1
|
Wang D, Yu L. Migrasome biogenesis: when biochemistry meets biophysics on membranes. Trends Biochem Sci 2024; 49:829-840. [PMID: 38945731 DOI: 10.1016/j.tibs.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/17/2024] [Accepted: 06/07/2024] [Indexed: 07/02/2024]
Abstract
Migrasomes, newly identified organelles, play crucial roles in intercellular communication, contributing to organ development and angiogenesis. These vesicles, forming on retraction fibers of migrating cells, showcase a sophisticated architecture. Recent research reveals that migrasome biogenesis is a complicated and highly regulated process. This review summarizes the mechanisms governing migrasome formation, proposing a model in which biogenesis is understood through the lens of membrane microdomain assembly. It underscores the critical interplay between biochemistry and biophysics. The biogenesis unfolds in three distinct stages: nucleation, maturation, and expansion, each characterized by unique morphological, biochemical, and biophysical features. We also explore the broader implications of migrasome research in membrane biology and outline key unanswered questions that represent important directions for future investigation.
Collapse
Affiliation(s)
- Dongju Wang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Li Yu
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
2
|
Wong AH, Nga ME, Chin CY, Tai YK, Wong HC, Soo R, An O, Yang H, Seet JE, Lim YC, Tam JKC, Tran T. Impact of CD151 overexpression on prognosis and therapy in non-small cell lung cancer patients lacking EGFR mutations. Cell Prolif 2024; 57:e13708. [PMID: 38982031 PMCID: PMC11503249 DOI: 10.1111/cpr.13708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/08/2024] [Accepted: 06/18/2024] [Indexed: 07/11/2024] Open
Abstract
This study investigates CD151, a protein linked to cancer progression, in non-small cell lung cancer (NSCLC) patients without epidermal growth factor receptor (EGFR) mutations. These patients often have limited treatment options. The study used retrospective analysis to examine 157 adenocarcinoma biopsy specimens and 199 patient cases from The Cancer Genome Atlas, correlating CD151 expression with patient survival. Cellular studies revealed that CD151 interacts with EGFR, influencing epidermal growth factor (EGF)-induced cell proliferation and the effectiveness of the EGFR inhibitor, erlotinib. A strong association was found between CD151 expression and EGFR mutation status. High CD151 expression in the absence of EGFR mutations is correlated with poorer survival outcomes. Biological assays showed that CD151 colocalizes and associates with EGFR, playing a crucial role in regulating EGF-induced cell proliferation via the AKT and ERK1/2 pathways. Importantly, CD151 expression was found to influence the anti-proliferative effects of the EGFR tyrosine kinase inhibitor, erlotinib. High CD151 expression, in the absence of EGFR mutations, was associated with poorer survival outcomes. It could serve as a potential prognostic marker and influence cellular responses to EGFR-targeted treatments. This study highlights CD151 as a potential novel target for therapeutic intervention in NSCLC, especially in populations lacking EGFR mutations.
Collapse
Affiliation(s)
- Amanda Huee‐Ping Wong
- Department of Physiology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Min En Nga
- Department of PathologyNational University HospitalSingaporeSingapore
- Department of Pathology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Chin Yein Chin
- Department of Physiology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Yee Kit Tai
- Department of Physiology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Hung Chew Wong
- Department of Biostatistics, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Ross Soo
- Department of Haematology‐OncologyNational University HospitalSingaporeSingapore
| | - Omer An
- Cancer Science Institute of SingaporeNational University of SingaporeSingaporeSingapore
| | - Henry Yang
- Cancer Science Institute of SingaporeNational University of SingaporeSingaporeSingapore
| | - Ju Ee Seet
- Department of PathologyNational University HospitalSingaporeSingapore
| | - Yaw Chyn Lim
- Department of Pathology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Cancer Science Institute of SingaporeNational University of SingaporeSingaporeSingapore
| | - John Kit Chung Tam
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Cardiac, Thoracic and Vascular Surgery, National University Heart Centre, SingaporeNational University Health SystemSingaporeSingapore
| | - Thai Tran
- Department of Physiology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Infectious Disease Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| |
Collapse
|
3
|
Susa KJ, Kruse AC, Blacklow SC. Tetraspanins: structure, dynamics, and principles of partner-protein recognition. Trends Cell Biol 2024; 34:509-522. [PMID: 37783654 PMCID: PMC10980598 DOI: 10.1016/j.tcb.2023.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/18/2023] [Accepted: 09/07/2023] [Indexed: 10/04/2023]
Abstract
Tetraspanins are a large, highly conserved family of four-pass transmembrane (TM) proteins that play critical roles in a variety of essential cellular functions, including cell migration, protein trafficking, maintenance of membrane integrity, and regulation of signal transduction. Tetraspanins carry out these biological functions primarily by interacting with partner proteins. Here, we summarize significant advances that have revealed fundamental principles underpinning structure-function relationships in tetraspanins. We first review the structural features of tetraspanin ectodomains and full-length apoproteins, and then discuss how recent structural studies of tetraspanin complexes have revealed plasticity in partner-protein recognition that enables tetraspanins to bind to remarkably different protein families, viral proteins, and antibody fragments. Finally, we discuss major questions and challenges that remain in studying tetraspanin complexes.
Collapse
Affiliation(s)
- Katherine J Susa
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158, USA.
| | - Andrew C Kruse
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| | - Stephen C Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA.
| |
Collapse
|
4
|
Bae E, Huang P, Müller-Greven G, Hambardzumyan D, Sloan AE, Nowacki AS, Marko N, Carlin CR, Gladson CL. Integrin α3β1 promotes vessel formation of glioblastoma-associated endothelial cells through calcium-mediated macropinocytosis and lysosomal exocytosis. Nat Commun 2022; 13:4268. [PMID: 35879332 PMCID: PMC9314429 DOI: 10.1038/s41467-022-31981-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 07/12/2022] [Indexed: 12/13/2022] Open
Abstract
Therapeutic targeting of angiogenesis in glioblastoma has yielded mixed outcomes. Investigation of tumor-associated angiogenesis has focused on the factors that stimulate the sprouting, migration, and hyperproliferation of the endothelial cells. However, little is known regarding the processes underlying the formation of the tumor-associated vessels. To address this issue, we investigated vessel formation in CD31+ cells isolated from human glioblastoma tumors. The results indicate that overexpression of integrin α3β1 plays a central role in the promotion of tube formation in the tumor-associated endothelial cells in glioblastoma. Blocking α3β1 function reduced sprout and tube formation in the tumor-associated endothelial cells and vessel density in organotypic cultures of glioblastoma. The data further suggest a mechanistic model in which integrin α3β1-promoted calcium influx stimulates macropinocytosis and directed maturation of the macropinosomes in a manner that promotes lysosomal exocytosis during nascent lumen formation. Altogether, our data indicate that integrin α3β1 may be a therapeutic target on the glioblastoma vasculature.
Collapse
Affiliation(s)
- Eunnyung Bae
- Department of Cancer Biology, Cleveland, Clinic, Cleveland, OH, USA
| | - Ping Huang
- Department of Cancer Biology, Cleveland, Clinic, Cleveland, OH, USA
| | | | - Dolores Hambardzumyan
- Departments of Oncological Sciences and Neurosurgery, Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | - Andrew Edward Sloan
- Department of Neurosurgery, Seidman Cancer Center, Cleveland, OH, USA
- University Hospital-Cleveland Medical Center and the Case Comprehensive Cancer Center, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Amy S Nowacki
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Nicholas Marko
- Department of Neurosurgery, LewisGale Medical Center, Salem, VA, USA
| | - Cathleen R Carlin
- University Hospital-Cleveland Medical Center and the Case Comprehensive Cancer Center, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
- Department of Molecular Biology and Microbiology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Candece L Gladson
- Department of Cancer Biology, Cleveland, Clinic, Cleveland, OH, USA.
- University Hospital-Cleveland Medical Center and the Case Comprehensive Cancer Center, Case Western Reserve University, School of Medicine, Cleveland, OH, USA.
- The Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
5
|
Thölmann S, Seebach J, Otani T, Florin L, Schnittler H, Gerke V, Furuse M, Ebnet K. JAM-A interacts with α3β1 integrin and tetraspanins CD151 and CD9 to regulate collective cell migration of polarized epithelial cells. Cell Mol Life Sci 2022; 79:88. [PMID: 35067832 PMCID: PMC8784505 DOI: 10.1007/s00018-022-04140-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/22/2021] [Accepted: 01/10/2022] [Indexed: 01/23/2023]
Abstract
AbstractJunctional adhesion molecule (JAM)-A is a cell adhesion receptor localized at epithelial cell–cell contacts with enrichment at the tight junctions. Its role during cell–cell contact formation and epithelial barrier formation has intensively been studied. In contrast, its role during collective cell migration is largely unexplored. Here, we show that JAM-A regulates collective cell migration of polarized epithelial cells. Depletion of JAM-A in MDCK cells enhances the motility of singly migrating cells but reduces cell motility of cells embedded in a collective by impairing the dynamics of cryptic lamellipodia formation. This activity of JAM-A is observed in cells grown on laminin and collagen-I but not on fibronectin or vitronectin. Accordingly, we find that JAM-A exists in a complex with the laminin- and collagen-I-binding α3β1 integrin. We also find that JAM-A interacts with tetraspanins CD151 and CD9, which both interact with α3β1 integrin and regulate α3β1 integrin activity in different contexts. Mapping experiments indicate that JAM-A associates with α3β1 integrin and tetraspanins CD151 and CD9 through its extracellular domain. Similar to depletion of JAM-A, depletion of either α3β1 integrin or tetraspanins CD151 and CD9 in MDCK cells slows down collective cell migration. Our findings suggest that JAM-A exists with α3β1 integrin and tetraspanins CD151 and CD9 in a functional complex to regulate collective cell migration of polarized epithelial cells.
Collapse
Affiliation(s)
- Sonja Thölmann
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity", Institute of Medical Biochemistry, ZMBE, University of Münster, Von-Esmarch-Str. 56, 48149, Münster, Germany
- Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Jochen Seebach
- Institute of Anatomy and Vascular Biology, University of Münster, Münster, Germany
- Cells-in-Motion Interfaculty Center, University of Münster, 48149, Münster, Germany
| | - Tetsuhisa Otani
- Division of Cell Structure, National Institute for Physiological Sciences, National Institute of Natural Sciences, Okazaki, Aichi, Japan
| | - Luise Florin
- Institute for Virology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Hans Schnittler
- Institute of Anatomy and Vascular Biology, University of Münster, Münster, Germany
- Cells-in-Motion Interfaculty Center, University of Münster, 48149, Münster, Germany
| | - Volker Gerke
- Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
- Cells-in-Motion Interfaculty Center, University of Münster, 48149, Münster, Germany
| | - Mikio Furuse
- Division of Cell Structure, National Institute for Physiological Sciences, National Institute of Natural Sciences, Okazaki, Aichi, Japan
| | - Klaus Ebnet
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity", Institute of Medical Biochemistry, ZMBE, University of Münster, Von-Esmarch-Str. 56, 48149, Münster, Germany.
- Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany.
- Cells-in-Motion Interfaculty Center, University of Münster, 48149, Münster, Germany.
| |
Collapse
|
6
|
Luo W, Lin Z, Chen J, Chen G, Zhang S, Liu M, Li H, He D, Liang S, Luo Q, Zhang D, Nie Q, Zhang X. TMEM182 interacts with integrin beta 1 and regulates myoblast differentiation and muscle regeneration. J Cachexia Sarcopenia Muscle 2021; 12:1704-1723. [PMID: 34427057 PMCID: PMC8718073 DOI: 10.1002/jcsm.12767] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/28/2021] [Accepted: 07/10/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Transmembrane proteins are vital for intercellular signalling and play important roles in the control of cell fate. However, their physiological functions and mechanisms of action in myogenesis and muscle disorders remain largely unexplored. It has been found that transmembrane protein 182 (TMEM182) is dramatically up-regulated during myogenesis, but its detailed functions remain unclear. This study aimed to analyse the function of TMEM182 during myogenesis and muscle regeneration. METHODS RNA sequencing, quantitative real-time polymerase chain reaction, and immunofluorescence approaches were used to analyse TMEM182 expression during myoblast differentiation. A dual-luciferase reporter assay was used to identify the promoter region of the TMEM182 gene, and a chromatin immunoprecipitation assay was used to investigate the regulation TMEM182 transcription by MyoD. We used chickens and TMEM182-knockout mice as in vivo models to examine the function of TMEM182 in muscle growth and muscle regeneration. Chickens and mouse primary myoblasts were used to extend the findings to in vitro effects on myoblast differentiation and fusion. Co-immunoprecipitation and mass spectrometry were used to identify the interaction between TMEM182 and integrin beta 1 (ITGB1). The molecular mechanism by which TMEM182 regulates myogenesis and muscle regeneration was examined by Transwell migration, cell wound healing, adhesion, glutathione-S-transferse pull down, protein purification, and RNA immunoprecipitation assays. RESULTS TMEM182 was specifically expressed in skeletal muscle and adipose tissue and was regulated at the transcriptional level by the myogenic regulatory factor MyoD1. Functionally, TMEM182 inhibited myoblast differentiation and fusion. The in vivo studies indicated that TMEM182 induced muscle fibre atrophy and delayed muscle regeneration. TMEM182 knockout in mice led to significant increases in body weight, muscle mass, muscle fibre number, and muscle fibre diameter. Skeletal muscle regeneration was accelerated in TMEM182-knockout mice. Furthermore, we revealed that the inhibitory roles of TMEM182 in skeletal muscle depend on ITGB1, an essential membrane receptor involved in cell adhesion and muscle formation. TMEM182 directly interacted with ITGB1, and this interaction required an extracellular hybrid domain of ITGB1 (aa 387-470) and a conserved region (aa 52-62) within the large extracellular loop of TMEM182. Mechanistically, TMEM182 modulated ITGB1 activation by coordinating the association between ITGB1 and laminin and regulating the intracellular signalling of ITGB1. Myogenic deletion of TMEM182 increased the binding activity of ITGB1 to laminin and induced the activation of the FAK-ERK and FAK-Akt signalling axes during myogenesis. CONCLUSIONS Our data reveal that TMEM182 is a novel negative regulator of myogenic differentiation and muscle regeneration.
Collapse
Affiliation(s)
- Wen Luo
- Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou, China.,Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, China.,Department of Orthaepedics and Traumatology, The Chinese University of Hong Kong, Hongkong
| | - Zetong Lin
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, China
| | - Jiahui Chen
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, China
| | - Genghua Chen
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, China
| | - Siyu Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, China
| | - Manqing Liu
- Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou, China.,Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, China
| | - Hongmei Li
- Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou, China.,Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, China
| | - Danlin He
- Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou, China.,Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, China
| | - Shaodong Liang
- Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou, China.,Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, China
| | - Qingbin Luo
- Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou, China.,Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, China
| | - Dexiang Zhang
- Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou, China.,Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, China
| | - Qinghua Nie
- Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou, China.,Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, China.,State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, South China Agricultural University, Guangzhou, China
| | - Xiquan Zhang
- Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou, China.,Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, China.,State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, South China Agricultural University, Guangzhou, China
| |
Collapse
|
7
|
Li D, Lai W, Wang Q, Xiang Z, Nan X, Yang X, Fang Q. CD151 enrichment in exosomes of luminal androgen receptor breast cancer cell line contributes to cell invasion. Biochimie 2021; 189:65-75. [PMID: 34157361 DOI: 10.1016/j.biochi.2021.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/26/2021] [Accepted: 06/17/2021] [Indexed: 02/03/2023]
Abstract
Breast cancer is the most common and highly heterogeneous disease in women worldwide. Given the challenges in the treatment of advanced metastatic breast cancer, it is necessary to understand the molecular mechanisms related to disease progression. Exosomes play various roles in the progression of tumors, including promoting the invasion and advancing the distant metastasis. To study the molecular mechanisms related to the progression of luminal androgen receptor (LAR) breast cancer, we first isolated exosomes of MDA-MB-453 cells, a representative cell line of LAR. Through quantitative proteomic analysis, we identified 180 proteins specifically enriched in exosomes after comparing with those in cells, microvesicles, and the 150K supernatant. Among these, CD151, a protein involved in the regulation of cell motility was the most enriched one. CD151-knockdown exosomes reduced the invasion ability of the recipient breast cancer cell and lowered the phosphorylation level of tyrosine-protein kinase Lck, indicating that the invasion of LAR breast cancer may be due to CD151-enriched exosomes. Our work reports for the first time that CD151 was highly abundant in the exosomes of MDA-MB-453 cells and expands the understanding of the development process of LAR subtype, suggesting CD151 may be a potential candidate for the treatment of LAR breast cancer.
Collapse
Affiliation(s)
- Dan Li
- Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100190, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Wenjia Lai
- Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Qingsong Wang
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing, 100871, PR China
| | - Zhichu Xiang
- Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Xiaohui Nan
- Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100190, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Xiaoliang Yang
- Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Qiaojun Fang
- Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100190, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing, 100190, PR China.
| |
Collapse
|
8
|
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: 1.7] [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
|
9
|
The cell surface hyaluronidase TMEM2 regulates cell adhesion and migration via degradation of hyaluronan at focal adhesion sites. J Biol Chem 2021; 296:100481. [PMID: 33647313 PMCID: PMC8042168 DOI: 10.1016/j.jbc.2021.100481] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/18/2021] [Accepted: 02/25/2021] [Indexed: 12/31/2022] Open
Abstract
The extracellular matrix (ECM) plays an important role in maintaining tissue homeostasis and poses a significant physical barrier to in vivo cell migration. Accordingly, as a means of enhancing tissue invasion, tumor cells use matrix metalloproteinases to degrade ECM proteins. However, the in vivo ECM is comprised not only of proteins but also of a variety of nonprotein components. Hyaluronan (HA), one of the most abundant nonprotein components of the interstitial ECM, forms a gel-like antiadhesive barrier that is impenetrable to particulate matter and cells. Mechanisms by which tumor cells penetrate the HA barrier have not been addressed. Here, we demonstrate that transmembrane protein 2 (TMEM2), the only known transmembrane hyaluronidase, is the predominant mediator of contact-dependent HA degradation and subsequent integrin-mediated cell–substrate adhesion. We show that a variety of tumor cells are able to eliminate substrate-bound HA in a tightly localized pattern corresponding to the distribution of focal adhesions (FAs) and stress fibers. This FA-targeted HA degradation is mediated by TMEM2, which itself is localized at site of FAs. TMEM2 depletion inhibits the ability of tumor cells to attach and migrate in an HA-rich environment. Importantly, TMEM2 directly binds at least two integrins via interaction between extracellular domains. Our findings demonstrate a critical role for TMEM2-mediated HA degradation in the adhesion and migration of cells on HA-rich ECM substrates and provide novel insight into the early phase of FA formation.
Collapse
|
10
|
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: 3.8] [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
|
11
|
Takheaw N, Pata S, Laopajon W, Roytrakul S, Kasinrerk W. The presence of membrane bound CD99 ligands on leukocyte surface. BMC Res Notes 2020; 13:496. [PMID: 33092634 PMCID: PMC7583281 DOI: 10.1186/s13104-020-05347-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 10/16/2020] [Indexed: 11/30/2022] Open
Abstract
Objective CD99, a leukocyte surface molecule, reportedly plays an important role in several cellular processes. However, the role of CD99 in T cell regulation remains unclear, as the CD99 ligand associated with T-cell regulation has not yet been identified. Our previous study showed that recombinant CD99 bound to CD99 ligands was expressed on monocytes, NK cells and dendritic cells. This interaction regulates the expression of IL-6 and TNF-α in CD3 + T cells following T cell activation. In the present study, we confirmed the presence of CD99 ligands in immune cells. Results A recombinant CD99-human IgG fusion protein, CD99HIgG, was produced and used to search for CD99 ligand expression in various hematopoietic cell lines. Among several cell lines, THP-1 monocytic cell line showed strong positive reaction for CD99HIgG, and CD99 and CD99 ligand complexes were pulled-down using a DTSSP cross-linker. The study demonstrated the presence of the membrane bound CD99 ligand, and CD99 ligand candidates were identified via LC–MS/MS. These results may be useful to further identify the CD99 ligands, and to fully comprehend the role of CD99 in immunoregulation.
Collapse
Affiliation(s)
- Nuchjira Takheaw
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Supansa Pata
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Witida Laopajon
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Sittiruk Roytrakul
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumtani, Thailand
| | - Watchara Kasinrerk
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand. .,Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
12
|
Jankovičová J, Neuerová Z, Sečová P, Bartóková M, Bubeníčková F, Komrsková K, Postlerová P, Antalíková J. Tetraspanins in mammalian reproduction: spermatozoa, oocytes and embryos. Med Microbiol Immunol 2020; 209:407-425. [PMID: 32424440 DOI: 10.1007/s00430-020-00676-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/02/2020] [Indexed: 12/21/2022]
Abstract
It is known that tetraspanin proteins are involved in many physiological somatic cell mechanisms. Additionally, research has indicated they also have a role in various infectious diseases and cancers. This review focuses on the molecular interactions underlying the tetraspanin web formation in gametes. Primarily, tetraspanins act in the reproductive tract as organizers of membrane complexes, which include the proteins involved in the contact and association of sperm and oocyte membranes. In addition, recent data shows that tetraspanins are likely to be involved in these processes in a complex way. In mammalian fertilization, an important role is attributed to CD molecules belonging to the tetraspanin superfamily, particularly CD9, CD81, CD151, and also CD63; mostly as part of extracellular vesicles, the significance of which and their potential in reproduction is being intensively investigated. In this article, we reviewed the existing knowledge regarding the expression of tetraspanins CD9, CD81, CD151, and CD63 in mammalian spermatozoa, oocytes, and embryos and their involvement in reproductive processes, including pathological events.
Collapse
Affiliation(s)
- Jana Jankovičová
- Laboratory of Reproductive Physiology, Center of Biosciences, Institute of Animal Biochemistry and Genetics, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Zdeňka Neuerová
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
- Faculty of Science, University of Hradec Králové, Hradec Králové, Czech Republic
| | - Petra Sečová
- Laboratory of Reproductive Physiology, Center of Biosciences, Institute of Animal Biochemistry and Genetics, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Michaela Bartóková
- Laboratory of Reproductive Physiology, Center of Biosciences, Institute of Animal Biochemistry and Genetics, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Filipa Bubeníčková
- Department of Veterinary Sciences, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Prague, Czech Republic
| | - Kateřina Komrsková
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
- Department of Zoology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Pavla Postlerová
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
- Department of Veterinary Sciences, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Prague, Czech Republic
| | - Jana Antalíková
- Laboratory of Reproductive Physiology, Center of Biosciences, Institute of Animal Biochemistry and Genetics, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| |
Collapse
|
13
|
Susa KJ, Seegar TCM, Blacklow SC, Kruse AC. A dynamic interaction between CD19 and the tetraspanin CD81 controls B cell co-receptor trafficking. eLife 2020; 9:e52337. [PMID: 32338599 PMCID: PMC7228769 DOI: 10.7554/elife.52337] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 04/26/2020] [Indexed: 12/20/2022] Open
Abstract
CD81 and its binding partner CD19 are core subunits of the B cell co-receptor complex. While CD19 belongs to the extensively studied Ig superfamily, CD81 belongs to a poorly understood family of four-pass transmembrane proteins called tetraspanins. Tetraspanins play important physiological roles by controlling protein trafficking and other processes. Here, we show that CD81 relies on its ectodomain to traffic CD19 to the cell surface. Moreover, the anti-CD81 antibody 5A6, which binds selectively to activated B cells, recognizes a conformational epitope on CD81 that is masked when CD81 is bound to CD19. Mutations of CD81 in this interface suppress its CD19 export activity. These data indicate that the CD81 - CD19 interaction is dynamically regulated upon B cell activation and this dynamism can be exploited to regulate B cell function. These results are not only valuable for understanding B cell biology, but also have important implications for understanding tetraspanin function generally.
Collapse
Affiliation(s)
- Katherine J Susa
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
| | - Tom CM Seegar
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
| | - Stephen C Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
- Dana Farber Cancer Institute, Department of Cancer BiologyBostonUnited States
| | - Andrew C Kruse
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
14
|
Finke J, Mikuličić S, Loster AL, Gawlitza A, Florin L, Lang T. Anatomy of a viral entry platform differentially functionalized by integrins α3 and α6. Sci Rep 2020; 10:5356. [PMID: 32210347 PMCID: PMC7093462 DOI: 10.1038/s41598-020-62202-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/10/2020] [Indexed: 12/26/2022] Open
Abstract
During cell invasion, human papillomaviruses use large CD151 patches on the cell surface. Here, we studied whether these patches are defined architectures with features for virus binding and/or internalization. Super-resolution microscopy reveals that the patches are assemblies of closely associated nanoclusters of CD151, integrin α3 and integrin α6. Integrin α6 is required for virus attachment and integrin α3 for endocytosis. We propose that CD151 organizes viral entry platforms with different types of integrin clusters for different functionalities. Since numerous viruses use tetraspanin patches, we speculate that this building principle is a blueprint for cell-surface architectures utilized by viral particles.
Collapse
Affiliation(s)
- Jérôme Finke
- Department of Membrane Biochemistry, Life & Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115, Bonn, Germany
| | - Snježana Mikuličić
- Institute for Virology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher Straße 67, 55131, Mainz, Germany
| | - Anna-Lena Loster
- Institute for Virology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher Straße 67, 55131, Mainz, Germany
| | - Alexander Gawlitza
- Institute for Virology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher Straße 67, 55131, Mainz, Germany
| | - Luise Florin
- Institute for Virology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher Straße 67, 55131, Mainz, Germany
| | - Thorsten Lang
- Department of Membrane Biochemistry, Life & Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115, Bonn, Germany.
| |
Collapse
|
15
|
Li H, Li J, Han R, Deng X, Shi J, Huang H, Hamad N, McCaughley A, Liu J, Wang C, Chen K, Wei D, Qiang J, Thatcher S, Wu Y, Liu C, Thibault O, Wei X, Chen S, Qian H, Zhou BP, Xu P, Yang XH. Deletion of tetraspanin CD151 alters the Wnt oncogene-induced mammary tumorigenesis: A cell type-linked function and signaling. Neoplasia 2019; 21:1151-1163. [PMID: 31783316 PMCID: PMC6888732 DOI: 10.1016/j.neo.2019.08.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 12/16/2022] Open
Abstract
Tetraspanin CD151 is increasingly implicated as a multifaceted mediator of cancer development and progression. Here we investigated the role of CD151 in breast cancer in the context of the Wnt oncogenic activation. Our data showed that removal of one or both of CD151 alleles in the MMTV-Wnt1 model significantly decreased the tumor-free survival of mice from 34 weeks on average to 22 weeks and 18 weeks, respectively. This effect coincided with an accelerated tumor growth and an increased number of Ki-67+ proliferative cells. Mechanistically, the CD151-deficient tumors were largely ER+, and exhibited hyperactivation of the Wnt pathway as reflected by a marked upregulation in β-catenin and Cyclin D1, and their target genes. In addition, E-cadherin displayed a cytosolic distribution and transcription factor Snail was markedly upregulated. Collectively, this data implies that CD151 suppresses the Wnt1-driven tumorigenesis, at least in part, via counteracting the epithelial-mesenchymal transition (EMT)-like program in luminal epithelial cells. Meanwhile, the proportion of tumor cells expressing CK5 or p63, the biomarkers of myoepithelial/basal cells, markedly decreased in the absence of CD151. This change was accompanied by a decreased invasiveness of tumors and their incompetence to form a long-term cell culture. Consistent with this basal cell-linked role, the CD151 downregulation impairs mammosphere formation in MCF-10A cells and the defect was rescued by re-expression of intact CD151 ORF, but not its integrin binding-defective mutant. Overall, our study suggests that CD151 is a key player in the Wnt oncogene-driven tumorigenesis and impacts breast cancer malignancy in a cell type-dependent manner.
Collapse
Affiliation(s)
- Hongxia Li
- Freshwater Fisheries Research Center, Ministry of Agriculture, and Fisheries College, Nanjing Agricultural University, Wuxi, Jiangsu Province, PR China; Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Jieming Li
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky, Lexington, KY; Center of Drug Discovery, China Pharmaceutical University, Nanjing, Jiangsu Province, PR China
| | - Rongbo Han
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky, Lexington, KY; Department of Oncology, Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Xinyu Deng
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Junfong Shi
- Department of Oncology, Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Huanhuan Huang
- Department of Oncology, Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Nevean Hamad
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Abigail McCaughley
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Jinpeng Liu
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Chi Wang
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Kuey Chen
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Dongping Wei
- Department of Oncology, Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Jun Qiang
- Freshwater Fisheries Research Center, Ministry of Agriculture, and Fisheries College, Nanjing Agricultural University, Wuxi, Jiangsu Province, PR China
| | - Sean Thatcher
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Yadi Wu
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Chunming Liu
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Olivier Thibault
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Xiaowei Wei
- Department of Oncology, Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Song Chen
- Institute of Medicinal Biotechnology, Jiangsu College of Nursing, Huaian, Jiangsu Province, PR China.
| | - Hai Qian
- Center of Drug Discovery, China Pharmaceutical University, Nanjing, Jiangsu Province, PR China.
| | - Binhua P Zhou
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky, Lexington, KY.
| | - Pao Xu
- Freshwater Fisheries Research Center, Ministry of Agriculture, and Fisheries College, Nanjing Agricultural University, Wuxi, Jiangsu Province, PR China.
| | - Xiuwei H Yang
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky, Lexington, KY.
| |
Collapse
|
16
|
Purushothaman G, Thiruvenkatam V. High Yield Expression of Recombinant CD151 in E. coli and a Structural Insight into Cholesterol Binding Domain. Mol Biotechnol 2019; 61:905-915. [PMID: 31541430 DOI: 10.1007/s12033-019-00212-3] [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: 10/26/2022]
Abstract
CD151 is an abundantly expressed eukaryotic transmembrane protein on the cell surface. It is involved in cell adhesion, angiogenesis and signal transduction as well in disease conditions such as cancer and viral infections. However, the molecular mechanism of CD151 activation is poorly understood due to the lack of structural information. By considering the difficulties in expressing the membrane protein in E. coli, herein we introduce the strategic design for the effective expression of recombinant CD151 protein in E. coli with high yield, that would aid for the structural studies. CD151 having four transmembrane domain (TMD's) along with small and a large extracellular loop (LEL) is constructed in parts to enhance the soluble expression of the protein attached with fusion tag. This has led to the high yield of the recombinant CD151 protein in the designed constructs. The recombinant CD151 protein is characterized and confirmed by western blot, CD and Mass peptide fingerprint. The molecular dynamics simulations (MDS) for the full-length CD151 shows conformational changes in the LEL of the protein in the presence and absence of cholesterol and indicate the certainty of closed and open conformation of CD151 based on cholesterol binding. The MDS results have led to the understanding of the possible underlying mechanism for the activation of the CD151 protein.
Collapse
Affiliation(s)
- Gayathri Purushothaman
- Discipline of Biological Engineering, Indian Institute of Technology Gandhinagar, Simkheda, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Vijay Thiruvenkatam
- Discipline of Biological Engineering, Indian Institute of Technology Gandhinagar, Simkheda, Palaj, Gandhinagar, 382355, Gujarat, India.
| |
Collapse
|
17
|
Takheaw N, Earwong P, Laopajon W, Pata S, Kasinrerk W. Interaction of CD99 and its ligand upregulates IL-6 and TNF-α upon T cell activation. PLoS One 2019; 14:e0217393. [PMID: 31120992 PMCID: PMC6532917 DOI: 10.1371/journal.pone.0217393] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 05/10/2019] [Indexed: 01/23/2023] Open
Abstract
CD99 has been reported to be involved in T cell regulation. CD99 ligand involvement in the regulation of T cell activation has been postulated. In this study, recombinant CD99 proteins were produced and used as a tool for determining the role of CD99 and its ligand interaction. Recombinant CD99 proteins induced the upregulation of IL-6 and TNF-α expression, but not IFN-γ, in anti-CD3 monoclonal antibody activated T cells. The cytokine alteration was not observed in unstimulated T cells indicating the cytokine upregulation required the signal from T cell activation. The upregulation of IL-6 and TNF-α was, in addition, observed in CD3- mononuclear cell population including monocytes and NK cells. The recombinant CD99 proteins, however, did not affect either CD25, CD69 or MHC class II expression or T cell proliferation, upon T cell activation. The CD99 ligands were demonstrated to be expressed on monocytes, NK cells and dendritic cells, but not on B and T cells. Our results indicated the presence of CD99 ligands on leukocyte surface. Interaction between CD99 and its ligands involves the regulation of cytokine production.
Collapse
Affiliation(s)
- Nuchjira Takheaw
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Papawadee Earwong
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Witida Laopajon
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Supansa Pata
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- * E-mail:
| | - Watchara Kasinrerk
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
18
|
Hwang S, Takimoto T, Hemler ME. Integrin-independent support of cancer drug resistance by tetraspanin CD151. Cell Mol Life Sci 2019; 76:1595-1604. [PMID: 30778617 PMCID: PMC6439156 DOI: 10.1007/s00018-019-03014-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/21/2018] [Accepted: 01/15/2019] [Indexed: 12/19/2022]
Abstract
Tetraspanin protein CD151 has typically been studied as binding partner and functional regulator of laminin-binding integrins. However, we show here that CD151 supports anti-cancer drug resistance independent of integrins. CD151 ablation sensitized multiple tumor cell types to several anti-cancer drugs (e.g., gefitinib and camptothecin), thus increasing apoptosis, as seen using cleaved caspase-3, cleaved PARP (poly (ADP-ribose) polymerase), annexin V, and propidium iodide staining assays. Drug sensitization due to CD151 ablation is integrin-independent, because, (1) effects occurred in cells when integrins were unengaged with ligand, (2) integrin ablation (α3 and α6 subunits) did not mimic effects of CD151 ablation, (3) the CD151QRD mutant, with diminished integrin association, and CD151WT (unmutated CD151) similarly reconstituted drug protection, and (4) treatment with anti-cancer drugs selectively upregulated intracellular nonintegrin-associated CD151 (NIA-CD151), consistent with its role in drug resistance. Together, these results suggest that upregulated CD151 expression may support not only typical integrin-dependent functions, but also integrin-independent survival of circulating (and possibly metastatic) cancer cells during anti-cancer drug therapy.
Collapse
Affiliation(s)
- Soonyean Hwang
- Department of Cancer Immunology and Virology, Rm SM-520C, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Takayuki Takimoto
- Department of Cancer Immunology and Virology, Rm SM-520C, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA.,Department of Internal Medicine, National Hospital Organization Kinki-Chuo Chest Medical Center 1180 Nagasone-cho, Kita-ku, Sakai, Osaka, 591-8555, Japan
| | - Martin E Hemler
- Department of Cancer Immunology and Virology, Rm SM-520C, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA.
| |
Collapse
|
19
|
Naudin C, Smith B, Bond DR, Dun MD, Scott RJ, Ashman LK, Weidenhofer J, Roselli S. Characterization of the early molecular changes in the glomeruli of Cd151 -/- mice highlights induction of mindin and MMP-10. Sci Rep 2017; 7:15987. [PMID: 29167507 PMCID: PMC5700190 DOI: 10.1038/s41598-017-15993-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/26/2017] [Indexed: 01/06/2023] Open
Abstract
In humans and FVB/N mice, loss of functional tetraspanin CD151 is associated with glomerular disease characterised by early onset proteinuria and ultrastructural thickening and splitting of the glomerular basement membrane (GBM). To gain insight into the molecular mechanisms associated with disease development, we characterised the glomerular gene expression profile at an early stage of disease progression in FVB/N Cd151 -/- mice compared to Cd151 +/+ controls. This study identified 72 up-regulated and 183 down-regulated genes in FVB/N Cd151 -/- compared to Cd151 +/+ glomeruli (p < 0.05). Further analysis highlighted induction of the matrix metalloprotease MMP-10 and the extracellular matrix protein mindin (encoded by Spon2) in the diseased FVB/N Cd151 -/- GBM that did not occur in the C57BL/6 diseased-resistant strain. Interestingly, mindin was also detected in urinary samples of FVB/N Cd151 -/- mice, underlining its potential value as a biomarker for glomerular diseases associated with GBM alterations. Gene set enrichment and pathway analysis of the microarray dataset showed enrichment in axon guidance and actin cytoskeleton signalling pathways as well as activation of inflammatory pathways. Given the known function of mindin, its early expression in the diseased GBM could represent a trigger of both further podocyte cytoskeletal changes and inflammation, thereby playing a key role in the mechanisms of disease progression.
Collapse
Affiliation(s)
- Crystal Naudin
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Newcastle, New South Wales, Australia.,Hunter Medical Research Institute, New Lambton, New South Wales, Australia.,Emory University, Atlanta, Georgia, USA
| | - Brian Smith
- School of Mathematics and Physical Sciences, University of Newcastle, Newcastle, New South Wales, Australia
| | - Danielle R Bond
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Newcastle, New South Wales, Australia.,Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| | - Matthew D Dun
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Newcastle, New South Wales, Australia.,Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| | - Rodney J Scott
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Newcastle, New South Wales, Australia.,Hunter Medical Research Institute, New Lambton, New South Wales, Australia.,Hunter Area Pathology Service, John Hunter Hospital, New Lambton, New South Wales, Australia
| | - Leonie K Ashman
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Newcastle, New South Wales, Australia.,Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| | - Judith Weidenhofer
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Newcastle, New South Wales, Australia.,Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| | - Séverine Roselli
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Newcastle, New South Wales, Australia. .,Hunter Medical Research Institute, New Lambton, New South Wales, Australia.
| |
Collapse
|
20
|
Earnest JT, Hantak MP, Li K, McCray PB, Perlman S, Gallagher T. The tetraspanin CD9 facilitates MERS-coronavirus entry by scaffolding host cell receptors and proteases. PLoS Pathog 2017; 13:e1006546. [PMID: 28759649 PMCID: PMC5552337 DOI: 10.1371/journal.ppat.1006546] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/10/2017] [Accepted: 07/21/2017] [Indexed: 01/27/2023] Open
Abstract
Infection by enveloped coronaviruses (CoVs) initiates with viral spike (S) proteins binding to cellular receptors, and is followed by proteolytic cleavage of receptor-bound S proteins, which prompts S protein-mediated virus-cell membrane fusion. Infection therefore requires close proximity of receptors and proteases. We considered whether tetraspanins, scaffolding proteins known to facilitate CoV infections, hold receptors and proteases together on cell membranes. Using knockout cell lines, we found that the tetraspanin CD9, but not the tetraspanin CD81, formed cell-surface complexes of dipeptidyl peptidase 4 (DPP4), the MERS-CoV receptor, and the type II transmembrane serine protease (TTSP) member TMPRSS2, a CoV-activating protease. This CD9-facilitated condensation of receptors and proteases allowed MERS-CoV pseudoviruses to enter cells rapidly and efficiently. Without CD9, MERS-CoV viruses were not activated by TTSPs, and they trafficked into endosomes to be cleaved much later and less efficiently by cathepsins. Thus, we identified DPP4:CD9:TTSP as the protein complexes necessary for early, efficient MERS-CoV entry. To evaluate the importance of these complexes in an in vivo CoV infection model, we used recombinant Adenovirus 5 (rAd5) vectors to express human DPP4 in mouse lungs, thereby sensitizing the animals to MERS-CoV infection. When the rAd5-hDPP4 vectors co-expressed small RNAs silencing Cd9 or Tmprss2, the animals were significantly less susceptible, indicating that CD9 and TMPRSS2 facilitated robust in vivo MERS-CoV infection of mouse lungs. Furthermore, the S proteins of virulent mouse-adapted MERS-CoVs acquired a CD9-dependent cell entry character, suggesting that CD9 is a selective agent in the evolution of CoV virulence.
Collapse
Affiliation(s)
- James T. Earnest
- Department of Microbiology and Immunology, Loyola University Medical Center, Maywood, IL, United States of America
| | - Michael P. Hantak
- Department of Microbiology and Immunology, Loyola University Medical Center, Maywood, IL, United States of America
| | - Kun Li
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
| | - Paul B. McCray
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
| | - Stanley Perlman
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
- Department of Microbiology, University of Iowa, Iowa City, IA, United States of America
| | - Tom Gallagher
- Department of Microbiology and Immunology, Loyola University Medical Center, Maywood, IL, United States of America
- * E-mail:
| |
Collapse
|
21
|
Joint features and complementarities of Tspan8 and CD151 revealed in knockdown and knockout models. Biochem Soc Trans 2017; 45:437-447. [PMID: 28408484 DOI: 10.1042/bst20160298] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/04/2017] [Accepted: 02/13/2017] [Indexed: 02/06/2023]
Abstract
Tetraspanins are highly conserved 4-transmembrane proteins which form molecular clusters with a large variety of transmembrane and cytosolic proteins. By these associations tetraspanins are engaged in a multitude of biological processes. Furthermore, tetraspanin complexes are located in specialized microdomains, called tetraspanin-enriched microdomains (TEMs). TEMs provide a signaling platform and are poised for invagination and vesicle formation. These vesicles can be released as exosomes (Exo) and are important in cell contact-independent intercellular communication. Here, we summarize emphasizing knockdown and knockout models' pathophysiological joint and selective activities of CD151 and Tspan8, and discuss the TEM-related engagement of CD151 and Tspan8 in Exo activities.
Collapse
|
22
|
Saint-Pol J, Billard M, Dornier E, Eschenbrenner E, Danglot L, Boucheix C, Charrin S, Rubinstein E. New insights into the tetraspanin Tspan5 using novel monoclonal antibodies. J Biol Chem 2017; 292:9551-9566. [PMID: 28428248 DOI: 10.1074/jbc.m116.765669] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 03/29/2017] [Indexed: 01/02/2023] Open
Abstract
Tspan5 is a member of a subgroup of tetraspanins referred to as TspanC8. These tetraspanins directly interact with the metalloprotease ADAM10, regulate its exit from the endoplasmic reticulum and subsequent trafficking, and differentially regulate its ability to cleave various substrates and activate Notch signaling. The study of Tspan5 has been limited by the lack of good antibodies. This study provides new insights into Tspan5 using new monoclonal antibodies (mAbs), including two mAbs recognizing both Tspan5 and the highly similar tetraspanin Tspan17. Using these mAbs, we show that endogenous Tspan5 associates with ADAM10 in human cell lines and in mouse tissues where it is the most abundant, such as the brain, the lung, the kidney, or the intestine. We also uncover two TspanC8-specific motifs in the large extracellular domain of Tspan5 that are important for ADAM10 interaction and exit from the endoplasmic reticulum. One of the anti-Tspan5 mAbs does not recognize Tspan5 associated with ADAM10, providing a convenient way to measure the fraction of Tspan5 not associated with ADAM10. This fraction is minor in the cell lines tested, and it increases upon transfection of cells with TspanC8 tetraspanins such as Tspan15 or Tspan33 that inhibit Notch signaling. Finally, two antibodies inhibit ligand-induced Notch signaling, and this effect is stronger in cells depleted of the TspanC8 tetraspanin Tspan14, further indicating that Tspan5 and Tspan14 can compensate for each other in Notch signaling.
Collapse
Affiliation(s)
- Julien Saint-Pol
- From Inserm, U935, F-94807 Villejuif.,the Université Paris-Sud, Institut André Lwoff, F-94807 Villejuif
| | - Martine Billard
- From Inserm, U935, F-94807 Villejuif.,the Université Paris-Sud, Institut André Lwoff, F-94807 Villejuif
| | - Emmanuel Dornier
- the Université Paris-Sud, Institut André Lwoff, F-94807 Villejuif.,Inserm, U1004, F-94807 Villejuif
| | - Etienne Eschenbrenner
- From Inserm, U935, F-94807 Villejuif.,the Université Paris-Sud, Institut André Lwoff, F-94807 Villejuif
| | - Lydia Danglot
- the CNRS, UMR7592, Université Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, F-75205 Paris, and.,Inserm, ERL U950, 75205 Paris, France
| | - Claude Boucheix
- From Inserm, U935, F-94807 Villejuif.,the Université Paris-Sud, Institut André Lwoff, F-94807 Villejuif
| | - Stéphanie Charrin
- From Inserm, U935, F-94807 Villejuif.,the Université Paris-Sud, Institut André Lwoff, F-94807 Villejuif
| | - Eric Rubinstein
- From Inserm, U935, F-94807 Villejuif, .,the Université Paris-Sud, Institut André Lwoff, F-94807 Villejuif
| |
Collapse
|
23
|
The CD9, CD81, and CD151 EC2 domains bind to the classical RGD-binding site of integrin αvβ3. Biochem J 2016; 474:589-596. [PMID: 27993971 DOI: 10.1042/bcj20160998] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 12/02/2016] [Accepted: 12/19/2016] [Indexed: 11/17/2022]
Abstract
Tetraspanins play important roles in normal (e.g. cell adhesion, motility, activation, and proliferation) and pathological conditions (e.g. metastasis and viral infection). Tetraspanins interact with integrins and regulate integrin functions, but the specifics of tetraspanin-integrin interactions are unclear. Using co-immunoprecipitation with integrins as a sole method to detect interaction between integrins and full-length tetraspanins, it has been proposed that the variable region (helices D and E) of the extracellular-2 (EC2) domain of tetraspanins laterally associates with a non-ligand-binding site of integrins. We describe that, using adhesion assays, the EC2 domain of CD81, CD9, and CD151 bound to integrin αvβ3, and this binding was suppressed by cRGDfV, a specific inhibitor of αvβ3, and antibody 7E3, which is mapped to the ligand-binding site of β3. We also present evidence that the specificity loop of β3 directly bound to the EC2 domains. This suggests that the EC2 domains specifically bind to the classical ligand-binding site of αvβ3. αvβ3 was a more effective receptor for the EC2 domains than the previously known tetraspanin receptors α3β1, α4β1, and α6β1. Docking simulation predicted that the helices A and B of CD81 EC2 bind to the RGD-binding site of αvβ3. Substituting Lys residues at positions 116 and 144/148 of CD81 EC2 in the predicted integrin-binding interface reduced the binding of CD81 EC2 to αvβ3, consistent with the docking model. These findings suggest that, in contrast with previous models, the ligand-binding site of integrin αvβ3, a new tetraspanin receptor, binds to the constant region (helices A and B) of the EC2 domain.
Collapse
|
24
|
Tilghman J, Schiapparelli P, Lal B, Ying M, Quinones-Hinojosa A, Xia S, Laterra J. Regulation of Glioblastoma Tumor-Propagating Cells by the Integrin Partner Tetraspanin CD151. Neoplasia 2016; 18:185-98. [PMID: 26992919 PMCID: PMC4796809 DOI: 10.1016/j.neo.2016.02.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 01/29/2016] [Accepted: 02/09/2016] [Indexed: 01/06/2023] Open
Abstract
Glioblastoma (GBM) stem cells (GSCs) represent tumor-propagating cells with stem-like characteristics (stemness) that contribute disproportionately to GBM drug resistance and tumor recurrence. Understanding the mechanisms supporting GSC stemness is important for developing therapeutic strategies for targeting GSC-dependent oncogenic mechanisms. Using GBM-derived neurospheres, we identified the cell surface tetraspanin family member CD151 as a novel regulator of glioma cell stemness, GSC self-renewal capacity, migration, and tumor growth. CD151 was found to be overexpressed in GBM tumors and GBM neurospheres enriched in GSCs. Silencing CD151 inhibited neurosphere forming capacity, neurosphere cell proliferation, and migration and attenuated the expression of markers and transcriptional drivers of the GSC phenotype. Conversely, forced CD151 expression promoted neurosphere self-renewal, cell migration, and expression of stemness-associated transcription factors. CD151 was found to complex with integrins α3, α6, and β1 in neurosphere cells, and blocking CD151 interactions with integrins α3 and α6 inhibited AKT phosphorylation, a downstream effector of integrin signaling, and impaired sphere formation and neurosphere cell migration. Additionally, targeting CD151 in vivo inhibited the growth of GBM neurosphere-derived xenografts. These findings identify CD151 and its interactions with integrins α3 and α6 as potential therapeutic targets for inhibiting stemness-driving mechanisms and stem cell populations in GBM.
Collapse
Affiliation(s)
- Jessica Tilghman
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, 21205, USA; Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Paula Schiapparelli
- Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Bachuchu Lal
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, 21205, USA; Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Mingyao Ying
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, 21205, USA; Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Alfredo Quinones-Hinojosa
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA; Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Shuli Xia
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, 21205, USA; Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - John Laterra
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, 21205, USA; Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA; Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA; Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
25
|
Integrin-mediated regulation of epidermal wound functions. Cell Tissue Res 2016; 365:467-82. [PMID: 27351421 DOI: 10.1007/s00441-016-2446-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 06/02/2016] [Indexed: 01/14/2023]
Abstract
During cutaneous wound healing, keratinocyte proliferation and migration are critical for re-epithelialization. In addition the epidermis secretes growth factors, cytokines, proteases, and matricellular proteins into the wound microenvironment that modify the extracellular matrix and stimulate other wound cells that control the inflammatory response, promote angiogenesis and facilitate tissue contraction and remodeling. Wound keratinocytes express at least seven different integrins-the major cell adhesion receptors for the extracellular matrix-that collectively control essential cell-autonomous functions to ensure proper re-epithelialization, including migration, proliferation, survival and basement membrane assembly. Moreover, it has become evident in recent years that some integrins can regulate paracrine signals from wound epidermis that stimulate other wound cells involved in angiogenesis, contraction and inflammation. Importantly, it is likely that abnormal integrin expression or function in the epidermis contributes to wound pathologies such as over-exuberant healing (e.g., hypertrophic scar formation) or diminished healing (e.g., chronic wounds). In this review, we discuss current knowledge of integrin function in the epidermis, which implicates them as attractive therapeutic targets to promote wound healing or treat wound pathologies. We also discuss challenges that arise from the complex roles that multiple integrins play in wound epidermis, which may be regulated through extracellular matrix remodeling that determines ligand availability. Indeed, understanding how different integrin functions are temporally coordinated in wound epidermis and which integrin functions go awry in pathological wounds, will be important to determine how best to target them clinically to achieve maximum therapeutic benefit. Graphical abstract In addition to their well-characterized roles in keratinocyte adhesion, migration and wound re-epithelialization, epidermal integrins play important roles in modifying the wound microenvironment by regulating the expression and secretion of growth factors, extracellular proteases, and matricellular proteins that stimulate other wound cells, including vascular endothelial cells and fibroblasts/myofibroblasts.
Collapse
|
26
|
Tesfay L, Schulz VV, Frank SB, Lamb LE, Miranti CK. Receptor tyrosine kinase Met promotes cell survival via kinase-independent maintenance of integrin α3β1. Mol Biol Cell 2016; 27:2493-504. [PMID: 27307589 PMCID: PMC4966988 DOI: 10.1091/mbc.e15-09-0649] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 06/08/2016] [Indexed: 01/13/2023] Open
Abstract
This study identifies a new mechanism by which the receptor tyrosine kinase Met promotes cell survival. The ectodomain and transmembrane domain of Met, independently of kinase activity, are required to maintain integrin α3β1 on the cell surface to prevent activation of intrinsic and extrinsic cell death pathways and maintain autophagic flux. Matrix adhesion via integrins is required for cell survival. Adhesion of epithelial cells to laminin via integrin α3β1 was previously shown to activate at least two independent survival pathways. First, integrin α3β1 is required for autophagy-induced cell survival after growth factor deprivation. Second, integrin α3β1 independently activates two receptor tyrosine kinases, EGFR and Met, in the absence of ligands. EGFR signaling to Erk promotes survival independently of autophagy. To determine how Met promotes cell survival, we inhibited Met kinase activity or blocked its expression with RNA interference. Loss of Met expression, but not inhibition of Met kinase activity, induced apoptosis by reducing integrin α3β1 levels, activating anoikis, and blocking autophagy. Met was specifically required for the assembly of autophagosomes downstream of LC3II processing. Reexpression of wild-type Met, kinase-dead Met, or integrin α3 was sufficient to rescue death upon removal of endogenous Met. Integrin α3β1 coprecipitated and colocalized with Met in cells. The extracellular and transmembrane domain of Met was required to fully rescue cell death and restore integrin α3 expression. Thus Met promotes survival of laminin-adherent cells by maintaining integrin α3β1 via a kinase-independent mechanism.
Collapse
Affiliation(s)
- Lia Tesfay
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503
| | - Veronique V Schulz
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503
| | - Sander B Frank
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503
| | - Laura E Lamb
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503
| | - Cindy K Miranti
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503
| |
Collapse
|
27
|
Noy PJ, Yang J, Reyat JS, Matthews AL, Charlton AE, Furmston J, Rogers DA, Rainger GE, Tomlinson MG. TspanC8 Tetraspanins and A Disintegrin and Metalloprotease 10 (ADAM10) Interact via Their Extracellular Regions: EVIDENCE FOR DISTINCT BINDING MECHANISMS FOR DIFFERENT TspanC8 PROTEINS. J Biol Chem 2015; 291:3145-57. [PMID: 26668317 PMCID: PMC4751363 DOI: 10.1074/jbc.m115.703058] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Indexed: 01/01/2023] Open
Abstract
A disintegrin and metalloprotease 10 (ADAM10) is a ubiquitously expressed transmembrane metalloprotease that cleaves the extracellular regions from its transmembrane substrates. ADAM10 is essential for embryonic development and is implicated in cancer, Alzheimer, and inflammatory diseases. The tetraspanins are a superfamily of 33 four-transmembrane proteins in mammals, of which the TspanC8 subgroup (Tspan5, 10, 14, 15, 17, and 33) promote ADAM10 intracellular trafficking and enzymatic maturation. However, the interaction between TspanC8s and ADAM10 has only been demonstrated in overexpression systems and the interaction mechanism remains undefined. To address these issues, an antibody was developed to Tspan14, which was used to show co-immunoprecipitation of Tspan14 with ADAM10 in primary human cells. Chimeric Tspan14 constructs demonstrated that the large extracellular loop of Tspan14 mediated its co-immunoprecipitation with ADAM10, and promoted ADAM10 maturation and trafficking to the cell surface. Chimeric ADAM10 constructs showed that membrane-proximal stalk, cysteine-rich, and disintegrin domains of ADAM10 mediated its co-immunoprecipitation with Tspan14 and other TspanC8s. This TspanC8-interacting region was required for ADAM10 exit from the endoplasmic reticulum. Truncated ADAM10 constructs revealed differential TspanC8 binding requirements for the stalk, cysteine-rich, and disintegrin domains. Moreover, Tspan15was the only TspanC8 to promote cleavage of the ADAM10 substrate N-cadherin, whereas Tspan14 was unique in reducing cleavage of the platelet collagen receptor GPVI. These findings suggest that ADAM10 may adopt distinct conformations in complex with different TspanC8s, which could impact on substrate selectivity. Furthermore, this study identifies regions of TspanC8s and ADAM10 for potential interaction-disrupting therapeutic targeting.
Collapse
Affiliation(s)
- Peter J Noy
- From the School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom and
| | - Jing Yang
- From the School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom and
| | - Jasmeet S Reyat
- From the School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom and
| | - Alexandra L Matthews
- From the School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom and
| | - Alice E Charlton
- From the School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom and
| | - Joanna Furmston
- From the School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom and
| | - David A Rogers
- From the School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom and
| | - G Ed Rainger
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Michael G Tomlinson
- From the School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom and
| |
Collapse
|
28
|
Beckwith KA, Byrd JC, Muthusamy N. Tetraspanins as therapeutic targets in hematological malignancy: a concise review. Front Physiol 2015; 6:91. [PMID: 25852576 PMCID: PMC4369647 DOI: 10.3389/fphys.2015.00091] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 03/05/2015] [Indexed: 12/11/2022] Open
Abstract
Tetraspanins belong to a family of transmembrane proteins which play a major role in the organization of the plasma membrane. While all immune cells express tetraspanins, most of these are present in a variety of other cell types. There are a select few, such as CD37 and CD53, which are restricted to hematopoietic lineages. Tetraspanins associate with numerous partners involved in a diverse set of biological processes, including cell activation, survival, proliferation, adhesion, and migration. The historical view has assigned them a scaffolding role, but recent discoveries suggest some tetraspanins can directly participate in signaling through interactions with cytoplasmic proteins. Given their potential roles in supporting tumor survival and immune evasion, an improved understanding of tetraspanin activity could prove clinically valuable. This review will focus on emerging data in the study of tetraspanins, advances in the clinical development of anti-CD37 therapeutics, and the future prospects of targeting tetraspanins in hematological malignancy.
Collapse
Affiliation(s)
- Kyle A Beckwith
- Division of Hematology, Department of Internal Medicine, The Ohio State University Columbus, OH, USA
| | - John C Byrd
- Division of Hematology, Department of Internal Medicine, The Ohio State University Columbus, OH, USA ; Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University Columbus, OH, USA
| | - Natarajan Muthusamy
- Division of Hematology, Department of Internal Medicine, The Ohio State University Columbus, OH, USA ; Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University Columbus, OH, USA
| |
Collapse
|
29
|
Kumari S, Devi G, Badana A, Dasari VR, Malla RR. CD151-A Striking Marker for Cancer Therapy. BIOMARKERS IN CANCER 2015; 7:7-11. [PMID: 25861224 PMCID: PMC4372031 DOI: 10.4137/bic.s21847] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 01/28/2015] [Accepted: 01/28/2015] [Indexed: 12/28/2022]
Abstract
Cluster of differentiation 151 (CD151) is a member of the mammalian tetraspanin family, which is involved in diverse functions such as maintaining normal cellular integrity, cell-to-cell communication, wound healing, platelet aggregation, trafficking, cell motility and angiogenesis. CD151 also supports de novo carcinogenesis in human skin squamous cell carcinoma (SCC) and tumor metastasis. CD151 interacts with α3β1 and α6β4 integrins through palmitoylation where cysteine plays an important role in the association of CD151 with integrins and non-integrin proteins. Invasion and metastasis of cancer cells were diminished by decreasing CD151 association with integrins. CD151 functions at various stages of cancer, including metastatic cascade and primary tumor growth, thus reinforcing the importance of CD151 as a target in oncology. The present review highlights the role of CD151 in tumor metastasis and its importance in cancer therapy.
Collapse
Affiliation(s)
- Seema Kumari
- Cancer Biology Lab, Department of Biochemistry, Institute of Science, GITAM University, Visakhapatnam, Andhra Pradesh, India
| | - Gayatri Devi
- Cancer Biology Lab, Department of Biochemistry, Institute of Science, GITAM University, Visakhapatnam, Andhra Pradesh, India
| | - Anil Badana
- Cancer Biology Lab, Department of Biochemistry, Institute of Science, GITAM University, Visakhapatnam, Andhra Pradesh, India
| | - Venkata Ramesh Dasari
- Department of Cancer Biology and Pharmacology, College of Medicine, University of Illinois, Peoria, IL, USA
| | - Rama Rao Malla
- Cancer Biology Lab, Department of Biochemistry, Institute of Science, GITAM University, Visakhapatnam, Andhra Pradesh, India
| |
Collapse
|
30
|
Longmate WM, DiPersio CM. Integrin Regulation of Epidermal Functions in Wounds. Adv Wound Care (New Rochelle) 2014; 3:229-246. [PMID: 24669359 DOI: 10.1089/wound.2013.0516] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 01/13/2014] [Indexed: 12/12/2022] Open
Abstract
Significance: Integrins are bidirectional signaling receptors for extracellular matrix that regulate both inside-out signaling that controls keratinocyte-mediated changes to the wound microenvironment and outside-in signaling that controls keratinocyte responses to microenvironmental changes. As such, integrins represent attractive therapeutic targets for treatment of chronic wounds or general promotion of wound healing. Advances in wound management are particularly important as the elderly and diabetic populations within the United States continue to grow. Recent Advances: Although integrins are best known for mediating cell adhesion and migration, integrins in wound epidermis also control cell survival, proliferation, matrix remodeling, and paracrine crosstalk to other cellular compartments of the wound. Importantly, the concept of targeting integrins in the clinic has been established for treatment of certain cancers and other diseases, laying the groundwork for similar exploitation of integrins as targets to treat chronic wounds. Critical Issues: Despite their attractiveness as therapeutic targets, integrins have complex roles in wound healing that are impacted by both their own expression and a highly dynamic wound microenvironment that determines ligand availability. Therefore, identifying relevant integrin ligands in the wound and understanding both distinct and overlapping functions that different integrins play in the epidermis will be critical to determine their precise roles in wound healing. Future Directions: Future research should focus on gaining a thorough understanding of the highly coordinated functions of different integrins in wound epidermis, and on determining which of these functions go awry in pathological wounds. This focus should facilitate development of integrin-targeting therapeutics for treating chronic wounds.
Collapse
Affiliation(s)
- Whitney M. Longmate
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York
| | - C. Michael DiPersio
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York
| |
Collapse
|
31
|
Abstract
An abundance of evidence shows supporting roles for tetraspanin proteins in human cancer. Many studies show that the expression of tetraspanins correlates with tumour stage, tumour type and patient outcome. In addition, perturbations of tetraspanins in tumour cell lines can considerably affect cell growth, morphology, invasion, tumour engraftment and metastasis. This Review emphasizes new studies that have used de novo mouse cancer models to show that select tetraspanin proteins have key roles in tumour initiation, promotion and metastasis. This Review also emphasizes how tetraspanin proteins can sometimes participate in tumour angiogenesis. These recent data build an increasingly strong case for tetraspanins as therapeutic targets.
Collapse
|
32
|
Kerrisk ME, Cingolani LA, Koleske AJ. ECM receptors in neuronal structure, synaptic plasticity, and behavior. PROGRESS IN BRAIN RESEARCH 2014; 214:101-31. [PMID: 25410355 DOI: 10.1016/b978-0-444-63486-3.00005-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
During central nervous system development, extracellular matrix (ECM) receptors and their ligands play key roles as guidance molecules, informing neurons where and when to send axonal and dendritic projections, establish connections, and form synapses between pre- and postsynaptic cells. Once stable synapses are formed, many ECM receptors transition in function to control the maintenance of stable connections between neurons and regulate synaptic plasticity. These receptors bind to and are activated by ECM ligands. In turn, ECM receptor activation modulates downstream signaling cascades that control cytoskeletal dynamics and synaptic activity to regulate neuronal structure and function and thereby impact animal behavior. The activities of cell adhesion receptors that mediate interactions between pre- and postsynaptic partners are also strongly influenced by ECM composition. This chapter highlights a number of ECM receptors, their roles in the control of synapse structure and function, and the impact of these receptors on synaptic plasticity and animal behavior.
Collapse
Affiliation(s)
- Meghan E Kerrisk
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Lorenzo A Cingolani
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Anthony J Koleske
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA; Department of Neurobiology, Yale University, New Haven, CT, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT, USA; Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University, New Haven, CT, USA.
| |
Collapse
|
33
|
Disease-associated single amino acid mutation in the calf-1 domain of integrin α3 leads to defects in its processing and cell surface expression. Biochem Biophys Res Commun 2013; 441:988-93. [DOI: 10.1016/j.bbrc.2013.11.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 11/02/2013] [Indexed: 11/22/2022]
|
34
|
Detchokul S, Newell B, Williams ED, Frauman AG. CD151 is associated with prostate cancer cell invasion and lymphangiogenesis in vivo. Oncol Rep 2013; 31:241-7. [PMID: 24174171 DOI: 10.3892/or.2013.2823] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 09/18/2013] [Indexed: 11/06/2022] Open
Abstract
CD151, a member of the tetraspanin family, is associated with regulation of migration of normal and tumour cells via cell surface microdomain formation. CD151 was found in our laboratory to have a prognostic value in prostate cancer and is a promoter of prostate cancer migration and invasion. These roles involve association with integrins on both cell-cell and cell-stroma levels. Furthermore, CD151 plays a role in endothelial cell motility. CD151 expression was examined in three commonly used prostate cancer cell lines. We investigated CD151 expression, angiogenesis (microvessel density; MVD) and lymphangiogenesis (lymphatic vessel density; LVD) in an orthotopic xenograft model of prostate cancer in matched tumours from primary and secondary sites. CD151 was found to be heterogeneously expressed across different prostate cancer cell lines and the levels of CD151 expression were significantly higher in the highly tumorigenic, androgen-insensitive cells PC-3 and DU-145 compared to the androgen-sensitive cell line LNCaP (P<0.05). The majority of in vivo xenografts developed pelvic lymph node metastases. Importantly, primary tumours that developed metastasis had significantly higher CD151 expression and MVD compared to those which did not develop metastasis (P<0.05). We identified, for the first time, that CD151 expression is associated with LVD in prostate cancer. These findings underscore the potential role of CD151 and angiogenesis in the metastatic potential of prostate cancer. CD151 has a prognostic value in this mouse model of prostate cancer and may play a role in lymphangiogenesis. CD151 is likely an important regulator of cancer cell communication with the surrounding microenvironment.
Collapse
Affiliation(s)
- Sujitra Detchokul
- Clinical Pharmacology and Therapeutics Unit, Department of Medicine (Austin Health/Northern Health), The University of Melbourne, Heidelberg, VIC, Australia
| | | | | | | |
Collapse
|
35
|
Verpelli C, Montani C, Vicidomini C, Heise C, Sala C. Mutations of the synapse genes and intellectual disability syndromes. Eur J Pharmacol 2013; 719:112-116. [PMID: 23872408 DOI: 10.1016/j.ejphar.2013.07.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 06/04/2013] [Accepted: 07/01/2013] [Indexed: 01/10/2023]
Abstract
Intellectual disability syndromes have been found associated to numerous mutated genes that code for proteins functionally involved in synapse formation, the regulation of dendritic spine morphology, the regulation of the synaptic cytoskeleton or the synthesis and degradation of specific synapse proteins. These studies have strongly demonstrated that even mild alterations in synapse morphology and function give rise to mild or severe alteration in intellectual abilities. Interestingly, pharmacological agents that are able to counteract these morphological and functional synaptic anomalies can also improve the symptoms of some of these conditions. This review is summarizing recent discoveries on the functions of some of the genes responsible for intellectual disability syndromes connected with synapse dysfunctions.
Collapse
Affiliation(s)
- Chiara Verpelli
- CNR Institute of Neuroscience and Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli 32, 20129 Milano, Italy
| | - Caterina Montani
- CNR Institute of Neuroscience and Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli 32, 20129 Milano, Italy
| | - Cinzia Vicidomini
- CNR Institute of Neuroscience and Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli 32, 20129 Milano, Italy
| | - Christopher Heise
- CNR Institute of Neuroscience and Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli 32, 20129 Milano, Italy
| | - Carlo Sala
- CNR Institute of Neuroscience and Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli 32, 20129 Milano, Italy; Neuromuscular Diseases and Neuroimmunology, Neurological Institute Foundation Carlo Besta, Via Celoria 11, 20133 Milan, Italy.
| |
Collapse
|
36
|
Zhang BH, Liu W, Li L, Lu JG, Sun YN, Jin DJ, Xu XY. KAI1/CD82 and MRP1/CD9 Serve as Markers of Infiltration, Metastasis, and Prognosis in Laryngeal Squamous Cell Carcinomas. Asian Pac J Cancer Prev 2013; 14:3521-6. [DOI: 10.7314/apjcp.2013.14.6.3521] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
37
|
Gustafson-Wagner E, Stipp CS. The CD9/CD81 tetraspanin complex and tetraspanin CD151 regulate α3β1 integrin-dependent tumor cell behaviors by overlapping but distinct mechanisms. PLoS One 2013; 8:e61834. [PMID: 23613949 PMCID: PMC3629153 DOI: 10.1371/journal.pone.0061834] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 03/15/2013] [Indexed: 01/16/2023] Open
Abstract
Integrin α3β1 potently promotes cell motility on its ligands, laminin-332 and laminin-511, and this may help to explain why α3β1 has repeatedly been linked to breast carcinoma progression and metastasis. The pro-migratory functions of α3β1 depend strongly on lateral interactions with cell surface tetraspanin proteins. Tetraspanin CD151 interacts directly with the α3 integrin subunit and links α3β1 integrin to other tetraspanins, including CD9 and CD81. Loss of CD151 disrupts α3β1 association with other tetraspanins and impairs α3β1-dependent motility. However, the extent to which tetraspanins other than CD151 are required for specific α3β1 functions is unclear. To begin to clarify which aspects of α3β1 function require which tetraspanins, we created breast carcinoma cells depleted of both CD9 and CD81 by RNA interference. Silencing both of these closely related tetraspanins was required to uncover their contributions to α3β1 function. We then directly compared our CD9/CD81-silenced cells to CD151-silenced cells. Both CD9/CD81-silenced cells and CD151-silenced cells showed delayed α3β1-dependent cell spreading on laminin-332. Surprisingly, however, once fully spread, CD9/CD81-silenced cells, but not CD151-silenced cells, displayed impaired α3β1-dependent directed motility and altered front-rear cell morphology. Also unexpectedly, the CD9/CD81 complex, but not CD151, was required to promote α3β1 association with PKCα in breast carcinoma cells, and a PKC inhibitor mimicked aspects of the CD9/CD81-silenced cell motility defect. Our data reveal overlapping, but surprisingly distinct contributions of specific tetraspanins to α3β1 integrin function. Importantly, some of CD9/CD81's α3β1 regulatory functions may not require CD9/CD81 to be physically linked to α3β1 by CD151.
Collapse
Affiliation(s)
| | - Christopher S. Stipp
- Department of Biology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa, United States of America
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa, United States of America
| |
Collapse
|
38
|
Li Q, Yang XH, Xu F, Sharma C, Wang HX, Knoblich K, Rabinovitz I, Granter SR, Hemler ME. Tetraspanin CD151 plays a key role in skin squamous cell carcinoma. Oncogene 2013; 32:1772-83. [PMID: 22824799 PMCID: PMC3482293 DOI: 10.1038/onc.2012.205] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Revised: 03/30/2012] [Accepted: 04/23/2012] [Indexed: 11/23/2022]
Abstract
Here we provide the first evidence that tetraspanin CD151 can support de novo carcinogenesis. During two-stage mouse skin chemical carcinogenesis, CD151 reduces tumor lag time and increases incidence, multiplicity, size and progression to malignant squamous cell carcinoma (SCC), while supporting both cell survival during tumor initiation and cell proliferation during the promotion phase. In human skin SCC, CD151 expression is selectively elevated compared with other skin cancer types. CD151 support of keratinocyte survival and proliferation may depend on activation of transcription factor STAT3 (signal transducers and activators of transcription), a regulator of cell proliferation and apoptosis. CD151 also supports protein kinase C (PKC)α-α6β4 integrin association and PKC-dependent β4 S1424 phosphorylation, while regulating α6β4 distribution. CD151-PKCα effects on integrin β4 phosphorylation and subcellular localization are consistent with epithelial disruption to a less polarized, more invasive state. CD151 ablation, while minimally affecting normal cell and normal mouse functions, markedly sensitized mouse skin and epidermoid cells to chemicals/drugs including 7,12-dimethylbenz[α]anthracene (mutagen) and camptothecin (topoisomerase inhibitor), as well as to agents targeting epidermal growth factor receptor, PKC, Jak2/Tyk2 and STAT3. Hence, CD151 'co-targeting' may be therapeutically beneficial. These findings not only support CD151 as a potential tumor target, but also should apply to other cancers utilizing CD151/laminin-binding integrin complexes.
Collapse
Affiliation(s)
- Qinglin Li
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute and Harvard Medical School, Boston MA
| | - Xiuwei H. Yang
- Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington, KY
| | - Fenghui Xu
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute and Harvard Medical School, Boston MA
| | - Chandan Sharma
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute and Harvard Medical School, Boston MA
| | - Hong-Xing Wang
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute and Harvard Medical School, Boston MA
| | - Konstantin Knoblich
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute and Harvard Medical School, Boston MA
| | - Isaac Rabinovitz
- Division of Cancer Biology and Angiogenesis, Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston MA
| | - Scott R. Granter
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston MA
| | - Martin E. Hemler
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute and Harvard Medical School, Boston MA
| |
Collapse
|
39
|
Toricelli M, Melo FHM, Peres GB, Silva DCP, Jasiulionis MG. Timp1 interacts with beta-1 integrin and CD63 along melanoma genesis and confers anoikis resistance by activating PI3-K signaling pathway independently of Akt phosphorylation. Mol Cancer 2013; 12:22. [PMID: 23522389 PMCID: PMC3635912 DOI: 10.1186/1476-4598-12-22] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 03/14/2013] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Anoikis resistance is one of the abilities acquired along tumor progression. This characteristic is associated with metastasis development, since tumorigenic cells must survive independently of cell-matrix interactions in this process. In our laboratory, it was developed a murine melanocyte malignant transformation model associated with a sustained stressful condition. After subjecting melan-a melanocytes to 1, 2, 3 and 4 cycles of anchorage impediment, anoikis resistant cells were established and named 1C, 2C, 3C and 4C, respectively. These cells showed altered morphology and PMA independent cell growth, but were not tumorigenic, corresponding to pre-malignant cells. After limiting dilution of 4C pre-malignant cells, melanoma cell lines with different characteristics were obtained. Previous data from our group showed that increased Timp1 expression correlated with anoikis-resistant phenotype. Timp1 was shown to confer anchorage-independent growth capability to melan-a melanocytes and render melanoma cells more aggressive when injected into mice. However, the mechanisms involved in anoikis regulation by Timp1 in tumorigenic cells are not clear yet. METHODS The β1-integrin and Timp1 expression were evaluated by Western blotting and CD63 protein expression by flow cytometry using specific antibodies. To analyze the interaction among Timp1, CD63 and β1-integrin, immunoprecipitation assays were performed, anoikis resistance capability was evaluated in the presence or not of the PI3-K inhibitors, Wortmannin and LY294002. Relative expression of TIMP1 and CD63 in human metastatic melanoma cells was analyzed by real time PCR. RESULTS Differential association among Timp1, CD63 and β1-integrins was observed in melan-a melanocytes, 4C pre-malignant melanocytes and 4C11- and 4C11+ melanoma cells. Timp1 present in conditioned medium of melanoma cells rendered melan-a melanocytes anoikis-resistant through PI3-K signaling pathway independently of Akt activation. In human melanoma cell lines, in which TIMP1 and beta-1 integrin were also found to be interacting, TIMP1 and CD63 levels together was shown to correlate significantly with colony formation capacity. CONCLUSIONS Our results show that Timp1 is assembled in a supramolecular complex containing CD63 and β1-integrins along melanoma genesis and confers anoikis resistance by activating PI3-K signaling pathway, independently of Akt phosphorylation. In addition, our data point TIMP1, mainly together with CD63, as a potential biomarker of melanoma.
Collapse
Affiliation(s)
- Mariana Toricelli
- Pharmacology Department, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
40
|
Schröder HM, Hoffmann SC, Hecker M, Korff T, Ludwig T. The tetraspanin network modulates MT1-MMP cell surface trafficking. Int J Biochem Cell Biol 2013; 45:1133-44. [PMID: 23500527 DOI: 10.1016/j.biocel.2013.02.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 02/11/2013] [Accepted: 02/21/2013] [Indexed: 10/27/2022]
Abstract
The membrane-type 1 matrix metalloproteinase (MT1-MMP) drives fundamental physiological and pathophysiological processes. Among other substrates, MT1-MMP cleaves components of the extracellular matrix and activates other matrix-cleaving proteases such as MMP-2. Trafficking is a highly effective means to modulate MT1-MMP cell surface expression, and hence regulate its function. Here, we describe the complex interaction of MT1-MMP with tetraspanins, their effects on MT1-MMP intracellular trafficking and proteolytic function. Tetraspanins are credited as membrane organizers that form a network within the membrane to regulate the trafficking of associated proteins. In short, we found MT1-MMP to interact with the tetraspanin-associated EWI-2 protein by a yeast two-hybrid screen. Immunoprecipitation analysis confirmed this interaction and further revealed that MT1-MMP also stably interacts with distinct tetraspanins (CD9, CD37, CD53, CD63, CD81, and CD82) and the tetraspanin-like MAL protein. By using different MT1-MMP truncation constructs and mutants, we observed that all tetraspanins and MAL associated with the hemopexin domain of MT1-MMP. Moreover, this interaction was independent of O-glycosylation of MT1-MMP and exclusively occurred in the endoplasmic reticulum. Here, the respective subcellular compartment was identified by fitting the MT1-MMP interaction pattern to a model for post-translational processing of MT1-MMP. In addition, tetraspanins differentially affected the cell surface localization of MT1-MMP, its capacity to activate pro-MMP-2, and the collagen invasion capacity. Interestingly, the degree of tetraspanin-MT1-MMP association did not correlate with its impact on MT1-MMP function. Tetraspanins thus distinctly affect MT1-MMP subcellular localization and function, and may constitute an effective mechanism to control MT1-MMP-dependent proteolysis at the cell surface.
Collapse
Affiliation(s)
- H M Schröder
- Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, Heidelberg University, 69120 Heidelberg, Germany.
| | | | | | | | | |
Collapse
|
41
|
Peddibhotla SSD, Brinkmann BF, Kummer D, Tuncay H, Nakayama M, Adams RH, Gerke V, Ebnet K. Tetraspanin CD9 links junctional adhesion molecule-A to αvβ3 integrin to mediate basic fibroblast growth factor-specific angiogenic signaling. Mol Biol Cell 2013; 24:933-44. [PMID: 23389628 PMCID: PMC3608503 DOI: 10.1091/mbc.e12-06-0481] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Junctional adhesion molecule-A (JAM-A) is a member of the immunoglobulin family with diverse functions in epithelial cells, including cell migration, cell contact maturation, and tight junction formation. In endothelial cells, JAM-A has been implicated in basic fibroblast growth factor (bFGF)-regulated angiogenesis through incompletely understood mechanisms. In this paper, we identify tetraspanin CD9 as novel binding partner for JAM-A in endothelial cells. CD9 acts as scaffold and assembles a ternary JAM-A-CD9-αvβ3 integrin complex from which JAM-A is released upon bFGF stimulation. CD9 interacts predominantly with monomeric JAM-A, which suggests that bFGF induces signaling by triggering JAM-A dimerization. Among the two vitronectin receptors, αvβ3 and αvβ5 integrin, which have been shown to cooperate during angiogenic signaling with bFGF and vascular endothelial growth factor (VEGF), respectively, CD9 links JAM-A specifically to αvβ3 integrin. In line with this, knockdown of CD9 blocks bFGF- but not VEGF-induced ERK1/2 activation. JAM-A or CD9 knockdown impairs endothelial cell migration and tube formation. Our findings indicate that CD9 incorporates monomeric JAM-A into a complex with αvβ3 integrin, which responds to bFGF stimulation by JAM-A release to regulate mitogen-activated protein kinase (MAPK) activation, endothelial cell migration, and angiogenesis. The data also provide new mechanistic insights into the cooperativity between bFGF and αvβ3 integrin during angiogenic signaling.
Collapse
|
42
|
Sachs N, Sonnenberg A. Cell-matrix adhesion of podocytes in physiology and disease. Nat Rev Nephrol 2013; 9:200-10. [PMID: 23338211 DOI: 10.1038/nrneph.2012.291] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Cell-matrix adhesion is crucial for maintaining the mechanical integrity of epithelial tissues. Podocytes--a key component of the glomerular filtration barrier--are exposed to permanent transcapillary filtration pressure and must therefore adhere tightly to the underlying glomerular basement membrane (GBM). The major cell-matrix adhesion receptor in podocytes is the integrin α3β1, which connects laminin 521 in the GBM through various adaptor proteins to the intracellular actin cytoskeleton. Other cell-matrix adhesion receptors expressed by podocytes include the integrins α2β1 and αvβ3, α-dystroglycan, syndecan-4 and type XVII collagen. Mutations in genes encoding any of the components critical for podocyte adhesion cause glomerular disease. This Review highlights recent advances in our understanding of the cell biology and genetics of podocyte adhesion with special emphasis on glomerular disease.
Collapse
Affiliation(s)
- Norman Sachs
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | |
Collapse
|
43
|
Nicolaou N, Margadant C, Kevelam SH, Lilien MR, Oosterveld MJS, Kreft M, van Eerde AM, Pfundt R, Terhal PA, van der Zwaag B, Nikkels PGJ, Sachs N, Goldschmeding R, Knoers NVAM, Renkema KY, Sonnenberg A. Gain of glycosylation in integrin α3 causes lung disease and nephrotic syndrome. J Clin Invest 2012; 122:4375-87. [PMID: 23114595 DOI: 10.1172/jci64100] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 09/06/2012] [Indexed: 12/23/2022] Open
Abstract
Integrins are transmembrane αβ glycoproteins that connect the extracellular matrix to the cytoskeleton. The laminin-binding integrin α3β1 is expressed at high levels in lung epithelium and in kidney podocytes. In podocytes, α3β1 associates with the tetraspanin CD151 to maintain a functional filtration barrier. Here, we report on a patient homozygous for a novel missense mutation in the human ITGA3 gene, causing fatal interstitial lung disease and congenital nephrotic syndrome. The mutation caused an alanine-to-serine substitution in the integrin α3 subunit, thereby introducing an N-glycosylation motif at amino acid position 349. We expressed this mutant form of ITGA3 in murine podocytes and found that hyperglycosylation of the α3 precursor prevented its heterodimerization with β1, whereas CD151 association with the α3 subunit occurred normally. Consequently, the β1 precursor accumulated in the ER, and the mutant α3 precursor was degraded by the ubiquitin-proteasome system. Thus, these findings uncover a gain-of-glycosylation mutation in ITGA3 that prevents the biosynthesis of functional α3β1, causing a fatal multiorgan disorder.
Collapse
Affiliation(s)
- Nayia Nicolaou
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
α3β1 integrins regulate CD151 complex assembly and membrane dynamics in carcinoma cells within 3D environments. Oncogene 2012; 32:3965-79. [PMID: 22986527 DOI: 10.1038/onc.2012.415] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 07/23/2012] [Accepted: 07/31/2012] [Indexed: 11/09/2022]
Abstract
Integrins are extracellular matrix (ECM) receptors that are key players in the regulation of tumour cell invasion. The laminin-binding integrin α3β1 has previously been shown to regulate adhesion and migration of carcinoma cells in part through co-operative signalling with the tetraspanin family of transmembrane proteins. However, the spatial and temporal regulation of crosstalk between these families of transmembrane proteins in intact cells remains poorly understood. Here we have used fluorescence resonance energy transfer (FRET) to demonstrate for the first time that α3β1 and the tetraspanin CD151 directly associate at the front and retracting rear of polarised migrating breast carcinoma cells in both two-dimentional (2D) and three-dimentional (3D)matrices. Furthermore, localised α3β1-CD151 binding correlates with lower CD151 homodimerisation in cells migrating on laminin or within matrigel. Loss of α3β1 integrin leads to increased CD151 homodimer formation, increased activation of Rho GTPase, loss of cell polarity and decreased invasion in 3D ECM. As a result, α3-silenced cells show decreased actin-based membrane protrusion and retraction in both 2D and 3D environments. These data demonstrate that associations between α3β1 and CD151 occur dynamically within discrete subcellular compartments and act to establish local GTPase signalling to promote tumour cell invasion. These novel findings shed light on the complex crosstalk and switching between receptor complexes in response to different extracellular cues during cell invasion in 3D environments.
Collapse
|
45
|
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: 1.9] [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
|
46
|
Yang XH, Mirchev R, Deng X, Yacono P, Yang HL, Golan DE, Hemler ME. CD151 restricts the α6 integrin diffusion mode. J Cell Sci 2012; 125:1478-87. [PMID: 22328509 DOI: 10.1242/jcs.093963] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Laminin-binding integrins (α3β1, α6β1, α6β4, α7β1) are almost always expressed together with tetraspanin CD151. In every coexpressing cell analyzed to date, CD151 makes a fundamental contribution to integrin-dependent motility, invasion, morphology, adhesion and/or signaling. However, there has been minimal mechanistic insight into how CD151 affects integrin functions. In MDA-MB-231 mammary cells, tetraspanin CD151 knockdown impairs α6 integrin clustering and functions without decreasing α6 integrin expression or activation. Furthermore, CD151 knockdown minimally affects the magnitude of α6 integrin diffusion, as measured using single particle tracking. Instead, CD151 knockdown has a novel and unexpected dysregulating effect on the mode of α6 integrin diffusion. In control cells α6 integrin shows mostly random-confined diffusion (RCD) and some directed motion (DMO). In sharp contrast, in CD151-knockdown cells α6 integrin shows mostly DMO. In control cells α6 diffusion mode is sensitive to actin disruption, talin knockdown and phorbol ester stimulation. By contrast, CD151 knockdown cell α6 integrin is sensitive to actin disruption but desensitized to talin knockdown or phorbol ester stimulation, indicating dysregulation. Both phorbol ester and EGF stimulate cell spreading and promote α6 RCD in control cells. By contrast, CD151-ablated cells retain EGF effects but lose phorbol-ester-stimulated spreading and α6 RCD. For α6 integrins, physical association with CD151 promotes α6 RCD, in support of α6-mediated cable formation and adhesion. By comparison, for integrins not associated with CD151 (e.g. αv integrins), CD151 affects neither diffusion mode nor αv function. Hence, CD151 support of α6 RCD is specific and functionally relevant, and probably underlies diverse CD151 functions in skin, kidney and cancer cells.
Collapse
Affiliation(s)
- Xiuwei H Yang
- Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115, USA.
| | | | | | | | | | | | | |
Collapse
|
47
|
Bassani S, Cingolani LA. Tetraspanins: Interactions and interplay with integrins. Int J Biochem Cell Biol 2012; 44:703-8. [PMID: 22326999 DOI: 10.1016/j.biocel.2012.01.020] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 01/20/2012] [Accepted: 01/27/2012] [Indexed: 12/14/2022]
Abstract
Tetraspanins are small transmembrane proteins present on the cell surface of almost every eukaryotic cell. Through binding with other transmembrane and intracellular proteins, they regulate diverse cellular processes ranging from cell adhesion and motility to synapse formation and tumor progression. Here, we provide a brief overview of molecular, cellular and clinical studies to illustrate how the multiple functions of this fascinating family of molecules stem from their interplay with multiple molecular partners. In particular, we emphasize the special relationship between tetraspanins and the cell adhesion molecules integrins in regulating cell physiology in health and disease.
Collapse
Affiliation(s)
- Silvia Bassani
- CNR Institute of Neuroscience, Cellular and Molecular Pharmacology, Department of Pharmacology, University of Milan, Italy
| | | |
Collapse
|
48
|
Abstract
Tetraspanins compose a family of structurally related molecules with four transmembrane domains. A total of 33 tetraspanins are present in the human genome, and tetraspanins are also found in plants and certain fungi. A well-known property of tetraspanins is their ability to interact with one another and many other surface proteins, which led to the suggestion that they organize a network of molecular interaction referred to as the 'tetraspanin web', and that they play a role in membrane compartmentalization. Recent studies of the dynamics of these molecules provided important new information that helped refining the models of this 'web'. Several genetic studies in mammals and invertebrates have demonstrated key physiological roles for some of the tetraspanins, in particular in immune response, sperm-egg fusion, photoreceptor function and the normal function of certain epitheliums or vascular development. However, in several examples, the phenotypes of tetraspanin-knockout mice are relatively mild or restricted to a particular organ, despite a wide tissue distribution.
Collapse
|
49
|
Zhu GH, Huang C, Qiu ZJ, Liu J, Zhang ZH, Zhao N, Feng ZZ, Lv XH. Expression and prognostic significance of CD151, c-Met, and integrin alpha3/alpha6 in pancreatic ductal adenocarcinoma. Dig Dis Sci 2011; 56:1090-8. [PMID: 20927591 DOI: 10.1007/s10620-010-1416-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Accepted: 08/26/2010] [Indexed: 12/20/2022]
Abstract
BACKGROUND CD151, c-Met, and integrin alpha3/alpha6 are all involved in the hepatocyte growth factor (HGF)/c-Met signal pathway, which plays an important role in the malignant progression of tumors. AIMS The purpose of this study was to explore the expression and prognostic significance of these proteins in pancreatic ductal adenocarcinoma (PDAC). METHODS We used immunohistochemical methods to investigate the expression patterns of CD151, c-Met, and integrin alpha3/alpha6proteins in 71 patients with PDAC and in ten samples of normal pancreatic tissue. We also assessed correlations between these proteins and clinicopathological parameters and survival of PDAC patients using various statistical methods. RESULTS CD151, c-Met, and integrin alpha3/alpha6 were all overexpressed in PDAC. CD151 and c-Met overexpressions were significantly associated with TNM stage (p=0.001 and p=0.038, respectively) and lymph node invasion (p=0.000, p=0.012, respectively). A significant positive linear correlation was found between CD151 and c-Met (r=0.583; p=0.000), integrin alpha3 (r=0.457; p=0.000), and integrin alpha6 (r=0.671; p=0.000). Overexpression of CD151, c-Met, integrin alpha3, or integrin alpha6 was related to poor survival of PDAC patients (p=0.000, p=0.000, p=0.005, and p=0.003, respectively), and CD151 and c-Met were independent factors in prognosis of PDAC. CONCLUSIONS CD151, c-Met, and integrin alpha3/alpha6 were all overexpressed in PDAC. CD151 and c-Met might be new molecular markers to predict the prognosis of PDAC patients.
Collapse
Affiliation(s)
- Guang-Hui Zhu
- Department of General Surgery, Affiliated First People's Hospital, Shanghai Jiao Tong University, 200080, Shanghai, China.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
McCabe KL, Bronner M. Tetraspanin, CD151, is required for maintenance of trigeminal placode identity. J Neurochem 2011; 117:221-30. [PMID: 21250998 DOI: 10.1111/j.1471-4159.2011.07190.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The trigeminal ganglion is the largest of the cranial ganglia and responsible for transmitting sensory information for much of the face. The cell surface glycoprotein CD151 is an early marker of the trigeminal placode, the precursor to the ganglion. Here, we investigate the role of CD151 during specification of trigeminal placode cells in the developing chicken embryo. Expression of the transcription factor Pax3, the earliest known marker of the trigeminal placode, briefly precedes that of CD151, but they then subsequently overlap in the trigeminal placode. Loss of CD151 protein dramatically decreases the number of Pax3+ placode cells in Stage 13-14 embryos, leading to loss of ophthalmic trigeminal neurons by Stages 16 and 17. Although the initial size of the Pax3 population is similar to that in controls, the number of Pax3+ cells decreases with time without alterations in cell death or proliferation. This suggests a role for CD151 in maintenance of the specification state in the trigeminal placode, uncovering the first known role for a tetraspanin in a developmental system.
Collapse
|