.
Collapse
Affiliation(s)
- Sarah Bui
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Isabel Mejia
- Department of Internal Medicine, Division of Medical Hematology and Oncology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Begoña Díaz
- Department of Internal Medicine, Division of Medical Hematology and Oncology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States.,David Geffen School of Medicine and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yanzhuang Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, United States.,Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| |
Collapse
|
12
|
Erfani S, Hua H, Pan Y, Zhou BP, Yang XH. The Context-Dependent Impact of Integrin-Associated CD151 and Other Tetraspanins on Cancer Development and Progression: A Class of Versatile Mediators of Cellular Function and Signaling, Tumorigenesis and Metastasis. Cancers (Basel) 2021; 13:cancers13092005. [PMID: 33919420 PMCID: PMC8122392 DOI: 10.3390/cancers13092005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/18/2021] [Accepted: 04/01/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Tetraspanins are a family of molecules abundantly expressed on the surface of normal or tumor cells. They have been implicated in recruiting or sequestering key molecular regulators of malignancy of a variety of human cancers, including breast and lung cancers, glioblastoma and leukemia. Yet, how their actions take place remains mysterious due to a lack of traditional platform for molecular interactions. The current review digs into this mystery by examining findings from recent studies of multiple tetraspanins, particularly CD151. The molecular basis for differential impact of tetraspanins on tumor development, progression, and spreading to secondary sites is highlighted, and the complexity and plasticity of their control over tumor cell activities and interaction with their surroundings is discussed. Finally, an outlook is provided regarding tetraspanins as candidate biomarkers and targets for the diagnosis and treatment of human cancer. Abstract As a family of integral membrane proteins, tetraspanins have been functionally linked to a wide spectrum of human cancers, ranging from breast, colon, lung, ovarian, prostate, and skin carcinomas to glioblastoma. CD151 is one such prominent member of the tetraspanin family recently suggested to mediate tumor development, growth, and progression in oncogenic context- and cell lineage-dependent manners. In the current review, we summarize recent advances in mechanistic understanding of the function and signaling of integrin-associated CD151 and other tetraspanins in multiple cancer types. We also highlight emerging genetic and epigenetic evidence on the intrinsic links between tetraspanins, the epithelial-mesenchymal transition (EMT), cancer stem cells (CSCs), and the Wnt/β-catenin pathway, as well as the dynamics of exosome and cellular metabolism. Finally, we discuss the implications of the highly plastic nature and epigenetic susceptibility of CD151 expression, function, and signaling for clinical diagnosis and therapeutic intervention for human cancer.
Collapse
Affiliation(s)
- Sonia Erfani
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536, USA;
- Markey Cancer Center, University of Kentucky Medical Center, Lexington, KY 40536, USA
- Pharmacy Department, St. Elizabeth Healthcare, Edgewood, KY 41017, USA
| | - Hui Hua
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui 230001, China; (H.H.); (Y.P.)
- Provincial Hospital, Hefei, Anhui 230001, China
| | - Yueyin Pan
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui 230001, China; (H.H.); (Y.P.)
- Provincial Hospital, Hefei, Anhui 230001, China
| | - Binhua P. Zhou
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY 40536, USA;
| | - Xiuwei H. Yang
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536, USA;
- Markey Cancer Center, University of Kentucky Medical Center, Lexington, KY 40536, USA
- Correspondence: ; Tel.: +1-859-323-1996
| |
Collapse
|
13
|
Chen D, He F, Lu T, Huang J, Li M, Cai D, Huang C, Chen D, Xiong F. VPS4B deficiency causes early embryonic lethality and induces signal transduction disorders of cell endocytosis. Genesis 2021; 59:e23415. [PMID: 33682352 DOI: 10.1002/dvg.23415] [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: 11/12/2020] [Revised: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 11/08/2022]
Abstract
VPS4B (vacuolar protein sorting 4B), a member of the ATPase associated with diverse cellular activities (AAA) protein family, is a component of the endosomal sorting complexes required for transport machinery which regulates the internalization and lysosomal degradation of membrane proteins. We previously reported that VPS4B is one of the pathogenic genes related to dentin dysplasia type I, although its function was largely unknown. To investigate the role of VPS4B in tooth development, we deleted the Vps4b gene in mice. We found that heterozygous knockout mice (Vps4b+/- ) developed normally and were fertile. However, homozygous deletion of the Vps4b gene resulted in early embryonic lethality of Vps4b-/- mice at approximately embryonic day 9.5 (E9.5). To investigate the underlying molecular mechanisms, we examined the molecular functions of VPS4B in vivo and in vitro. Cell experiments showed that VPS4B influenced the proliferation, apoptosis, and cell cycle of transfected human neuroblastoma cells (IMR-32 cells) with over-expression or knockdown of VPS4B. Moreover, qRT-PCR detection showed that the mRNA expression levels of apoptosis-, cell cycle-, and endocytosis-related genes was significantly down or up-regulated in RNA interference-mediated knockdown of VPS4B in IMR-32 cells and Vps4b+/- E12.5 embryos. We accordingly speculated that signal transduction disorders of cell endocytosis are a contributing factor to the prenatal lethality of Vps4b-/- mice.
Collapse
Affiliation(s)
- Danna Chen
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Fei He
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ting Lu
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jin Huang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Meiyi Li
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Decheng Cai
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Cheng Huang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Dong Chen
- Department of Stomatology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fu Xiong
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Key Laboratory of Single Cell Technology and Application in Guangdong, Guangzhou, China.,Guangdong Province Key Laboratory of Psychiatric Disorders, Guangzhou, China
| |
Collapse
|
14
|
Lorico A, Lorico-Rappa M, Karbanová J, Corbeil D, Pizzorno G. CD9, a tetraspanin target for cancer therapy? Exp Biol Med (Maywood) 2021; 246:1121-1138. [PMID: 33601913 DOI: 10.1177/1535370220981855] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In the present minireview, we intend to provide a brief history of the field of CD9 involvement in oncogenesis and in the metastatic process of cancer, considering its potential value as a tumor-associated antigenic target. Over the years, CD9 has been identified as a favorable prognostic marker or predictor of metastatic potential depending on the cancer type. To understand its implications in cancer beside its use as an antigenic biomarker, it is essential to know its physiological functions, including its molecular partners in a given cell system. Moreover, the discovery that CD9 is one of the most specific and broadly expressed markers of extracellular membrane vesicles, nanometer-sized entities that are released into extracellular space and various physiological body fluids and play a role in intercellular communication under physiological and pathological conditions, notably the establishment of cancer metastases, has added a new dimension to our knowledge of CD9 function in cancer. Here, we will discuss these issues as well as the possible cancer therapeutic implications of CD9, their limitations, and pitfalls.
Collapse
Affiliation(s)
- Aurelio Lorico
- Touro University College of Medicine, Henderson, NV 89014, USA.,Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | | | - Jana Karbanová
- Biotechnology Center and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden 01307, Germany
| | - Denis Corbeil
- Biotechnology Center and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden 01307, Germany
| | - Giuseppe Pizzorno
- University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Erlanger Health System, Chattanooga, TN 37403 , USA
| |
Collapse
|
15
|
He J, Gu H, Wang W, Hu Y. Two CD9 tetraspanin family members of Japanese flounder (Paralichthys olivaceus): characterization and comparative analysis of the anti-infectious immune function. Vet Res 2021; 52:28. [PMID: 33597018 PMCID: PMC7890607 DOI: 10.1186/s13567-021-00903-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 01/10/2021] [Indexed: 12/14/2022] Open
Abstract
CD9 is a glycoprotein of the transmembrane 4 superfamily that is involved in various cellular processes. Studies related to the immune functions and activities of CD9 in teleost fish are limited. In this study, we characterized two CD9 homologs, PoCD9.1 and PoCD9.3, from Japanese flounder (Paralichthys olivaceus). Sequence analysis showed that PoCD9.1 and PoCD9.3 possess characteristic transmembrane 4 superfamily (TM4SF) structures. PoCD9.1 shares 70.61% sequence identity with PoCD9.3. The expression of PoCD9.1 and PoCD9.3 in the three main immune tissues was significantly induced in a time-dependent manner by extracellular and intracellular pathogen infection, which indicates that the two CD9 homologs play an important role in the response to pathogenic infection. Following infection with the extracellular pathogen Vibrio anguillarum, the expression profiles of both PoCD9.1 and PoCD9.3 were similar. After infection with the intracellular pathogen Edwardsiella piscicida, the expression levels of PoCD9.1 and PoCD9.3 were different at different stages of infection, especially in the spleen. The spleen was the most important tissue for the PoCD9.1 and PoCD9.3 responses to pathogen infection among the three examined immune tissues. Knockdown of PoCD9.1 and PoCD9.3 attenuated the ability of host cells to eliminate pathogenic bacteria, and PoCD9.1 knockdown was more lethal than PoCD9.3 knockdown for host cells with E. piscicida infection. Overexpression of PoCD9.1 and PoCD9.3 promoted host or host cell defence against E. piscicida infection. These findings suggest that PoCD9.1 and PoCD9.3 serve as immune-related factors, play an important role in the immune defence system of Japanese flounder, and display different functions in response to different pathogens at different stages of infection.
Collapse
Affiliation(s)
- Jiaojiao He
- Marine Science and Engineering College, Qingdao Agricultural University, Qingdao, 266109, China.,Institute of Tropical Bioscience and Biotechnology, Hainan Academy of Tropical Agricultural Resource, CATAS, Haikou, 571101, China
| | - Hanjie Gu
- Institute of Tropical Bioscience and Biotechnology, Hainan Academy of Tropical Agricultural Resource, CATAS, Haikou, 571101, China.,Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bioresources, Haikou, 571101, China
| | - Wenqi Wang
- Marine Science and Engineering College, Qingdao Agricultural University, Qingdao, 266109, China.
| | - Yonghua Hu
- Institute of Tropical Bioscience and Biotechnology, Hainan Academy of Tropical Agricultural Resource, CATAS, Haikou, 571101, China. .,Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266071, China. .,Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bioresources, Haikou, 571101, China.
| |
Collapse
|
16
|
Bobrowicz M, Kubacz M, Slusarczyk A, Winiarska M. CD37 in B Cell Derived Tumors-More than Just a Docking Point for Monoclonal Antibodies. Int J Mol Sci 2020; 21:ijms21249531. [PMID: 33333768 PMCID: PMC7765243 DOI: 10.3390/ijms21249531] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/10/2020] [Accepted: 12/13/2020] [Indexed: 12/20/2022] Open
Abstract
CD37 is a tetraspanin expressed prominently on the surface of B cells. It is an attractive molecular target exploited in the immunotherapy of B cell-derived lymphomas and leukemia. Currently, several monoclonal antibodies targeting CD37 as well as chimeric antigen receptor-based immunotherapies are being developed and investigated in clinical trials. Given the unique role of CD37 in the biology of B cells, it seems that CD37 constitutes more than a docking point for monoclonal antibodies, and targeting this molecule may provide additional benefit to relapsed or refractory patients. In this review, we aimed to provide an extensive overview of the function of CD37 in B cell malignancies, providing a comprehensive view of recent therapeutic advances targeting CD37 and delineating future perspectives.
Collapse
MESH Headings
- Antibodies, Monoclonal/therapeutic use
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Antineoplastic Agents, Immunological/therapeutic use
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- B-Lymphocytes/pathology
- Humans
- Immunotherapy/methods
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Lymphoma, B-Cell/drug therapy
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Tetraspanins/immunology
- Tetraspanins/metabolism
Collapse
|
17
|
Tetraspanins: useful multifunction proteins for the possible design and development of small-molecule therapeutic tools. Drug Discov Today 2020; 26:56-68. [PMID: 33137483 DOI: 10.1016/j.drudis.2020.10.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/21/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023]
Abstract
Tetraspanins constitute a well-conserved superfamily of four-span small membrane proteins (TM4SF), with >30 members in humans, with important roles in numerous mechanisms of cell biology. Moreover, tetraspanins associate with either specific partner proteins or another tetraspanin, generating a network of interactions involved in cell and membrane compartmentalization and having a role in cellular development, proliferation, activation, motility, and membrane fusions. Therefore, tetraspanins are considered regulators of cellular signaling and are often depicted as 'molecular facilitators'. In view of these many physiological functions, it is likely that these molecules are important actors in pathological processes. In this review, we present the main characteristics of this superfamily, providing a more detailed description of some significant representatives and discuss their relevance as potential targets for the design and development of small-molecule therapeutics in different pathologies.
Collapse
|
18
|
Kgk D, Kumari S, G S, Malla RR. Marine natural compound cyclo(L-leucyl-L-prolyl) peptide inhibits migration of triple negative breast cancer cells by disrupting interaction of CD151 and EGFR signaling. Chem Biol Interact 2019; 315:108872. [PMID: 31669320 DOI: 10.1016/j.cbi.2019.108872] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/30/2019] [Accepted: 10/21/2019] [Indexed: 12/14/2022]
Abstract
Cyclo (L-Leucyl-L-Prolyl) peptide/CLP is a marine natural metabolite and well recognized as an antimicrobial and antioxidant agent with limited studies on anticancer activity. The current study aims to determine the effect of CLP on migration and growth of triple negative breast cancer cell lines. The anti-growth potential was evaluated by MTT, BrdU and TUNEL assays; DNA damage by γH2AX and Dead green assays; antimigration activity by Boyden chamber invasion and wound healing assays. Interaction of CLP with CD151 was resolved by PatchDock. Effect of CLP on the expression of transmembrane CD151 was evaluated by cell-based ELISA assay. The interaction between CD151 and EGFR was predicted by using FireDoc Web server. Impact of CLP on the interaction of CD151 with EGFR was evaluated by co-immunoprecipitation assay. The effect of CLP on the cell cycle and its controlling proteins was determined by Western blotting. CLP reduced the viability of MDA-MB-231 and MDA-MB-468 TNBC cell lines but not human breast healthy epithelial cell line (MCF-12A) similar to eribulin, standard. CLP also inhibited proliferation; cell cycle and migration. It induced DNA strand breaks, DNA damage, and cell death. It showed the most favorable interactions with CD151 in in silico docking and significantly reduced the expression of membrane-bound CD151 proteins. FireDoc Web study predicted the association between CD151 and EGFR with -29.13 kcal/mol of binding energy. CLP reduced the interaction of CD151 with EGFR along with the expression of cyclin D, CDK4, PAK, RAC1, and P27kiP1. This study concludes that CLP suppresses growth and migration by attenuating cell cycle of TNBC cell lines via EGFR and CD151 signaling. Thus, exploring the EGFR and CD151 signaling pathway targeted by CLP may provide a new approach in the treatment of TNBC.
Collapse
Affiliation(s)
- Deepak Kgk
- Cancer Biology Lab, Department of Biochemistry, GIS, GITAM (Deemed to be University), Visakhapatnam, 530045, Andhra Pradesh, India
| | - Seema Kumari
- Cancer Biology Lab, Department of Biochemistry, GIS, GITAM (Deemed to be University), Visakhapatnam, 530045, Andhra Pradesh, India
| | - Shailender G
- Cancer Biology Lab, Department of Biochemistry, GIS, GITAM (Deemed to be University), Visakhapatnam, 530045, Andhra Pradesh, India
| | - Rama Rao Malla
- Cancer Biology Lab, Department of Biochemistry, GIS, GITAM (Deemed to be University), Visakhapatnam, 530045, Andhra Pradesh, India.
| |
Collapse
|
19
|
Lazareth H, Henique C, Lenoir O, Puelles VG, Flamant M, Bollée G, Fligny C, Camus M, Guyonnet L, Millien C, Gaillard F, Chipont A, Robin B, Fabrega S, Dhaun N, Camerer E, Kretz O, Grahammer F, Braun F, Huber TB, Nochy D, Mandet C, Bruneval P, Mesnard L, Thervet E, Karras A, Le Naour F, Rubinstein E, Boucheix C, Alexandrou A, Moeller MJ, Bouzigues C, Tharaux PL. The tetraspanin CD9 controls migration and proliferation of parietal epithelial cells and glomerular disease progression. Nat Commun 2019; 10:3303. [PMID: 31341160 PMCID: PMC6656772 DOI: 10.1038/s41467-019-11013-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 06/07/2019] [Indexed: 01/02/2023] Open
Abstract
The mechanisms driving the development of extracapillary lesions in focal segmental glomerulosclerosis (FSGS) and crescentic glomerulonephritis (CGN) remain poorly understood. A key question is how parietal epithelial cells (PECs) invade glomerular capillaries, thereby promoting injury and kidney failure. Here we show that expression of the tetraspanin CD9 increases markedly in PECs in mouse models of CGN and FSGS, and in kidneys from individuals diagnosed with these diseases. Cd9 gene targeting in PECs prevents glomerular damage in CGN and FSGS mouse models. Mechanistically, CD9 deficiency prevents the oriented migration of PECs into the glomerular tuft and their acquisition of CD44 and β1 integrin expression. These findings highlight a critical role for de novo expression of CD9 as a common pathogenic switch driving the PEC phenotype in CGN and FSGS, while offering a potential therapeutic avenue to treat these conditions. In both focal segmental glomerulosclerosis (FSGS) and crescentic glomerulonephritis (CGN), kidney injury is characterised by the invasion of glomerular tufts by parietal epithelial cells (PECs). Here Lazareth et al. identify the tetraspanin CD9 as a key regulator of PEC migration, and find its upregulation in FSGS and CGN contributes to kidney injury in both diseases.
Collapse
Affiliation(s)
- Hélène Lazareth
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France.,Renal Division, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, Paris, F-75015, France.,Laboratoire d'Optique et Biosciences, Ecole polytechnique, CNRS UMR7645, INSERM U1182, Université Paris-Saclay, Palaiseau, F-91128, France
| | - Carole Henique
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France. .,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France. .,Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe 21, Université Paris Est Créteil, Créteil, F-94010, France.
| | - Olivia Lenoir
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France
| | - Victor G Puelles
- Department of Nephrology and Clinical Immunology, University Hospital RWTH Aachen, Pauwelsstrasse 30, D-52074, Aachen, Germany.,Department of Medicine III, Faculty of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany.,Department of Nephrology and Center for Inflammatory Diseases, Monash University, Melbourne, VIC 3168, Australia
| | - Martin Flamant
- Xavier Bichat University Hospital, Université de Paris, Paris, F-75018, France
| | - Guillaume Bollée
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France
| | - Cécile Fligny
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France
| | - Marine Camus
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France
| | - Lea Guyonnet
- National Cytometry Platform, Department of Infection and Immunity, Luxembourg Institute of Health, Luxembourg, L-4354, Luxembourg
| | - Corinne Millien
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France
| | - François Gaillard
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France
| | - Anna Chipont
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France
| | - Blaise Robin
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France
| | - Sylvie Fabrega
- Université de Paris, Institut Imagine, Plateforme Vecteurs Viraux et Transfert de Gènes, IFR94, Hôpital Necker Enfants-Malades, Paris, F-75015, France
| | - Neeraj Dhaun
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, EH16 4SA, Scotland, UK
| | - Eric Camerer
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France
| | - Oliver Kretz
- Department of Medicine III, Faculty of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany.,Renal Division, Faculty of Medicine, Medical Centre, University of Freiburg, Freiburg, D-79106, Germany
| | - Florian Grahammer
- Department of Medicine III, Faculty of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany.,Renal Division, Faculty of Medicine, Medical Centre, University of Freiburg, Freiburg, D-79106, Germany
| | - Fabian Braun
- Department of Medicine III, Faculty of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany.,Renal Division, Faculty of Medicine, Medical Centre, University of Freiburg, Freiburg, D-79106, Germany
| | - Tobias B Huber
- Department of Medicine III, Faculty of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany.,Renal Division, Faculty of Medicine, Medical Centre, University of Freiburg, Freiburg, D-79106, Germany
| | - Dominique Nochy
- Department of Pathology, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Paris, F-75015, France
| | - Chantal Mandet
- Department of Pathology, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Paris, F-75015, France
| | - Patrick Bruneval
- Department of Pathology, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Paris, F-75015, France
| | - Laurent Mesnard
- Critical Care Nephrology and Kidney Transplantation, Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, Unité Mixte de Recherche S1155, Pierre and Marie Curie University, Paris, F-75020, France
| | - Eric Thervet
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France.,Renal Division, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, Paris, F-75015, France
| | - Alexandre Karras
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France.,Renal Division, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, Paris, F-75015, France
| | | | - Eric Rubinstein
- Inserm U935, Université Paris-Sud, Villejuif, F-94800, France
| | - Claude Boucheix
- Inserm U935, Université Paris-Sud, Villejuif, F-94800, France
| | - Antigoni Alexandrou
- Laboratoire d'Optique et Biosciences, Ecole polytechnique, CNRS UMR7645, INSERM U1182, Université Paris-Saclay, Palaiseau, F-91128, France
| | - Marcus J Moeller
- Department of Nephrology and Clinical Immunology, University Hospital RWTH Aachen, Pauwelsstrasse 30, D-52074, Aachen, Germany
| | - Cédric Bouzigues
- Laboratoire d'Optique et Biosciences, Ecole polytechnique, CNRS UMR7645, INSERM U1182, Université Paris-Saclay, Palaiseau, F-91128, France
| | - Pierre-Louis Tharaux
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France. .,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France. .,Renal Division, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, Paris, F-75015, France.
| |
Collapse
|
20
|
Baek J, Jang N, Choi JE, Kim JR, Bae YK. CD9 Expression in Tumor Cells Is Associated with Poor Prognosis in Patients with Invasive Lobular Carcinoma. J Breast Cancer 2019; 22:77-85. [PMID: 30941235 PMCID: PMC6438839 DOI: 10.4048/jbc.2019.22.e9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/15/2019] [Indexed: 12/23/2022] Open
Abstract
Purpose We investigated the prognostic significance of CD9 expression in tumor cells of patients with invasive lobular carcinoma (ILC). Methods CD9 expression was evaluated by immunohistochemistry in 113 ILC tissue samples. Correlation of CD9 expression with the patients' clinicopathological parameters and overall survival was assessed. Results CD9 expression was detected in 48 (42.5%) ILC patients. However, no significant relation could be determined between CD9 expression and the clinicopathological parameters of the patient including tumor size, lymph node metastasis, lymphovascular invasion, histologic grade, expression of hormone receptors, human epidermal growth factor receptor 2 status, and Ki-67 labeling index. Patients with CD9 expression had worse overall survival (p = 0.051) and disease-free survival (DFS, p = 0.014) compared to patients without CD9 expression. Multivariate analysis revealed that CD9 expression was an independent prognostic factor for DFS (p = 0.049). Conclusion CD9 expression in tumor cells could be a significant prognostic marker in patients with ILC.
Collapse
Affiliation(s)
- Jina Baek
- Department of Pathology, Yeungnam University College of Medicine, Daegu, Korea
| | - Nuri Jang
- Department of Pathology, Yeungnam University College of Medicine, Daegu, Korea
| | - Jung Eun Choi
- Department of Breast Surgery, Yeungnam University College of Medicine, Daegu, Korea
| | - Jae-Ryong Kim
- Department of Biochemistry and Molecular Biology, Yeungnam University College of Medicine, Daegu, Korea
| | - Young Kyung Bae
- Department of Pathology, Yeungnam University College of Medicine, Daegu, Korea
| |
Collapse
|
21
|
Reyes R, Cardeñes B, Machado-Pineda Y, Cabañas C. Tetraspanin CD9: A Key Regulator of Cell Adhesion in the Immune System. Front Immunol 2018; 9:863. [PMID: 29760699 PMCID: PMC5936783 DOI: 10.3389/fimmu.2018.00863] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 04/09/2018] [Indexed: 12/21/2022] Open
Abstract
The tetraspanin CD9 is expressed by all the major subsets of leukocytes (B cells, CD4+ T cells, CD8+ T cells, natural killer cells, granulocytes, monocytes and macrophages, and immature and mature dendritic cells) and also at a high level by endothelial cells. As a typical member of the tetraspanin superfamily, a prominent feature of CD9 is its propensity to engage in a multitude of interactions with other tetraspanins as well as with different transmembrane and intracellular proteins within the context of defined membranal domains termed tetraspanin-enriched microdomains (TEMs). Through these associations, CD9 influences many cellular activities in the different subtypes of leukocytes and in endothelial cells, including intracellular signaling, proliferation, activation, survival, migration, invasion, adhesion, and diapedesis. Several excellent reviews have already covered the topic of how tetraspanins, including CD9, regulate these cellular processes in the different cells of the immune system. In this mini-review, however, we will focus particularly on describing and discussing the regulatory effects exerted by CD9 on different adhesion molecules that play pivotal roles in the physiology of leukocytes and endothelial cells, with a particular emphasis in the regulation of adhesion molecules of the integrin and immunoglobulin superfamilies.
Collapse
Affiliation(s)
- Raquel Reyes
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Beatriz Cardeñes
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Yesenia Machado-Pineda
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Carlos Cabañas
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.,Departamento de Inmunología, Oftalmología y OTR (IO2), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| |
Collapse
|
22
|
Saiz ML, Cibrian D, Ramírez-Huesca M, Torralba D, Moreno-Gonzalo O, Sánchez-Madrid F. Tetraspanin CD9 Limits Mucosal Healing in Experimental Colitis. Front Immunol 2017; 8:1854. [PMID: 29312336 PMCID: PMC5742144 DOI: 10.3389/fimmu.2017.01854] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/07/2017] [Indexed: 12/19/2022] Open
Abstract
Tetraspanins are a family of proteins with four transmembrane domains that associate between themselves and cluster with other partner proteins, conforming a distinct class of membrane domains, the tetraspanin-enriched microdomains (TEMs). These TEMs constitute macromolecular signaling platforms that regulate key processes in several cellular settings controlling signaling thresholds and avidity of receptors. In this study, we investigated the role of CD9, a tetraspanin that regulates major biological processes such as cell migration and immunological responses, in two mouse models of colitis that have been used to study the pathogenesis of inflammatory bowel disease (IBD). Previous in vitro studies revealed an important role in the interaction of leukocytes with inflamed endothelium, but in vivo evidence of the involvement of CD9 in inflammatory diseases is scarce. Here, we studied the role of CD9 in the pathogenesis of colitis in vivo. Colitis was induced by administration of dextran sodium sulfate (DSS), a chemical colitogen that causes epithelial disruption and intestinal inflammation. CD9−/− mice showed less severe colitis than wild-type counterparts upon exposure to DSS (2% solution) and enhanced survival in response to a lethal DSS dose (4%). Decreased neutrophil and macrophage cell infiltration was observed in colonic tissue from CD9−/− animals, in accordance with their lower serum levels of TNF-α, IL-6, and other proinflammatory cytokines in the colon. The specific role of CD9 in IBD was further dissected by transfer of CD4+ CD45RBhi naive T cells into the Rag1−/− mouse colitis model. However, no significant differences were observed in these settings between both groups, ruling out a role for CD9 in IBD in the lymphoid compartment. Experiments with bone marrow chimeras revealed that CD9 in the non-hematopoietic compartment is involved in colon injury and limits the proliferation of epithelial cells. Our data indicate that CD9 in non-hematopoietic cells plays an important role in colitis by limiting epithelial cell proliferation. Future strategies to repress CD9 expression may be of therapeutic benefit in the treatment of IBD.
Collapse
Affiliation(s)
- María Laura Saiz
- Immunology Service, Hospital de la Princesa, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa, Madrid, Spain.,Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Danay Cibrian
- Immunology Service, Hospital de la Princesa, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa, Madrid, Spain.,Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,CIBER Cardiovascular, Madrid, Spain
| | - Marta Ramírez-Huesca
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Daniel Torralba
- Immunology Service, Hospital de la Princesa, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa, Madrid, Spain.,Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Olga Moreno-Gonzalo
- Immunology Service, Hospital de la Princesa, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa, Madrid, Spain.,Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Francisco Sánchez-Madrid
- Immunology Service, Hospital de la Princesa, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa, Madrid, Spain.,Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,CIBER Cardiovascular, Madrid, Spain
| |
Collapse
|
23
|
Soekmadji C, Corcoran NM, Oleinikova I, Jovanovic L, Ramm GA, Nelson CC, Jenster G, Russell PJ. Extracellular vesicles for personalized therapy decision support in advanced metastatic cancers and its potential impact for prostate cancer. Prostate 2017; 77:1416-1423. [PMID: 28856701 DOI: 10.1002/pros.23403] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 08/03/2017] [Indexed: 12/31/2022]
Abstract
The use of circulating tumor cells (CTCs) and circulating extracellular vesicles (EVs), such as exosomes, as liquid biopsy-derived biomarkers for cancers have been investigated. CTC enumeration using the CellSearch based platform provides an accurate insight on overall survival where higher CTC counts indicate poor prognosis for patients with advanced metastatic cancer. EVs provide information based on their lipid, protein, and nucleic acid content and can be isolated from biofluids and analyzed from a relatively small volume, providing a routine and non-invasive modality to monitor disease progression. Our pilot experiment by assessing the level of two subpopulations of small EVs, the CD9 positive and CD63 positive EVs, showed that the CD9 positive EV level is higher in plasma from patients with advanced metastatic prostate cancer with detectable CTCs. These data show the potential utility of a particular EV subpopulation to serve as biomarkers for advanced metastatic prostate cancer. EVs can potentially be utilized as biomarkers to provide accurate genotypic and phenotypic information for advanced prostate cancer, where new strategies to design a more personalized therapy is currently the focus of considerable investigation.
Collapse
Affiliation(s)
- Carolina Soekmadji
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Niall M Corcoran
- Australian Prostate Cancer Research Centre Epworth, and Department of Surgery, University of Melbourne, Australia
| | - Irina Oleinikova
- Department of Urology, Queensland Health, Princess Alexandra Hospital, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Lidija Jovanovic
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- Translational Research Institute, Brisbane, Queensland, Australia
| | - Grant A Ramm
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Colleen C Nelson
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- Translational Research Institute, Brisbane, Queensland, Australia
| | - Guido Jenster
- Department of Urology, Erasmus Medical Centre, R,otterdam, The Netherlands
| | - Pamela J Russell
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- Translational Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
24
|
Reducing isoform complexity of human tetraspanins by optimized expression in Dictyostelium discoideum enables high-throughput functional read-out. Protein Expr Purif 2017; 135:8-15. [DOI: 10.1016/j.pep.2017.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 04/18/2017] [Accepted: 04/20/2017] [Indexed: 11/21/2022]
|
25
|
Xie J, Wang CL, Yang W, Wang J, Chen C, Zheng L, Sung KP, Zhou X. Modulation of MMP-2 and MMP-9 through connected pathways and growth factors is critical for extracellular matrix balance of intra-articular ligaments. J Tissue Eng Regen Med 2017; 12:e550-e565. [PMID: 27684403 DOI: 10.1002/term.2325] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 07/29/2016] [Accepted: 09/26/2016] [Indexed: 02/05/2023]
Affiliation(s)
- Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan Province China
| | - Chun-Li Wang
- Key Laboratory of Biorheological Science and Technology, Bioengineering College; Chongqing University; Chongqing China
| | - Wenbin Yang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan Province China
| | - Jue Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan Province China
| | - Cheng Chen
- Centre for Joint Surgery, Southwest Hospital; Third Military Medical University; Chongqing China
| | - Liwei Zheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan Province China
| | - K.L. Paul Sung
- Key Laboratory of Biorheological Science and Technology, Bioengineering College; Chongqing University; Chongqing China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology; Sichuan University; Chengdu Sichuan Province China
| |
Collapse
|
26
|
Termini CM, Gillette JM. Tetraspanins Function as Regulators of Cellular Signaling. Front Cell Dev Biol 2017; 5:34. [PMID: 28428953 PMCID: PMC5382171 DOI: 10.3389/fcell.2017.00034] [Citation(s) in RCA: 193] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 03/22/2017] [Indexed: 01/10/2023] Open
Abstract
Tetraspanins are molecular scaffolds that distribute proteins into highly organized microdomains consisting of adhesion, signaling, and adaptor proteins. Many reports have identified interactions between tetraspanins and signaling molecules, finding unique downstream cellular consequences. In this review, we will explore these interactions as well as the specific cellular responses to signal activation, focusing on tetraspanin regulation of adhesion-mediated (integrins/FAK), receptor-mediated (EGFR, TNF-α, c-Met, c-Kit), and intracellular signaling (PKC, PI4K, β-catenin). Additionally, we will summarize our current understanding for how tetraspanin post-translational modifications (palmitoylation, N-linked glycosylation, and ubiquitination) can regulate signal propagation. Many of the studies outlined in this review suggest that tetraspanins offer a potential therapeutic target to modulate aberrant signal transduction pathways that directly impact a host of cellular behaviors and disease states.
Collapse
Affiliation(s)
- Christina M Termini
- Department of Pathology, University of New Mexico Health Sciences CenterAlbuquerque, NM, USA
| | - Jennifer M Gillette
- Department of Pathology, University of New Mexico Health Sciences CenterAlbuquerque, NM, USA
| |
Collapse
|
27
|
Termini CM, Lidke KA, Gillette JM. Tetraspanin CD82 Regulates the Spatiotemporal Dynamics of PKCα in Acute Myeloid Leukemia. Sci Rep 2016; 6:29859. [PMID: 27417454 PMCID: PMC4945921 DOI: 10.1038/srep29859] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/22/2016] [Indexed: 02/08/2023] Open
Abstract
Patients with acute myeloid leukemia (AML) have increased myeloid cells within their bone marrow that exhibit aberrant signaling. Therefore, therapeutic targets that modulate disrupted signaling cascades are of significant interest. In this study, we demonstrate that the tetraspanin membrane scaffold, CD82, regulates protein kinase c alpha (PKCα)-mediated signaling critical for AML progression. Utilizing a palmitoylation mutant form of CD82 with disrupted membrane organization, we find that the CD82 scaffold controls PKCα expression and activation. Combining single molecule and ensemble imaging measurements, we determine that CD82 stabilizes PKCα activation at the membrane and regulates the size of PKCα membrane clusters. Further evaluation of downstream effector signaling identified robust and sustained activation of ERK1/2 upon CD82 overexpression that results in enhanced AML colony formation. Together, these data propose a mechanism where CD82 membrane organization regulates sustained PKCα signaling that results in an aggressive leukemia phenotype. These observations suggest that the CD82 scaffold may be a potential therapeutic target for attenuating aberrant signal transduction in AML.
Collapse
Affiliation(s)
- Christina M Termini
- Department of Pathology, University of New Mexico Health Sciences Center, University of New Mexico, MSC 08-4640, Albuquerque, NM 87131, USA
| | - Keith A Lidke
- Department of Physics and Astronomy, University of New Mexico, MSC 07-4220, Albuquerque, NM 87131, USA
| | - Jennifer M Gillette
- Department of Pathology, University of New Mexico Health Sciences Center, University of New Mexico, MSC 08-4640, Albuquerque, NM 87131, USA
| |
Collapse
|
28
|
ErbB receptors and tetraspanins: Casting the net wider. Int J Biochem Cell Biol 2016; 77:68-71. [PMID: 27262234 DOI: 10.1016/j.biocel.2016.05.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 05/19/2016] [Accepted: 05/23/2016] [Indexed: 01/15/2023]
|
29
|
Podergajs N, Motaln H, Rajčević U, Verbovšek U, Koršič M, Obad N, Espedal H, Vittori M, Herold-Mende C, Miletic H, Bjerkvig R, Turnšek TL. Transmembrane protein CD9 is glioblastoma biomarker, relevant for maintenance of glioblastoma stem cells. Oncotarget 2016; 7:593-609. [PMID: 26573230 PMCID: PMC4808020 DOI: 10.18632/oncotarget.5477] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 10/31/2015] [Indexed: 12/20/2022] Open
Abstract
The cancer stem cell model suggests that glioblastomas contain a subpopulation of stem-like tumor cells that reproduce themselves to sustain tumor growth. Targeting these cells thus represents a novel treatment strategy and therefore more specific markers that characterize glioblastoma stem cells need to be identified. In the present study, we performed transcriptomic analysis of glioblastoma tissues compared to normal brain tissues revealing sensible up-regulation of CD9 gene. CD9 encodes the transmembrane protein tetraspanin which is involved in tumor cell invasion, apoptosis and resistance to chemotherapy. Using the public REMBRANDT database for brain tumors, we confirmed the prognostic value of CD9, whereby a more than two fold up-regulation correlates with shorter patient survival. We validated CD9 gene and protein expression showing selective up-regulation in glioblastoma stem cells isolated from primary biopsies and in primary organotypic glioblastoma spheroids as well as in U87-MG and U373 glioblastoma cell lines. In contrast, no or low CD9 gene expression was observed in normal human astrocytes, normal brain tissue and neural stem cells. CD9 silencing in three CD133+ glioblastoma cell lines (NCH644, NCH421k and NCH660h) led to decreased cell proliferation, survival, invasion, and self-renewal ability, and altered expression of the stem-cell markers CD133, nestin and SOX2. Moreover, CD9-silenced glioblastoma stem cells showed altered activation patterns of the Akt, MapK and Stat3 signaling transducers. Orthotopic xenotransplantation of CD9-silenced glioblastoma stem cells into nude rats promoted prolonged survival. Therefore, CD9 should be further evaluated as a target for glioblastoma treatment.
Collapse
Affiliation(s)
- Neža Podergajs
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, 1000 Ljubljana, Slovenia
| | - Helena Motaln
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, 1000 Ljubljana, Slovenia
| | - Uroš Rajčević
- Department of Biochemistry, Blood Transfusion Centre of Slovenia, 1000 Ljubljana, Slovenia
| | - Urška Verbovšek
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, 1000 Ljubljana, Slovenia
| | - Marjan Koršič
- Department of Neurosurgery, University Medical Centre, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Nina Obad
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
| | - Heidi Espedal
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
| | - Miloš Vittori
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, 1000 Ljubljana, Slovenia
| | - Christel Herold-Mende
- Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg, 69120 Heidelberg, Germany
| | - Hrvoje Miletic
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
| | - Rolf Bjerkvig
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
- NorLux Neuro-Oncology Laboratory, Centre de Recherche Public de la Santé, 1526 Luxembourg, Luxembourg
| | - Tamara Lah Turnšek
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, 1000 Ljubljana, Slovenia
- Department of Biochemistry, Faculty of Chemistry and Chemical Engineering, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
30
|
Charming neighborhoods on the cell surface: plasma membrane microdomains regulate receptor tyrosine kinase signaling. Cell Signal 2015; 27:1963-76. [PMID: 26163824 DOI: 10.1016/j.cellsig.2015.07.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/07/2015] [Indexed: 12/14/2022]
Abstract
Receptor tyrosine kinases (RTK) are an important family of growth factor and hormone receptors that regulate many aspects of cellular physiology. Ligand binding by RTKs at the plasma membrane elicits activation of many signaling intermediates. The spatial and temporal regulation of RTK signaling within cells is an important determinant of receptor signaling outcome. In particular, the compartmentalization of the plasma membrane into a number of microdomains allows context-specific control of RTK signaling. Indeed various RTKs are recruited to and enriched within specific plasma membrane microdomains under various conditions, including lipid-ordered domains such as caveolae and lipid rafts, clathrin-coated structures, tetraspanin-enriched microdomains, and actin-dependent protrusive membrane microdomains such as dorsal ruffles and invadosomes. We examine the evidence for control of RTK signaling by each of these plasma membrane microdomains, as well as molecular mechanisms for how this spatial organization controls receptor signaling.
Collapse
|
31
|
Tang M, Yin G, Wang F, Liu H, Zhou S, Ni J, Chen C, Zhou Y, Zhao Y. Downregulation of CD9 promotes pancreatic cancer growth and metastasis through upregulation of epidermal growth factor on the cell surface. Oncol Rep 2015; 34:350-8. [PMID: 25955689 DOI: 10.3892/or.2015.3960] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 04/21/2015] [Indexed: 11/06/2022] Open
Abstract
The expression of CD9 has been shown to be inversely associated with pancreatic cancer metastasis but the underlying mechanism remains incompletely understood. Using the two closely associated pancreatic cancer cell lines, PaTu-8898s and PaTu-8898t, which are metastatic and non-metastatic, respectively, we showed that the PaTu-8988s cells expressed a lower level of CD9 but had higher proliferation and migration rates than the PaTu-8898t cells. An inverse correlation between CD9 expression and the cell surface level of epidermal growth factor receptor (EGFR) was observed in both cell lines. In the PaTu-8898s cells, overexpression of CD9 decreased the cell surface expression of EGFR, associated with increased expression of dynamin-2, whereas in the PaTu-8898t cells, knockdown of CD9 with RNA interference (RNAi) increased the cell surface expression of EGFR, associated with decreased expression of dynamin-2. However, the total EGFR level did not change by manipulation of CD9 expression, suggesting that CD9 plays a role in EGFR endocytosis. Furthermore, in the PaTu-8898ts cells, CD9 overexpression decreased the cell proliferation and migration, which were reversed by EGFR overexpression, whereas in the PaTu-8898t cells, CD9 knockdown enhanced the cell proliferation and migration which were blocked by EGFR RNAi both in vitro and in vivo. Thus, in pancreatic cancer cells, downregulation of CD9 may play a role in cancer growth and metastasis through, at least in part, enhancing cell surface expression of EGFR.
Collapse
Affiliation(s)
- Maochun Tang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Guojian Yin
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Feng Wang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Hua Liu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Shu Zhou
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Jianbo Ni
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Congying Chen
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Yingqun Zhou
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Yan Zhao
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| |
Collapse
|
32
|
Murayama Y, Oritani K, Tsutsui S. Novel CD9-targeted therapies in gastric cancer. World J Gastroenterol 2015; 21:3206-3213. [PMID: 25805926 PMCID: PMC4363749 DOI: 10.3748/wjg.v21.i11.3206] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 11/13/2014] [Accepted: 12/16/2014] [Indexed: 02/06/2023] Open
Abstract
There are 33 human tetraspanin proteins, emerging as key players in malignancy, the immune system, fertilization, cellular signaling, adhesion, morphology, motility, proliferation, and tumor invasion. CD9, a member of the tetraspanin family, associates with and influences a variety of cell-surface molecules. Through these interactions, CD9 modifies multiple cellular events, including adhesion, migration, proliferation, and survival. CD9 is therefore considered to play a role in several stages during cancer development. Reduced CD9 expression is generally related to venous vessel invasion and metastasis as well as poor prognosis. We found that treatment of mice bearing human gastric cancer cells with anti-CD9 antibody successfully inhibited tumor progression via antiproliferative, proapoptotic, and antiangiogenic effects, strongly indicating that CD9 is a possible therapeutic target in patients with gastric cancer. Here, we describe the possibility of CD9 manipulation as a novel therapeutic strategy in gastric cancer, which still shows poor prognosis.
Collapse
|
33
|
Sun H, Li G, Zhang W, Zhou Q, Yu Y, Shi Y, Offermanns S, Lu J, Zhou N. Niacin activates the PI3K/Akt cascade via PKC- and EGFR-transactivation-dependent pathways through hydroxyl-carboxylic acid receptor 2. PLoS One 2014; 9:e112310. [PMID: 25375133 PMCID: PMC4223033 DOI: 10.1371/journal.pone.0112310] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 10/04/2014] [Indexed: 01/27/2023] Open
Abstract
Niacin has been demonstrated to activate a PI3K/Akt signaling cascade to prevent brain damage after stroke and UV-induced skin damage; however, the underlying molecular mechanisms for HCA2-induced Akt activation remain to be elucidated. Using CHO-K1 cells stably expressing HCA2 and A431 cells, a human epidermoid cell line with high levels of endogenous expression of functional HCA2 receptors, we first demonstrated that niacin induced a robust Akt phosphorylation at both Thr308 and Ser473 in a time-dependent fashion, with a maximal activation at 5 min and a subsequent reduction to baseline by 30 min through HCA2, and that the activation was significantly blocked by pertussis toxin. The HCA2-mediated activation of Akt was also significantly inhibited by the PKC inhibitors GF109203x and Go6983 in both cell lines, by the PDGFR-selective inhibitor tyrphostin A9 in CHO-HCA2 cells and by the MMP inhibitor GM6001 and EGFR-specific inhibitor AG1478 in A431 cells. These results suggest that the PKC pathway and PDGFR/EGFR transactivation pathway play important roles in HCA2-mediated Akt activation. Further investigation indicated that PI3K and the Gβγ subunit were likely to play an essential role in HCA2-induced Akt activation. Moreover, Immunobloting analyses using an antibody that recognizes p70S6K1 phosphorylated at Thr389 showed that niacin evoked p70S6K1 activation via the PI3K/Akt pathway. The results of our study provide new insight into the signaling pathways involved in HCA2 activation.
Collapse
Affiliation(s)
- Huawang Sun
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guo Li
- College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Aging Research, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Wenjuan Zhang
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qi Zhou
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yena Yu
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ying Shi
- College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang, China
| | - Stefan Offermanns
- Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Jianxin Lu
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
- * E-mail: (NZ); (JL)
| | - Naiming Zhou
- College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang, China
- * E-mail: (NZ); (JL)
| |
Collapse
|
34
|
Tetraspanin CD9 regulates cell contraction and actin arrangement via RhoA in human vascular smooth muscle cells. PLoS One 2014; 9:e106999. [PMID: 25184334 PMCID: PMC4153684 DOI: 10.1371/journal.pone.0106999] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 08/07/2014] [Indexed: 02/07/2023] Open
Abstract
The most prevalent cardiovascular diseases arise from alterations in vascular smooth muscle cell (VSMC) morphology and function. Tetraspanin CD9 has been previously implicated in regulating vascular pathologies; however, insight into how CD9 may regulate adverse VSMC phenotypes has not been provided. We utilized a human model of aortic smooth muscle cells to understand the consequences of CD9 deficiency on VSMC phenotypes. Upon knocking down CD9, the cells developed an abnormally small and rounded morphology. We determined that this morphological change was due to a lack of typical parallel actin arrangement. We also found similar total RhoA but decreased GTP-bound (active) RhoA levels in CD9 deficient cells. As a result, cells lacking a full complement of CD9 were less contractile than their control treated counterparts. Upon restoration of RhoA activity in the CD9 deficient cells, the phenotype was reversed and cell contraction was restored. Conversely, inhibition of RhoA activity in the control cells mimicked the CD9-deficient cell phenotype. Thus, alteration in CD9 expression was sufficient to profoundly disrupt cellular actin arrangement and endogenous cell contraction by interfering with RhoA signaling. This study provides insight into how CD9 may regulate previously described vascular smooth muscle cell pathophysiology.
Collapse
|
35
|
Cheung KJ, Ewald AJ. Illuminating breast cancer invasion: diverse roles for cell-cell interactions. Curr Opin Cell Biol 2014; 30:99-111. [PMID: 25137487 DOI: 10.1016/j.ceb.2014.07.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 07/26/2014] [Accepted: 07/28/2014] [Indexed: 02/08/2023]
Abstract
Metastasis begins when tumors invade into surrounding tissues. In breast cancer, the study of cell interactions has provided fundamental insights into this complex process. Powerful intravital and 3D organoid culture systems have emerged that enable biologists to model the complexity of cell interactions during cancer invasion in real-time. Recent studies utilizing these techniques reveal distinct mechanisms through which multiple cancer cell and stromal cell subpopulations interact, including paracrine signaling, direct cell-cell adhesion, and remodeling of the extracellular matrix. Three cell interaction mechanisms have emerged to explain how breast tumors become invasive: epithelial-mesenchymal transition, collective invasion, and the macrophage-tumor cell feedback loop. Future work is needed to distinguish whether these mechanisms are mutually exclusive or whether they cooperate to drive metastasis.
Collapse
Affiliation(s)
- Kevin J Cheung
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, 855 N. Wolfe St, 452 Rangos Bldg, Baltimore, MD 21205, USA; Department of Oncology, School of Medicine, Johns Hopkins University, 855 N. Wolfe St, 452 Rangos Bldg, Baltimore, MD 21205, USA.
| | - Andrew J Ewald
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, 855 N. Wolfe St, 452 Rangos Bldg, Baltimore, MD 21205, USA; Department of Oncology, School of Medicine, Johns Hopkins University, 855 N. Wolfe St, 452 Rangos Bldg, Baltimore, MD 21205, USA.
| |
Collapse
|
36
|
Chandran RR, Iordanou E, Ajja C, Wille M, Jiang L. Gene expression profiling of Drosophila tracheal fusion cells. Gene Expr Patterns 2014; 15:112-23. [PMID: 24928808 DOI: 10.1016/j.gep.2014.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 05/27/2014] [Accepted: 05/31/2014] [Indexed: 10/25/2022]
Abstract
The Drosophila trachea is a premier genetic system to investigate the fundamental mechanisms of tubular organ formation. Tracheal fusion cells lead the branch fusion process to form an interconnected tubular network. Therefore, fusion cells in the Drosophila trachea will be an excellent model to study branch fusion in mammalian tubular organs, such as kidneys and blood vessels. The fusion process is a dynamic cellular process involving cell migration, adhesion, vesicle trafficking, cytoskeleton rearrangement, and membrane fusion. To understand how these cellular events are coordinated, we initiated the critical step to assemble a gene expression profile of fusion cells. For this study, we analyzed the expression of 234 potential tracheal-expressed genes in fusion cells during fusion cell development. 143 Tracheal genes were found to encode transcription factors, signal proteins, cytoskeleton and matrix proteins, transporters, and proteins with unknown function. These genes were divided into four subgroups based on their levels of expression in fusion cells compared to neighboring non-fusion cells revealed by in situ hybridization: (1) genes that have relative high abundance in fusion cells, (2) genes that are dynamically expressed in fusion cells, (3) genes that have relative low abundance in fusion cells, and (4) genes that are expressed at similar levels in fusion cells and non-fusion tracheal cells. This study identifies the expression profile of fusion cells and hypothetically suggests genes which are necessary for the fusion process and which play roles in distinct stages of fusion, as indicated by the location and timing of expression. These data will provide the basis for a comprehensive understanding of the molecular and cellular mechanisms of branch fusion.
Collapse
Affiliation(s)
- Rachana R Chandran
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, United States
| | - Ekaterini Iordanou
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, United States
| | - Crystal Ajja
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, United States
| | - Michael Wille
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, United States
| | - Lan Jiang
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, United States.
| |
Collapse
|
37
|
Singh B, Coffey RJ. From wavy hair to naked proteins: the role of transforming growth factor alpha in health and disease. Semin Cell Dev Biol 2014; 28:12-21. [PMID: 24631356 DOI: 10.1016/j.semcdb.2014.03.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 02/28/2014] [Accepted: 03/04/2014] [Indexed: 02/07/2023]
Abstract
Since its discovery in 1978 and cloning in 1984, transforming growth factor-alpha (TGF-α, TGFA) has been one of the most extensively studied EGF receptor (EGFR) ligands. In this review, we provide a historical perspective on TGFA-related studies, highlighting what we consider important advances related to its function in normal and disease states.
Collapse
Affiliation(s)
- Bhuminder Singh
- Departments of Medicine and Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Robert J Coffey
- Departments of Medicine and Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Veteran Affairs Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
38
|
Role of epidermal growth factor receptor signaling in the interaction of Neisseria meningitidis with endothelial cells. Infect Immun 2013; 82:1243-55. [PMID: 24379285 DOI: 10.1128/iai.01346-13] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Neisseria meningitidis, the causative agent of meningitis and septicemia, attaches to and invades various cell types. Both steps induce and/or require tyrosine phosphorylation of host cell proteins. Here, we used a phospho array platform to identify active receptor tyrosine kinases (RTKs) and key signaling nodes in N. meningitidis-infected brain endothelial cells to decipher RTK-dependent signaling pathways necessary for bacterial uptake. We detected several activated RTKs, including the ErbB family receptors epidermal growth factor receptor (EGFR), ErbB2, and ErbB4. We found that pharmacological inhibition and genetic ablation of ErbB receptor tyrosine phosphorylation and expression resulted in decreased bacterial uptake and heterologous expression of EGFR, ErbB2, or ErbB4 in Chinese ovary hamster (CHO-K1) cells, which do not express of EGFR and ErbB4; the decrease caused a significant increase in meningococcal invasion. Activation of EGFR and ErbB4 was mediated by transactivation via the common ligand HB-EGF (heparin-binding EGF-like ligand), which was significantly elevated in infected cell culture supernatants. We furthermore determined that N. meningitidis induced phosphorylation of EGFR at Tyr845 independent of ligand binding, which required c-Src activation and was involved in mediating uptake of N. meningitidis into eukaryotic cells. Increased uptake was repressed by expression of EGFR Y845F, which harbored a point mutation in the kinase domain. In addition, activation of ErbB4 at its autophosphorylation site, Tyr1284, and phosphorylation of ErbB2 Thr686 were observed. Altogether, our results provide evidence that EGFR, ErbB2, and ErbB4 are activated in response to N. meningitidis infection and shed new light on the role of ErbB signaling in meningococcal infection biology.
Collapse
|
39
|
Herr MJ, Mabry SE, Jameson JF, Jennings LK. Pro-MMP-9 upregulation in HT1080 cells expressing CD9 is regulated by epidermal growth factor receptor. Biochem Biophys Res Commun 2013; 442:99-104. [PMID: 24246676 DOI: 10.1016/j.bbrc.2013.11.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 11/04/2013] [Indexed: 11/26/2022]
Abstract
Degradation of the surrounding extracellular matrix (ECM) by matrix metalloproteinases (MMPs) drives invasion and metastasis of cancer cells. We previously demonstrated that tetraspanin CD9 expression upregulates pro-MMP-9 expression and release and promotes cellular invasion in a human fibrosarcoma cell line (HT1080). These events were dependent upon the highly functional second extracellular loop of CD9. We report here that the epidermal growth factor receptor (EGFR) tyrosine kinase expression and activity are involved in the CD9-mediated increase in pro-MMP-9 release and cellular invasion. Pro-MMP-9 expression was significantly decreased in a dose-dependent manner using first a broad spectrum receptor tyrosine kinase inhibitor and multiple specific EGFR inhibitors in CD9-HT1080 cells. Furthermore, gefitinib treatment of CD9-HT1080 cells reduced invasion through matrigel. EGFR knockdown using short interfering RNA resulted in decreased pro-MMP-9 expression and release into the media and subsequent cellular invasion without affecting CD9 expression or localization. Conclusively, this study points to EGFR as a key mediator between CD9-mediated pro-MMP-9 release and cellular invasion of HT1080 cells.
Collapse
Affiliation(s)
- Michael J Herr
- The Vascular Biology Center of Excellence, Department of Internal Medicine, USA; Department of Microbiology, Immunology, and Biochemistry, USA
| | | | | | | |
Collapse
|
40
|
Tetraspanins CD9 and CD151, epidermal growth factor receptor and cyclooxygenase-2 expression predict malignant progression in oral epithelial dysplasia. Br J Cancer 2013; 109:2864-74. [PMID: 24201754 PMCID: PMC3844903 DOI: 10.1038/bjc.2013.600] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 08/13/2013] [Accepted: 09/11/2013] [Indexed: 12/31/2022] Open
Abstract
Background: Prognostic biomarkers aim to improve on the current inadequate method of histological assessment to identify patients with oral epithelial dysplasia at greatest risk of malignant transformation. We aimed to assess the prognostic ability of six protein biomarkers linked to the epidermal growth factor receptor (EGFR) pathway, including three tetraspanins, in a large multicentre oral dysplasia cohort. Methods: One hundred and forty-eight cases with varying degrees of epithelial dysplasia underwent immunohistochemical assessment for CD9, CD151, CD82, EGFR, Her-2, and COX-2. Scoring was performed independently by two observers. Univariate analyses using both logistic and Cox regression models and a multivariate regression were performed. Results: Malignant progression was significantly greater in those cases with decreased expression of CD9 (P=0.02), and increased expression of CD151 (P=0.02), EGFR (P=0.04), and COX-2 (P=0.003). Histological grade (P=0.0002) and morphology (P=0.03) were also prognostic, whereas smoking and alcohol were not. The optimal combination by backward-variable selection was of histological grade (hazard ratio (HR) 1.64; 95% CI 1.12, 2.40), COX-2 overexpression (HR 1.12; 1.02, 1.24) and CD9 underexpression (HR 0.88; 0.80, 0.97). CD82 and Her-2 demonstrated no prognostic ability. Conclusion: This is the first study of the expression and prognostic potential of the tetraspanins in oral dysplasia. A combination of certain biomarkers with clinical factors appeared to improve the accuracy of determining the risk of malignancy in individuals with oral dysplasia. These findings may also offer potential new therapeutic approaches for this condition.
Collapse
|
41
|
Steinhauer J, Liu HH, Miller E, Treisman JE. Trafficking of the EGFR ligand Spitz regulates its signaling activity in polarized tissues. J Cell Sci 2013; 126:4469-78. [PMID: 23902690 DOI: 10.1242/jcs.131169] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) ligands undergo a complex series of processing events during their maturation to active signaling proteins. Like its mammalian homologs, the predominant Drosophila EGFR ligand Spitz is produced as a transmembrane pro-protein. In the secretory pathway, Spitz is cleaved within its transmembrane domain to release the extracellular signaling domain. This domain is modified with an N-terminal palmitate group that tethers it to the plasma membrane. We found that the pro-protein can reach the cell surface in the absence of proteolysis, but that it fails to activate the EGFR. To address why the transmembrane pro-protein is inactive, whereas membrane association through the palmitate group promotes activity, we generated a panel of chimeric constructs containing the Spitz extracellular region fused to exogenous transmembrane proteins. Although the orientation of the EGF domain and its distance from the plasma membrane varies in these chimeras, they are all active in vivo. Thus, tethering Spitz to the membrane via a transmembrane domain at either terminus does not prevent activity. Conversely, removing the N-terminal palmitate group from the C-terminally tethered pro-protein does not render it active. Furthermore, we show that the Spitz transmembrane pro-protein can activate the EGFR in a tissue culture assay, indicating that its failure to signal in vivo is not due to structural features. In polarized imaginal disc cells, unprocessed Spitz pro-protein localizes to apical puncta, whereas the active chimeric Spitz constructs are basolaterally localized. Taken together, our data support the model that localized trafficking of the pro-protein restricts its ability to activate the receptor in polarized tissues.
Collapse
|
42
|
Dale BM, Alvarez RA, Chen BK. Mechanisms of enhanced HIV spread through T-cell virological synapses. Immunol Rev 2013; 251:113-24. [PMID: 23278744 DOI: 10.1111/imr.12022] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
An elaborate network of cell-cell interactions in the immune system is essential for vertebrates to mount adaptive immune responses against invading pathogens. For lymphotropic viruses such as the human immunodeficiency virus type 1 (HIV-1), these immune cell interactions can also promote the spread of the virus within the host. The main target of HIV-1 infection is the CD4(+) helper T lymphocyte, a cell type that is responsible for coordinating immune responses and modulating effector responses to foreign antigens. As part of their normal immune surveillance duties, these cells migrate actively within lymphoid tissues and can travel from inductive sites to effector sites in search of their cognate antigen. For CD4(+) T cells, there is an ongoing search for a unique peptide antigen presented in the context of class II MHC that can activate a proliferative or tolerogenic response. This iterative and continual probing and interrogation of other cells determine the outcome of immune responses. Recent studies in vitro have revealed that the viral infection program induces cell-cell interactions called virological synapses between infected and uninfected CD4(+) T cells. These long-lived, virally induced adhesive contacts greatly enhance the rate of productive infection and may be central to the spread of the virus in vivo. Here, we review aspects of this efficient mode of cell-to-cell infection and the implications for our understanding of HIV-1 pathogenesis.
Collapse
Affiliation(s)
- Benjamin M Dale
- Division of Infectious Disease, Department of Medicine, Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | |
Collapse
|
43
|
Sanchez N, Gallagher M, Lao N, Gallagher C, Clarke C, Doolan P, Aherne S, Blanco A, Meleady P, Clynes M, Barron N. MiR-7 triggers cell cycle arrest at the G1/S transition by targeting multiple genes including Skp2 and Psme3. PLoS One 2013; 8:e65671. [PMID: 23762407 PMCID: PMC3675065 DOI: 10.1371/journal.pone.0065671] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 04/28/2013] [Indexed: 12/26/2022] Open
Abstract
MiR-7 acts as a tumour suppressor in many cancers and abrogates proliferation of CHO cells in culture. In this study we demonstrate that miR-7 targets key regulators of the G1 to S phase transition, including Skp2 and Psme3, to promote increased levels of p27KIP and temporary growth arrest of CHO cells in the G1 phase. Simultaneously, the down-regulation of DNA repair-specific proteins via miR-7 including Rad54L, and pro-apoptotic regulators such as p53, combined with the up-regulation of anti-apoptotic factors like p-Akt, promoted cell survival while arrested in G1. Thus miR-7 can co-ordinate the levels of multiple genes and proteins to influence G1 to S phase transition and the apoptotic response in order to maintain cellular homeostasis. This work provides further mechanistic insight into the role of miR-7 as a regulator of cell growth in times of cellular stress.
Collapse
Affiliation(s)
- Noelia Sanchez
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Mark Gallagher
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Nga Lao
- Conway Institute, University College Dublin, Dublin, Ireland
| | - Clair Gallagher
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Colin Clarke
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Padraig Doolan
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Sinead Aherne
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Alfonso Blanco
- Conway Institute, University College Dublin, Dublin, Ireland
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Martin Clynes
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Niall Barron
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
- * E-mail:
| |
Collapse
|
44
|
Intercellular contact augments epidermal growth factor receptor (EGFR) and signal transducer and activator of transcription 3 (STAT3)-activation which increases podoplanin-expression in order to promote squamous cell carcinoma motility. Cell Signal 2013; 25:760-5. [DOI: 10.1016/j.cellsig.2012.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 12/18/2012] [Indexed: 01/13/2023]
|
45
|
Liu WM, Zhang F, Moshiach S, Zhou B, Huang C, Srinivasan K, Khurana S, Zheng Y, Lahti JM, Zhang XA. Tetraspanin CD82 inhibits protrusion and retraction in cell movement by attenuating the plasma membrane-dependent actin organization. PLoS One 2012; 7:e51797. [PMID: 23251627 PMCID: PMC3522597 DOI: 10.1371/journal.pone.0051797] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 11/12/2012] [Indexed: 11/18/2022] Open
Abstract
To determine how tetraspanin KAI1/CD82, a tumor metastasis suppressor, inhibits cell migration, we assessed which cellular events critical for motility are altered by KAI1/CD82 and how KAI1/CD82 regulates these events. We found that KAI1/CD82-expressing cells typically exhibited elongated cellular tails and diminished lamellipodia. Live imaging demonstrated that the polarized protrusion and retraction of the plasma membrane became deficient upon KAI1/CD82 expression. The deficiency in developing these motility-related cellular events was caused by poor formations of actin cortical network and stress fiber and by aberrant dynamics in actin organization. Rac1 activity was reduced by KAI1/CD82, consistent with the diminution of lamellipodia and actin cortical network; while the growth factor-stimulated RhoA activity was blocked by KAI1/CD82, consistent with the loss of stress fiber and attenuation in cellular retraction. Upon KAI1/CD82 expression, Rac effector cofilin was not enriched at the cell periphery to facilitate lamellipodia formation while Rho kinase exhibited a significantly lower activity leading to less retraction. Phosphatidylinositol 4, 5-biphosphate, which initiates actin polymerization from the plasma membrane, became less detectable at the cell periphery in KAI1/CD82-expressing cells. Moreover, KAI1/CD82-induced phenotypes likely resulted from the suppression of multiple signaling pathways such as integrin and growth factor signaling. In summary, at the cellular level KAI1/CD82 inhibited polarized protrusion and retraction events by disrupting actin reorganization; at the molecular level, KAI1/CD82 deregulated Rac1, RhoA, and their effectors cofilin and Rho kinase by perturbing the plasma membrane lipids.
Collapse
Affiliation(s)
- Wei M. Liu
- Vascular Biology and Cancer Centers and Departments of Medicine and Molecular Science, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Feng Zhang
- Vascular Biology and Cancer Centers and Departments of Medicine and Molecular Science, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Simon Moshiach
- Department of Genetics and Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Bin Zhou
- Vascular Biology and Cancer Centers and Departments of Medicine and Molecular Science, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Chao Huang
- Vascular Biology and Cancer Centers and Departments of Medicine and Molecular Science, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Kamalakkannan Srinivasan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Seema Khurana
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Yi Zheng
- Division of Experimental Hematology, Cincinnati Children's Hospital, Cincinnati, Ohio, United States of America
| | - Jill M. Lahti
- Department of Genetics and Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Xin A. Zhang
- Vascular Biology and Cancer Centers and Departments of Medicine and Molecular Science, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
46
|
Zhou Q, Li G, Deng XY, He XB, Chen LJ, Wu C, Shi Y, Wu KP, Mei LJ, Lu JX, Zhou NM. Activated human hydroxy-carboxylic acid receptor-3 signals to MAP kinase cascades via the PLC-dependent PKC and MMP-mediated EGFR pathways. Br J Pharmacol 2012; 166:1756-73. [PMID: 22289163 DOI: 10.1111/j.1476-5381.2012.01875.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE 3-Hydroxy-octanoate, recently identified as a ligand for, the orphan GPCR, HCA(3), is of particular interest given its ability to treat lipid disorders and atherosclerosis. Here we demonstrate the pathway of HCA(3)-mediated activation of ERK1/2. EXPERIMENTAL APPROACH Using CHO-K1 cells stably expressing HCA(3) receptors and A431 cells, a human epidermoid cell line with high levels of endogenous expression of functional HCA(3) receptors, HCA(3)-mediated activation of ERK1/2 was measured by Western blot. KEY RESULTS HCA(3)-mediated activation of ERK1/2 was rapid, peaking at 5 min, and was Pertussis toxin sensitive. Our data, obtained by time course analyses in combination with different kinase inhibitors, demonstrated that on agonist stimulation, HCA(3) receptors evoked ERK1/2 activation via two distinct pathways, the PLC/PKC pathway at early time points (≤ 2 min) and the MMP/ epidermal growth factor receptor (EGFR) transactivation pathway with a maximum response at 5 min. Furthermore, our present results also indicated that the βγ-subunits of the G(i) protein play a critical role in HCA(3)-activated ERK1/2 phosphorylation, whereas β-arrestins and Src were not required for ERK1/2 activation. CONCLUSIONS AND IMPLICATIONS We have described the molecular mechanisms underlying the coupling of human HCA(3) receptors to the ERK1/2 MAP kinase pathway in CHO-K1 and A431 cells, which implicate the G(i) protein-initiated, PLC/PKC -and platelet-derived growth factor receptor/EGFR transactivation-dependent pathways. These observations may provide new insights into the pharmacological effects and the physiological functions modulated by the HCA(3)-mediated activation of ERK1/2.
Collapse
Affiliation(s)
- Q Zhou
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Science, Wenzhou Medical College, Wenzhou, Zhejiang, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Zhu YZ, Luo Y, Cao MM, Liu Y, Liu XQ, Wang W, Wu DG, Guan M, Xu QQ, Ren H, Zhao P, Qi ZT. Significance of palmitoylation of CD81 on its association with tetraspanin-enriched microdomains and mediating hepatitis C virus cell entry. Virology 2012; 429:112-23. [PMID: 22560863 DOI: 10.1016/j.virol.2012.03.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 03/03/2012] [Indexed: 12/15/2022]
Abstract
CD81, a co-receptor for hepatitis C virus (HCV), is a member of the tetraspanin superfamily and is heavily palmitoylated in the juxtamembrane cysteine residues. Palmitoylation plays an important role in protein-protein interactions and association with cholesterol-rich domains of membranes. In this study, Huh7 cells expressing wild-type or palmitoylation-defective CD81 were generated to analyze whether palmitoylation of CD81 is involved in HCV cell entry. Our data showed that de-palmitoylation of CD81 dramatically reduced its association with tetraspanin CD151, but did not influence CD81 partition in detergent-resistant membranes. Moreover, de-palmitoylated CD81 decreased the host cell susceptibility to HCV. Notably, CD151-specific antibodies and siRNA inhibited HCV cell entry, and detachment of CD81 with CD151 decreased the lateral movement of virus particle/CD81 complex to areas of cell-cell contact. These results suggest that palmitoylation of CD81 should facilitate HCV entry, at least in part, by regulating the association of CD81 with tetraspanin-enriched microdomains.
Collapse
Affiliation(s)
- Yong-Zhe Zhu
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Meleady P, Gallagher M, Clarke C, Henry M, Sanchez N, Barron N, Clynes M. Impact of miR-7 over-expression on the proteome of Chinese hamster ovary cells. J Biotechnol 2012; 160:251-62. [PMID: 22445466 DOI: 10.1016/j.jbiotec.2012.03.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 03/05/2012] [Accepted: 03/07/2012] [Indexed: 12/31/2022]
Abstract
MicroRNAs play critical roles in the regulation of biological processes such as growth, apoptosis, productivity and secretion thus representing a potential route toward enhancing desirable characteristics of mammalian cells for biopharmaceutical production. We have previously found that miR-7 over-expression significantly inhibits the growth of CHO-SEAP cells without impacting cellular viability, with an associated increase in normalised productivity. Understanding the biological basis of this effect might open the way to new strategies for bioprocess-relevant growth regulation. In this study we have carried out a quantitative label-free LC-MS profiling study of proteins exhibiting altered levels following over-expression of miR-7 to gain insights into potential mechanisms involved in the observed phenotype. From the analysis we found 93 proteins showing decreased levels and 74 proteins with increased levels following over-expression of miR-7. Pathway analysis suggests that proteins involved in protein translation (e.g. ribosomal proteins), RNA and DNA processing (including histones) are enriched in the list of proteins showing decreased expression. Proteins involved in protein folding and secretion were found to be up-regulated following miR-7 over-expression. In silico bioinformatic analysis using miRWalk, which combined the output from 6 selected miRNA target prediction algorithms, was used to evaluate if any of the down-regulated proteins were potential direct targets of miR-7. Two genes, stathmin and catalase, which both have known roles in the regulation of cellular growth, were found to overlap a number of the predictive target database searches in both mouse and rat, and are likely to be possible direct targets of miR-7 in CHO cells. This is the first report investigating the impact of a miRNA on the proteome of CHO cells.
Collapse
Affiliation(s)
- Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
| | | | | | | | | | | | | |
Collapse
|
49
|
Yáñez-Mó M, Gutiérrez-López MD, Cabañas C. Functional interplay between tetraspanins and proteases. Cell Mol Life Sci 2011; 68:3323-35. [PMID: 21687991 PMCID: PMC11114976 DOI: 10.1007/s00018-011-0746-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Revised: 05/04/2011] [Accepted: 05/30/2011] [Indexed: 12/14/2022]
Abstract
Several recent publications have described examples of physical and functional interations between tetraspanins and specific membrane proteases belonging to the TM-MMP and α-(ADAMs) and γ-secretases families. Collectively, these examples constitute an emerging body of evidence supporting the notion that tetraspanin-enriched microdomains (TEMs) represent functional platforms for the regulation of key cellular processes including the release of surface protein ectodomains ("shedding"), regulated intramembrane proteolysis ("RIPing") and matrix degradation and assembly. These cellular processes in turn play a crucial role in an array of physiological and pathological phenomena. Thus, TEMs may represent new therapeutical targets that may simultaneously affect the proteolytic activity of different enzymes and their substrates. Agonistic or antagonistic antibodies and blocking soluble peptides corresponding to tetraspanin functional regions may offer new opportunities in the treatment of pathologies such as chronic inflammation, cancer, or Alzheimer's disease. In this review article, we will discuss all these aspects of functional regulation of protease activities by tetraspanins.
Collapse
Affiliation(s)
- María Yáñez-Mó
- Servicio de Inmunología, Hospital de la Princesa, Instituto de Investigación Sanitaria Princesa, 28006 Madrid, Spain
| | | | - Carlos Cabañas
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), 28049 Madrid, Spain
- Facultad de Medicina, Departamento de Microbiología I (Inmunología), UCM, 28040 Madrid, Spain
| |
Collapse
|
50
|
Gutiérrez-López MD, Gilsanz A, Yáñez-Mó M, Ovalle S, Lafuente EM, Domínguez C, Monk PN, González-Alvaro I, Sánchez-Madrid F, Cabañas C. The sheddase activity of ADAM17/TACE is regulated by the tetraspanin CD9. Cell Mol Life Sci 2011; 68:3275-92. [PMID: 21365281 PMCID: PMC11115118 DOI: 10.1007/s00018-011-0639-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 12/23/2010] [Accepted: 01/20/2011] [Indexed: 01/06/2023]
Abstract
ADAM17/TACE is a metalloproteinase responsible for the shedding of the proinflammatory cytokine TNF-α and many other cell surface proteins involved in development, cell adhesion, migration, differentiation, and proliferation. Despite the important biological function of ADAM17, the mechanisms of regulation of its metalloproteinase activity remain largely unknown. We report here that the tetraspanin CD9 and ADAM17 partially co-localize on the surface of endothelial and monocytic cells. In situ proximity ligation, co-immunoprecipitation, crosslinking, and pull-down experiments collectively demonstrate a direct association between these molecules. Functional studies reveal that treatment with CD9-specific antibodies or neoexpression of CD9 exert negative regulatory effects on ADAM17 sheddase activity. Conversely, CD9 silencing increased the activity of ADAM17 against its substrates TNF-α and ICAM-1. Taken together, our results show that CD9 associates with ADAM17 and, through this interaction, negatively regulates the sheddase activity of ADAM17.
Collapse
Affiliation(s)
- Maria Dolores Gutiérrez-López
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Nicolás Cabrera 1, Campus de Cantoblanco, 28049 Madrid, Spain
- Present Address: Departamento de Farmacología, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain
| | - Alvaro Gilsanz
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Nicolás Cabrera 1, Campus de Cantoblanco, 28049 Madrid, Spain
| | - María Yáñez-Mó
- Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria Princesa, 28006 Madrid, Spain
| | - Susana Ovalle
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Nicolás Cabrera 1, Campus de Cantoblanco, 28049 Madrid, Spain
| | - Esther M. Lafuente
- Departamento de Microbiología I (Inmunología), Facultad de Medicina, UCM, 28040 Madrid, Spain
| | - Carmen Domínguez
- Servicio de Reumatología, Hospital Universitario de La Princesa, 28006 Madrid, Spain
| | - Peter N. Monk
- University of Sheffield Medical School, Sheffield S10 2RX, Sheffield, United Kingdom
| | | | - Francisco Sánchez-Madrid
- Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria Princesa, 28006 Madrid, Spain
- Departamento de Biología Vascular e Inflamación, CNIC, 28029 Madrid, Spain
| | - Carlos Cabañas
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Nicolás Cabrera 1, Campus de Cantoblanco, 28049 Madrid, Spain
- Departamento de Microbiología I (Inmunología), Facultad de Medicina, UCM, 28040 Madrid, Spain
| |
Collapse
|