1
|
Sahu VK, Lokhande KB, Swamy VK, Basu S, Ranjan A. Role of the transmembrane polar residues on CD151 in cholesterol binding. J Biomol Struct Dyn 2025:1-13. [PMID: 39743780 DOI: 10.1080/07391102.2024.2446663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/30/2024] [Indexed: 01/04/2025]
Abstract
Tetraspanins superfamily proteins have been shown to play an important role in several physiological processes and diseases such as cancer. Transmembrane polar residues of tetraspanins have an implication in regulating the process of cancer metastasis. Tetraspanin CD82 has been demonstrated to exert an anti-metastatic role while mutating polar residues in its transmembrane domains (TMDs) abrogates its metastasis inhibitory role. However, CD151, being pro-metastatic tetraspanin, the role of polar residues in TMDs of CD151 in cholesterol binding and its stability has not been investigated. In this study, we employed bioinformatics analysis of CD151 sequences to determine polar residue in TMDs, molecular docking, dynamics, and normal mode analysis studies to investigate the contribution of polar residues on cholesterol binding by modelling and comparing wild type with polar residue mutant of CD151. The study finds three polar residues viz Asn24, Glu104 and Gln234, in the TMD1, TMD2, and TMD4, respectively, in CD151 that are conserved in most vertebrates indicating their functional significance. Furthermore, we found that these polar residues are required for CD151 stability and facilitate stable interactions with cholesterol. Further cholesterol binding influenced the differences in the distance between large and small extracellular domains of CD151 indicating its important role in the open and closed conformation of the protein. In summary, CD151 exists in open and closed conformation by regulating its interactions with cholesterol. This interaction may modulate the cell membrane dynamics suggesting its potential implication in cancer metastasis.
Collapse
Affiliation(s)
- Vishal Kumar Sahu
- Cancer and Translational Research Centre, Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Kiran Bharat Lokhande
- Bioinformatics Research Laboratory, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Patil Vidyapeeth, Pune, Maharashtra, India
- Translational Bioinformatics and Computational Genomics Research Lab, Department of Life Sciences, Shiv Nadar Institution of Eminence, Gautam Buddha Nagar, Uttar Pradesh, India
| | - Venkateswar K Swamy
- Bioinformatics Research Laboratory, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Patil Vidyapeeth, Pune, Maharashtra, India
- Bioinformatics Research Group, MIT School of Bioengineering Science and Research, MIT- ADT University, Pune, Maharashtra, India
| | - Soumya Basu
- Cancer and Translational Research Centre, Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Amit Ranjan
- Cancer and Translational Research Centre, Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| |
Collapse
|
2
|
Gallo S, Folco CB, Crepaldi T. The MET Oncogene Network of Interacting Cell Surface Proteins. Int J Mol Sci 2024; 25:13692. [PMID: 39769452 PMCID: PMC11728269 DOI: 10.3390/ijms252413692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/11/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
The MET oncogene, encoding the hepatocyte growth factor (HGF) receptor, plays a key role in tumorigenesis, invasion, and resistance to therapy, yet its full biological functions and activation mechanisms remain incompletely understood. A feature of MET is its extensive interaction network, encompassing the following: (i) receptor tyrosine kinases (RTKs); (ii) co-receptors (e.g., CDCP1, Neuropilin1); (iii) adhesion molecules (e.g., integrins, tetraspanins); (iv) proteases (e.g., ADAM10); and (v) other receptors (e.g., CD44, plexins, GPCRs, and NMDAR). These interactions dynamically modulate MET's activation, signaling, intracellular trafficking, and degradation, enhancing its functional versatility and oncogenic potential. This review offers current knowledge on MET's partnerships, focusing on their functional impact on signaling output, therapeutic resistance, and cellular behavior. Finally, we evaluate emerging combination therapies targeting MET and its interactors, highlighting their potential to overcome resistance and improve clinical outcomes. By exploring the complex interplay within the MET network of interacting cell surface proteins, this review provides insights into advancing anti-cancer strategies and understanding the broader implications of RTK crosstalk in oncology.
Collapse
Affiliation(s)
- Simona Gallo
- Department of Oncology, University of Turin, Regione Gonzole 10, 10143 Orbassano, Italy; (S.G.); (C.B.F.)
- Candiolo Cancer Institute, FPO-IRCCS, SP142, Km 3.95, 10060 Candiolo, Italy
| | - Consolata Beatrice Folco
- Department of Oncology, University of Turin, Regione Gonzole 10, 10143 Orbassano, Italy; (S.G.); (C.B.F.)
- Candiolo Cancer Institute, FPO-IRCCS, SP142, Km 3.95, 10060 Candiolo, Italy
| | - Tiziana Crepaldi
- Department of Oncology, University of Turin, Regione Gonzole 10, 10143 Orbassano, Italy; (S.G.); (C.B.F.)
- Candiolo Cancer Institute, FPO-IRCCS, SP142, Km 3.95, 10060 Candiolo, Italy
| |
Collapse
|
3
|
Khan R, Azhar M, Umair M. Decoding the Genes Orchestrating Egg and Sperm Fusion Reactions and Their Roles in Fertility. Biomedicines 2024; 12:2850. [PMID: 39767756 PMCID: PMC11673484 DOI: 10.3390/biomedicines12122850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/07/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Mammalian fertilization is a complex and highly regulated process that has garnered significant attention, particularly with advancements in assisted reproductive technologies such as in vitro fertilization (IVF). The fusion of egg and sperm involves a sequence of molecular and cellular events, including capacitation, the acrosome reaction, adhesion, and membrane fusion. Critical genetic factors, such as IZUMO1, JUNO (also known as FOLR4), CD9, and several others, have been identified as essential mediators in sperm-egg recognition and membrane fusion. Additionally, glycoproteins such as ZP3 within the zona pellucida are crucial for sperm binding and triggering the acrosome reaction. Recent gene-editing technologies, such as CRISPR/Cas9 and conditional knockout models, have facilitated the functional annotation of genes such as SPAM1 and ADAM family members, further elucidating their roles in capacitation and adhesion. Furthermore, the integration of CRISPR-Cas9 with omics technologies, including transcriptomics, proteomics, and lipidomics, has unlocked new avenues for identifying previously unknown genetic players and pathways involved in fertilization. For instance, transcriptomics can uncover gene expression profiles during gamete maturation, while proteomics identifies key protein interactions critical for processes such as capacitation and the acrosome reaction. Lipidomics adds another dimension by revealing how membrane composition influences gamete fusion. Together, these tools enable the discovery of novel genes, pathways, and molecular mechanisms involved in fertility, providing insights that were previously unattainable. These approaches not only deepen our molecular understanding of fertility mechanisms but also hold promise for refining diagnostic tools and therapeutic interventions for infertility. This review summarizes the current molecular insights into genes orchestrating fertilization and highlights cutting-edge methodologies that propel the field toward novel discoveries. By integrating these findings, this review aims to provide valuable knowledge for clinicians, researchers, and technologists in the field of reproductive biology and assisted reproductive technologies.
Collapse
Affiliation(s)
- Ranjha Khan
- Department of Pediatrics, University of California, San Francisco, CA94143, USA;
| | - Muhammad Azhar
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China;
| | - Muhammad Umair
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia
| |
Collapse
|
4
|
Ruangsuwast A, Smout MJ, Brindley PJ, Loukas A, Laha T, Chaiyadet S. Tetraspanins from the liver fluke Opisthorchis viverrini stimulate cholangiocyte migration and inflammatory cytokine production. Folia Parasitol (Praha) 2023; 70:2023.017. [PMID: 37752807 DOI: 10.14411/fp.2023.017] [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: 05/16/2023] [Accepted: 07/26/2023] [Indexed: 09/28/2023]
Abstract
The liver fluke Opisthorchis viverrini (Poirier, 1886) (Digenea) secretes extracellular vesicles (EVs) bearing CD63-like tetraspanins on their surface. Fluke EVs are actively internalised by host cholangiocytes in the bile ducts, where they drive pathology and promote neoplasia through induction of cellular proliferation and secretion of inflammatory cytokines. We investigated the effects of tetraspanins of the CD63 superfamily by co-culturing recombinant forms of the large extracellular loop (LEL) of O. viverrini tetraspanin-2 (rLEL-Ov-TSP-2) and tetraspanin-3 (rLEL-Ov-TSP-3) with non-cancerous human bile duct (H69) and cholangiocarcinoma (CCA, M213) cell lines. The results showed that cell lines co-cultured with excretory/secretory products from adult O. viverrini (Ov-ES) underwent significantly increased cell proliferation at 48 hours but not 24 hours compared to untreated control cells (P < 0.05), whereas rLEL-Ov-TSP-3 co-culture resulted in significantly increased cell proliferation at both 24 hours (P < 0.05) and 48 hours (P < 0.01) time points. In like fashion, H69 cholangiocytes co-cultured with both Ov-ES and rLEL-Ov-TSP-3 underwent significantly elevated Il-6 and Il-8 gene expression for at least one of the time points assessed. Finally, both rLEL-Ov-TSP-2 and rLEL-Ov-TSP-3 significantly enhanced migration of both M213 and H69 cell lines. These findings indicated that O. viverrini CD63 family tetraspanins can promote a cancerous microenvironment by enhancing innate immune responses and migration of biliary epithelial cells.
Collapse
Affiliation(s)
- Apisit Ruangsuwast
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Michael J Smout
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Paul J Brindley
- Department of Microbiology, Immunology and Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, D.C., USA
| | - Alex Loukas
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Thewarach Laha
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sujittra Chaiyadet
- Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand *Address for correspondence: Sujittra Chaiyadet, Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand. E-mail
| |
Collapse
|
5
|
Liu Y, Zhu Z, Li Y, Yang M, Hu Q. Migrasomes released by HSV-2-infected cells serve as a conveyance for virus spread. Virol Sin 2023; 38:643-645. [PMID: 37295496 PMCID: PMC10436035 DOI: 10.1016/j.virs.2023.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 06/05/2023] [Indexed: 06/12/2023] Open
Abstract
•Cells infected with HSV-2 release migrasomes containing HSV-2 virions. •HSV-2 in the isolated migrasomes can be transmitted to uninfected cells and cause productive infection. •It is the first time that migrasomes have been found to play a role in virus spread.
Collapse
Affiliation(s)
- Yalan Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; Hubei Jiangxia Laboratory, Wuhan, 430200, China.
| | - Zhiyuan Zhu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuncheng Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mengshi Yang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Qinxue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; Institute for Infection and Immunity, St George's University of London, London, SW17 0RE, UK.
| |
Collapse
|
6
|
Ruangsuwast A, Smout MJ, Brindley PJ, Loukas A, Laha T, Chaiyadet S. Tetraspanins from Opisthorchis viverrini stimulate cholangiocyte migration and inflammatory cytokine production. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544604. [PMID: 37398394 PMCID: PMC10312640 DOI: 10.1101/2023.06.12.544604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
The liver fluke Opsithorchis viverrini secretes extracellular vesicles (EVs) bearing CD63-like tetraspanins on their surface. Fluke EVs are actively internalized by host cholangiocytes in the bile ducts, where they drive pathology and promote neoplasia through induction of cellular proliferation and secretion of inflammatory cytokines. We investigated the effects of tetraspanins of the CD63 superfamily by co-culturing recombinant forms of the large extracellular loop (LEL) of O. viverrini tetraspanin-2 (rLEL- Ov -TSP-2) and tetraspanin-3 (rLEL- Ov -TSP-3) with non-cancerous human bile duct (H69) and cholangiocarcinoma (CCA, M213) cell lines. The results showed that cell lines co-cultured with excretory/secretory products from adult O. viverrini ( Ov- ES) underwent significantly increased cell proliferation at 48 hours but not 24 hours compared to untreated control cells ( P <0.05), whereas rLEL- Ov -TSP-3 co-culture resulted in significantly increased cell proliferation at both 24 hr ( P <0.05) and 48 hr ( P <0.01) time points. In like fashion, H69 cholangiocytes co-cultured with both Ov -ES and rLEL- Ov -TSP-3 underwent significantly elevated Il-6 and Il-8 gene expression for at least one of the time points assessed. Finally, both rLEL- Ov -TSP-and rLEL- Ov -TSP-3 significantly enhanced migration of both M213 and H69 cell lines. These findings indicated that O. viverrini CD63 family tetraspanins can promote a cancerous microenvironment by enhancing innate immune responses and migration of biliary epithelial cells.
Collapse
Affiliation(s)
- Apisit Ruangsuwast
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Michael J. Smout
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, 4878, Australia
| | - Paul J. Brindley
- Department of Microbiology, Immunology and Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, D.C. 20037, USA
| | - Alex Loukas
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, 4878, Australia
| | - Thewarach Laha
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sujittra Chaiyadet
- Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand
| |
Collapse
|
7
|
Bovari-Biri J, Garai K, Banfai K, Csongei V, Pongracz JE. miRNAs as Predictors of Barrier Integrity. BIOSENSORS 2023; 13:bios13040422. [PMID: 37185497 PMCID: PMC10136429 DOI: 10.3390/bios13040422] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 05/17/2023]
Abstract
The human body has several barriers that protect its integrity and shield it from mechanical, chemical, and microbial harm. The various barriers include the skin, intestinal and respiratory epithelia, blood-brain barrier (BBB), and immune system. In the present review, the focus is on the physical barriers that are formed by cell layers. The barrier function is influenced by the molecular microenvironment of the cells forming the barriers. The integrity of the barrier cell layers is maintained by the intricate balance of protein expression that is partly regulated by microRNAs (miRNAs) both in the intracellular space and the extracellular microenvironment. The detection of changes in miRNA patterns has become a major focus of diagnostic, prognostic, and disease progression, as well as therapy-response, markers using a great variety of detection systems in recent years. In the present review, we highlight the importance of liquid biopsies in assessing barrier integrity and challenges in differential miRNA detection.
Collapse
Affiliation(s)
- Judit Bovari-Biri
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, 2 Rokus Str, H-7624 Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, 20 Ifjusag Str, H-7624 Pecs, Hungary
| | - Kitti Garai
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, 2 Rokus Str, H-7624 Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, 20 Ifjusag Str, H-7624 Pecs, Hungary
| | - Krisztina Banfai
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, 2 Rokus Str, H-7624 Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, 20 Ifjusag Str, H-7624 Pecs, Hungary
| | - Veronika Csongei
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, 2 Rokus Str, H-7624 Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, 20 Ifjusag Str, H-7624 Pecs, Hungary
| | - Judit E Pongracz
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, 2 Rokus Str, H-7624 Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, 20 Ifjusag Str, H-7624 Pecs, Hungary
| |
Collapse
|
8
|
McLaughlin K, Acreman S, Nawaz S, Cutteridge J, Clark A, Knudsen JG, Denwood G, Spigelman AF, Manning Fox JE, Singh SP, MacDonald PE, Hastoy B, Zhang Q. Loss of tetraspanin-7 expression reduces pancreatic β-cell exocytosis Ca 2+ sensitivity but has limited effect on systemic metabolism. Diabet Med 2022; 39:e14984. [PMID: 36264270 PMCID: PMC9828109 DOI: 10.1111/dme.14984] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/08/2022] [Accepted: 10/18/2022] [Indexed: 01/18/2023]
Abstract
BACKGROUND Tetraspanin-7 (Tspan7) is an islet autoantigen involved in autoimmune type 1 diabetes and known to regulate β-cell L-type Ca2+ channel activity. However, the role of Tspan7 in pancreatic β-cell function is not yet fully understood. METHODS Histological analyses were conducted using immunostaining. Whole-body metabolism was tested using glucose tolerance test. Islet hormone secretion was quantified using static batch incubation or dynamic perifusion. β-cell transmembrane currents, electrical activity and exocytosis were measured using whole-cell patch-clamping and capacitance measurements. Gene expression was studied using mRNA-sequencing and quantitative PCR. RESULTS Tspan7 is expressed in insulin-containing granules of pancreatic β-cells and glucagon-producing α-cells. Tspan7 knockout mice (Tspan7y/- mouse) exhibit reduced body weight and ad libitum plasma glucose but normal glucose tolerance. Tspan7y/- islets have normal insulin content and glucose- or tolbutamide-stimulated insulin secretion. Depolarisation-triggered Ca2+ current was enhanced in Tspan7y/- β-cells, but β-cell electrical activity and depolarisation-evoked exocytosis were unchanged suggesting that exocytosis was less sensitive to Ca2+ . TSPAN7 knockdown (KD) in human pseudo-islets led to a significant reduction in insulin secretion stimulated by 20 mM K+ . Transcriptomic analyses show that TSPAN7 KD in human pseudo-islets correlated with changes in genes involved in hormone secretion, apoptosis and ER stress. Consistent with rodent β-cells, exocytotic Ca2+ sensitivity was reduced in a human β-cell line (EndoC-βH1) following Tspan7 KD. CONCLUSION Tspan7 is involved in the regulation of Ca2+ -dependent exocytosis in β-cells. Its function is more significant in human β-cells than their rodent counterparts.
Collapse
Affiliation(s)
- Kerry McLaughlin
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of Oxford, Churchill HospitalOxfordUK
| | - Samuel Acreman
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of Oxford, Churchill HospitalOxfordUK
- Institute of Neuroscience and Physiology, Department of Physiology, Metabolic Research UnitUniversity of GoteborgGöteborgSweden
| | - Sameena Nawaz
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of Oxford, Churchill HospitalOxfordUK
| | - Joseph Cutteridge
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of Oxford, Churchill HospitalOxfordUK
| | - Anne Clark
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of Oxford, Churchill HospitalOxfordUK
| | - Jakob G. Knudsen
- Section for Cell Biology and Physiology, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| | - Geoffrey Denwood
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of Oxford, Churchill HospitalOxfordUK
| | - Aliya F. Spigelman
- Alberta Diabetes Institute and Department of PharmacologyUniversity of AlbertaEdmontonAlbertaCanada
| | - Jocelyn E. Manning Fox
- Alberta Diabetes Institute and Department of PharmacologyUniversity of AlbertaEdmontonAlbertaCanada
| | | | - Patrick E. MacDonald
- Alberta Diabetes Institute and Department of PharmacologyUniversity of AlbertaEdmontonAlbertaCanada
| | - Benoit Hastoy
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of Oxford, Churchill HospitalOxfordUK
| | - Quan Zhang
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of Oxford, Churchill HospitalOxfordUK
| |
Collapse
|
9
|
Shao S, Piao L, Guo L, Wang J, Wang L, Wang J, Tong L, Yuan X, Zhu J, Fang S, Wang Y. Tetraspanin 7 promotes osteosarcoma cell invasion and metastasis by inducing EMT and activating the FAK-Src-Ras-ERK1/2 signaling pathway. Cancer Cell Int 2022; 22:183. [PMID: 35524311 PMCID: PMC9074275 DOI: 10.1186/s12935-022-02591-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 04/18/2022] [Indexed: 02/08/2023] Open
Abstract
Background Tetraspanins are members of the 4-transmembrane protein superfamily (TM4SF) that function by recruiting many cell surface receptors and signaling proteins into tetraspanin-enriched microdomains (TEMs) that play vital roles in the regulation of key cellular processes including adhesion, motility, and proliferation. Tetraspanin7 (Tspan7) is a member of this superfamily that plays documented roles in hippocampal neurogenesis, synaptic transmission, and malignant transformation in certain tumor types. How Tspan7 influences the onset or progression of osteosarcoma (OS), however, remains to be defined. Herein, this study aimed to explore the relationship between Tspan7 and the malignant progression of OS, and its underlying mechanism of action. Methods In this study, the levels of Tspan7 expression in human OS cell lines were evaluated via qRT-PCR and western blotting. The effect of Tspan7 on proliferation was examined using CCK-8 and colony formation assays, while metastatic role of Tspan7 was assessed by functional assays both in vitro and in vivo. In addition, mass spectrometry and co-immunoprecipitation were performed to verify the interaction between Tspan7 and β1 integrin, and western blotting was used to explore the mechanisms of Tspan7 in OS progresses. Results We found that Tspan7 is highly expressed in primary OS tumors and OS cell lines. Downregulation of Tspan7 significantly suppressed OS growth, metastasis, and attenuated epithelial-mesenchymal transition (EMT), while its overexpression had the opposite effects in vitro. Furthermore, it exhibited reduced OS pulmonary metastases in Tspan7-deleted mice comparing control mice in vivo. Additionally, we proved that Tspan7 interacted with β1 integrin to facilitate OS metastasis through the activation of integrin-mediated downstream FAK-Src-Ras-ERK1/2 signaling pathway. Conclusion In summary, this study demonstrates for the first time that Tspan7 promotes OS metastasis via interacting with β1 integrin and activating the FAK-Src-Ras-ERK1/2 pathway, which could provide rationale for a new therapeutic strategy for OS. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02591-1.
Collapse
Affiliation(s)
- Shijie Shao
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, People's Republic of China
| | - Lianhua Piao
- Institute of Bioinformatics and Medical Engineering, Jiangsu University of Technology, Changzhou, 213000, People's Republic of China.
| | - Liwei Guo
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, People's Republic of China
| | - Jiangsong Wang
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, People's Republic of China
| | - Luhui Wang
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, People's Republic of China
| | - Jiawen Wang
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, People's Republic of China
| | - Lei Tong
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, People's Republic of China
| | - Xiaofeng Yuan
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, People's Republic of China
| | - Junke Zhu
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, People's Republic of China
| | - Sheng Fang
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, People's Republic of China
| | - Yimin Wang
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, People's Republic of China.
| |
Collapse
|
10
|
Di Daniele A, Antonucci Y, Campello S. Migrasomes, new vescicles as Hansel and Gretel white pebbles? Biol Direct 2022; 17:8. [PMID: 35484629 PMCID: PMC9047267 DOI: 10.1186/s13062-022-00321-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 04/16/2022] [Indexed: 02/07/2023] Open
Abstract
Migrasomes, released by migrating cells, belong to the heterogeneous world of extracellular vesicles (EVs). However, they can be distinguished from all other members of EVs by their size, biorigin and protein cargo. As far as we know, they can play important roles in various communication processes, by mediating the release of signals, such as mRNAs, proteins or damaged mitochondria. To extend and better understand the functional roles and importance of migrasomes, it is first essential to well understand the basic molecular mechanisms behind their formation and function. Herein, we endeavor to provide a brief and up-to-date description of migrasome biogenesis, release, characterization, biological properties and functional activities in cell-to-cell communication, and we will discuss and propose putative new functions for these vesicles.
Collapse
Affiliation(s)
- Arianna Di Daniele
- grid.6530.00000 0001 2300 0941Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
| | - Ylenia Antonucci
- grid.6530.00000 0001 2300 0941Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
| | - Silvia Campello
- grid.6530.00000 0001 2300 0941Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
| |
Collapse
|
11
|
Becic A, Leifeld J, Shaukat J, Hollmann M. Tetraspanins as Potential Modulators of Glutamatergic Synaptic Function. Front Mol Neurosci 2022; 14:801882. [PMID: 35046772 PMCID: PMC8761850 DOI: 10.3389/fnmol.2021.801882] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/07/2021] [Indexed: 12/16/2022] Open
Abstract
Tetraspanins (Tspans) comprise a membrane protein family structurally defined by four transmembrane domains and intracellular N and C termini that is found in almost all cell types and tissues of eukaryotes. Moreover, they are involved in a bewildering multitude of diverse biological processes such as cell adhesion, motility, protein trafficking, signaling, proliferation, and regulation of the immune system. Beside their physiological roles, they are linked to many pathophysiological phenomena, including tumor progression regulation, HIV-1 replication, diabetes, and hepatitis. Tetraspanins are involved in the formation of extensive protein networks, through interactions not only with themselves but also with numerous other specific proteins, including regulatory proteins in the central nervous system (CNS). Interestingly, recent studies showed that Tspan7 impacts dendritic spine formation, glutamatergic synaptic transmission and plasticity, and that Tspan6 is correlated with epilepsy and intellectual disability (formerly known as mental retardation), highlighting the importance of particular tetraspanins and their involvement in critical processes in the CNS. In this review, we summarize the current knowledge of tetraspanin functions in the brain, with a particular focus on their impact on glutamatergic neurotransmission. In addition, we compare available resolved structures of tetraspanin family members to those of auxiliary proteins of glutamate receptors that are known for their modulatory effects.
Collapse
|
12
|
Justo BL, Jasiulionis MG. Characteristics of TIMP1, CD63, and β1-Integrin and the Functional Impact of Their Interaction in Cancer. Int J Mol Sci 2021; 22:9319. [PMID: 34502227 PMCID: PMC8431149 DOI: 10.3390/ijms22179319] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 01/03/2023] Open
Abstract
Tissue Inhibitor of Metalloproteases 1, also known as TIMP-1, is named for its well-established function of inhibiting the proteolytic activity of matrix metalloproteases. Given this function, many studies were carried out to verify if TIMP-1 was able to interrupt processes such as tumor cell invasion and metastasis. In contrast, many studies have shown that TIMP-1 expression is increased in several types of tumors, and this increase was correlated with a poor prognosis and lower survival in cancer patients. Later, it was shown that TIMP-1 is also able to modulate cell behavior through the induction of signaling pathways involved in cell growth, proliferation, and survival. The mechanisms involved in the regulation of the pleiotropic functions of TIMP-1 are still poorly understood. Thus, this review aimed to present literature data that show its ability to form a membrane complex with CD63 and β1-integrin, and point to N-glycosylation as a potential regulatory mechanism of the functions exerted by TIMP-1. This article reviewed the characteristics and functions performed individually by TIMP1, CD63, and β1-integrin, the roles of the TIMP-1/CD63/β1-integrin complex, both in a physiological context and in cancer, and the regulatory mechanisms involved in its assembly.
Collapse
Affiliation(s)
| | - Miriam Galvonas Jasiulionis
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Rua Pedro de Toledo 669, 5 Floor, São Paulo 04039-032, Brazil;
| |
Collapse
|
13
|
Roupakia E, Chavdoula E, Karpathiou G, Vatsellas G, Chatzopoulos D, Mela A, Gillette JM, Kriegsmann K, Kriegsmann M, Batistatou A, Goussia A, Marcu KB, Karteris E, Klinakis A, Kolettas E. Canonical NF-κB Promotes Lung Epithelial Cell Tumour Growth by Downregulating the Metastasis Suppressor CD82 and Enhancing Epithelial-to-Mesenchymal Cell Transition. Cancers (Basel) 2021; 13:cancers13174302. [PMID: 34503110 PMCID: PMC8428346 DOI: 10.3390/cancers13174302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/17/2021] [Accepted: 08/24/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Canonical NF-κB signalling pathway acts as a tumour promoter in several types of cancer including non-small cell lung cancer (NSCLC), but the mechanism(s) by which it contributes to NSCLC is still under investigation. We show here that NF-κB RelA/p65 is required for the tumour growth of human NSCLC cells grown in vivo as xenografts in immune-compromised mice. RNA-seq transcriptome profile analysis identified the metastasis suppressor CD82/KAI1/TSPAN27 as a canonical NF-κB target. Loss of CD82 correlated with malignancy. RelA/p65 stimulates cell migration and epithelial-to-mesenchymal cell transition (EMT), mediated, in part, by CD82/KAI1, through integrin-mediated signalling, thus, identifying a mechanism mediating NF-κB RelA/p65 lung tumour promoting function. Abstract Background: The development of non-small cell lung cancer (NSCLC) involves the progressive accumulation of genetic and epigenetic changes. These include somatic oncogenic KRAS and EGFR mutations and inactivating TP53 tumour suppressor mutations, leading to activation of canonical NF-κB. However, the mechanism(s) by which canonical NF-κB contributes to NSCLC is still under investigation. Methods: Human NSCLC cells were used to knock-down RelA/p65 (RelA/p65KD) and investigate its impact on cell growth, and its mechanism of action by employing RNA-seq analysis, qPCR, immunoblotting, immunohistochemistry, immunofluorescence and functional assays. Results: RelA/p65KD reduced the proliferation and tumour growth of human NSCLC cells grown in vivo as xenografts in immune-compromised mice. RNA-seq analysis identified canonical NF-κB targets mediating its tumour promoting function. RelA/p65KD resulted in the upregulation of the metastasis suppressor CD82/KAI1/TSPAN27 and downregulation of the proto-oncogene ROS1, and LGR6 involved in Wnt/β-catenin signalling. Immunohistochemical and bioinformatics analysis of human NSCLC samples showed that CD82 loss correlated with malignancy. RelA/p65KD suppressed cell migration and epithelial-to-mesenchymal cell transition (EMT), mediated, in part, by CD82/KAI1, through integrin-mediated signalling involving the mitogenic ERK, Akt1 and Rac1 proteins. Conclusions: Canonical NF-κB signalling promotes NSCLC, in part, by downregulating the metastasis suppressor CD82/KAI1 which inhibits cell migration, EMT and tumour growth.
Collapse
Affiliation(s)
- Eugenia Roupakia
- Laboratory of Biology, School of Medicine, Faculty of Health Sciences, Institute of Biosciences, University Research Centre, University of Ioannina, University Campus, 45110 Ioannina, Greece;
- Biomedical Research Division, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, University of Ioannina Campus, 45115 Ioannina, Greece;
| | - Evangelia Chavdoula
- Biomedical Research Division, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, University of Ioannina Campus, 45115 Ioannina, Greece;
- Biomedical Research Foundation, Academy of Athens (BRFAA), 4 Soranou Ephessiou Street, 11527 Athens, Greece; (G.V.); (D.C.); (K.B.M.); (A.K.)
| | - Georgia Karpathiou
- Laboratory of Pathology, School of Medicine, Faculty of Health Sciences, University of Ioannina, 45500 Ioannina, Greece; (G.K.); (A.B.); (A.G.)
| | - Giannis Vatsellas
- Biomedical Research Foundation, Academy of Athens (BRFAA), 4 Soranou Ephessiou Street, 11527 Athens, Greece; (G.V.); (D.C.); (K.B.M.); (A.K.)
| | - Dimitrios Chatzopoulos
- Biomedical Research Foundation, Academy of Athens (BRFAA), 4 Soranou Ephessiou Street, 11527 Athens, Greece; (G.V.); (D.C.); (K.B.M.); (A.K.)
| | - Angeliki Mela
- Department of Pathology and Cell Biology Columbia University Medical Center, Irving Comprehensive Cancer Research Center, Columbia University, New York, NY 10032, USA;
| | - Jennifer M. Gillette
- Department of Pathology, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA;
| | - Katharina Kriegsmann
- Department of Internal Medicine V, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Mark Kriegsmann
- Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Anna Batistatou
- Laboratory of Pathology, School of Medicine, Faculty of Health Sciences, University of Ioannina, 45500 Ioannina, Greece; (G.K.); (A.B.); (A.G.)
| | - Anna Goussia
- Laboratory of Pathology, School of Medicine, Faculty of Health Sciences, University of Ioannina, 45500 Ioannina, Greece; (G.K.); (A.B.); (A.G.)
| | - Kenneth B. Marcu
- Biomedical Research Foundation, Academy of Athens (BRFAA), 4 Soranou Ephessiou Street, 11527 Athens, Greece; (G.V.); (D.C.); (K.B.M.); (A.K.)
- Department of Biochemistry and Cell Biology, Microbiology and Pathology, Stony Brook University, New York, NY 11794, USA
| | - Emmanouil Karteris
- Division of Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, Middlesex, London UB8 PH, UK;
| | - Apostolos Klinakis
- Biomedical Research Foundation, Academy of Athens (BRFAA), 4 Soranou Ephessiou Street, 11527 Athens, Greece; (G.V.); (D.C.); (K.B.M.); (A.K.)
| | - Evangelos Kolettas
- Laboratory of Biology, School of Medicine, Faculty of Health Sciences, Institute of Biosciences, University Research Centre, University of Ioannina, University Campus, 45110 Ioannina, Greece;
- Biomedical Research Division, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, University of Ioannina Campus, 45115 Ioannina, Greece;
- Correspondence: ; Tel.: +30-26510-07578; Fax: +30-26510-07863
| |
Collapse
|
14
|
Kim M, Lin J, Huh JE, Park JH, Go M, Lee H, Hwang D, Kim HS, Kim T, Lee D, Lee SY. Tetraspanin 7 regulates osteoclast function through association with the RANK/αvβ3 integrin complex. J Cell Physiol 2021; 237:846-855. [PMID: 34407208 DOI: 10.1002/jcp.30559] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/20/2021] [Accepted: 08/10/2021] [Indexed: 12/13/2022]
Abstract
Actin rings are unique structures that facilitate the attachment of osteoclasts to the bone matrix during bone resorption. Previous studies have shown that tetraspanin7 (TSPAN7) plays an important role in the reorganization of the cytoskeleton necessary for the bone-resorbing activity of osteoclasts. However, questions remain as to the mechanisms by which TSPAN7 regulates this cytoskeletal rearrangement. In this study, we investigated the roles of TSPAN7 in osteoclasts by deleting the Tm4sf2 gene in mice, which encodes TSPAN7. The Tm4sf2 global knockout model showed protective effects on pathological bone loss, but no discernible changes in bone phenotypes under physiological conditions. In vitro study revealed that ablation of Tm4sf2 caused significant defects in integrin-mediated actin ring formation, thereby leading to significantly decreased bone resorption. Additionally, we demonstrated an association between TSPAN7 and the receptor activator of nuclear factor-кB/αvβ3 integrin. Overall, our findings suggest that TSPAN7 acts as a novel modulator regulating the bone-resorbing function of osteoclasts.
Collapse
Affiliation(s)
- Minhee Kim
- Department of Life Science, Ewha Womans University, Seoul, South Korea.,Department of Life Science, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Jingjing Lin
- Department of Life Science, Ewha Womans University, Seoul, South Korea.,Department of Life Science, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Jeong-Eun Huh
- Department of Life Science, Ewha Womans University, Seoul, South Korea.,Department of Life Science, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Jin Hee Park
- Department of Life Science, Ewha Womans University, Seoul, South Korea.,Department of Life Science, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Miyeon Go
- Department of Life Science, Ewha Womans University, Seoul, South Korea.,Department of Life Science, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Hana Lee
- Department of Biomedical Engineering, Yonsei University, Wonju, South Korea
| | - Donghyun Hwang
- Department of Biomedical Engineering, Yonsei University, Wonju, South Korea
| | - Han Sung Kim
- Department of Biomedical Engineering, Yonsei University, Wonju, South Korea
| | - Taesoo Kim
- Department of Life Science, Ewha Womans University, Seoul, South Korea.,Department of Life Science, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Daekee Lee
- Department of Life Science, Ewha Womans University, Seoul, South Korea
| | - Soo Young Lee
- Department of Life Science, Ewha Womans University, Seoul, South Korea.,Department of Life Science, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| |
Collapse
|
15
|
Zhu J, Cai T, Zhou J, Du W, Zeng Y, Liu T, Fu Y, Li Y, Qian Q, Yang XH, Li Q, Huang JA, Liu Z. CD151 drives cancer progression depending on integrin α3β1 through EGFR signaling in non-small cell lung cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:192. [PMID: 34108040 PMCID: PMC8191020 DOI: 10.1186/s13046-021-01998-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 05/28/2021] [Indexed: 01/07/2023]
Abstract
Background Tetraspanins CD151, a transmembrane 4 superfamily protein, has been identified participating in the initiation of a variety of cancers. However, the precise function of CD151 in non-small cell lung cancer (NSCLC) remains unclear. Here, we addressed the pro-tumoral role of CD151 in NSCLC by targeting EGFR/ErbB2 which favors tumor proliferation, migration and invasion. Methods First, the mRNA expression levels of CD151 in NSCLC tissues and cell lines were measured by RT-PCR. Meanwhile, CD151 and its associated proteins were analyzed by western blotting. The expression levels of CD151 in NSCLC samples and its paired adjacent lung tissues were then verified by Immunohistochemistry. The protein interactions are evaluated by co-immunoprecipitation. Flow cytometry was applied to cell cycle analysis. CCK-8, EdU Incorporation, and clonogenic assays were used to analyze cell viability. Wound healing, transwell migration, and matrigel invasion assays were utilized to assess the motility of tumor cells. To investigate the role of CD151 in vivo, lung carcinoma xenograft mouse model was applied. Results High CD151 expression was identified in NSCLC tissues and cell lines, and its high expression was significantly associated with poor prognosis of NSCLC patients. Further, knockdown of CD151 in vitro inhibited tumor proliferation, migration, and invasion. Besides, inoculation of nude mice with CD151-overexpressing tumor cells exhibited substantial tumor proliferation compared to that in control mice which inoculated with vector-transfected tumor cells. Noteworthy, we found that overexpression of CD151 conferred cell migration and invasion by interacting with integrins. We next sought to demonstrate that CD151 regulated downstream signaling pathways via activation of EGFR/ErbB2 in NSCLC cells. Therefore, we infer that CD151 probably affects the sensitivity of NSCLC in response to anti-cancer drugs. Conclusions Based on these results, we demonstrated a new mechanism of CD151-mediated tumor progression by targeting EGFR/ErbB2 signaling pathway, by which CD151 promotes NSCLC proliferation, migration, and invasion, which may considered as a potential target of NSCLC treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01998-4.
Collapse
Affiliation(s)
- Jianjie Zhu
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Institute of Respiratory Diseases, Soochow University, 215006, Suzhou, China.,Suzhou Key Laboratory for Respiratory Diseases, 215006, Suzhou, China
| | - Tingting Cai
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Institute of Respiratory Diseases, Soochow University, 215006, Suzhou, China
| | - Jieqi Zhou
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Institute of Respiratory Diseases, Soochow University, 215006, Suzhou, China
| | - Wenwen Du
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Institute of Respiratory Diseases, Soochow University, 215006, Suzhou, China
| | - Yuanyuan Zeng
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Institute of Respiratory Diseases, Soochow University, 215006, Suzhou, China.,Suzhou Key Laboratory for Respiratory Diseases, 215006, Suzhou, China
| | - Ting Liu
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Institute of Respiratory Diseases, Soochow University, 215006, Suzhou, China
| | - Yulong Fu
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Institute of Respiratory Diseases, Soochow University, 215006, Suzhou, China
| | - Yue Li
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Institute of Respiratory Diseases, Soochow University, 215006, Suzhou, China
| | - Qian Qian
- Department of Medicine, Division of Allergy and Clinical Immunology, National Jewish Health, Denver, 80206, USA
| | - Xiuwei H Yang
- Department of Pharmacology and Nutritional Sciences, Department of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky, Lexington, KY, 40506, USA
| | - Qinglin Li
- Department of Traditional Chinese Medicine, Cancer Hospital of the University of Chinese Academy of Sciences, 310022, Hangzhou, People's Republic of China.
| | - Jian-An Huang
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China. .,Institute of Respiratory Diseases, Soochow University, 215006, Suzhou, China. .,Suzhou Key Laboratory for Respiratory Diseases, 215006, Suzhou, China.
| | - Zeyi Liu
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China. .,Institute of Respiratory Diseases, Soochow University, 215006, Suzhou, China. .,Suzhou Key Laboratory for Respiratory Diseases, 215006, Suzhou, China.
| |
Collapse
|
16
|
Exploring interactions between extracellular vesicles and cells for innovative drug delivery system design. Adv Drug Deliv Rev 2021; 173:252-278. [PMID: 33798644 DOI: 10.1016/j.addr.2021.03.017] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/15/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) are submicron cell-secreted structures containing proteins, nucleic acids and lipids. EVs can functionally transfer these cargoes from one cell to another to modulate physiological and pathological processes. Due to their presumed biocompatibility and capacity to circumvent canonical delivery barriers encountered by synthetic drug delivery systems, EVs have attracted considerable interest as drug delivery vehicles. However, it is unclear which mechanisms and molecules orchestrate EV-mediated cargo delivery to recipient cells. Here, we review how EV properties have been exploited to improve the efficacy of small molecule drugs. Furthermore, we explore which EV surface molecules could be directly or indirectly involved in EV-mediated cargo transfer to recipient cells and discuss the cellular reporter systems with which such transfer can be studied. Finally, we elaborate on currently identified cellular processes involved in EV cargo delivery. Through these topics, we provide insights in critical effectors in the EV-cell interface which may be exploited in nature-inspired drug delivery strategies.
Collapse
|
17
|
Ahmed W, Neelakanta G, Sultana H. Tetraspanins as Potential Therapeutic Candidates for Targeting Flaviviruses. Front Immunol 2021; 12:630571. [PMID: 33968023 PMCID: PMC8097176 DOI: 10.3389/fimmu.2021.630571] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/06/2021] [Indexed: 12/18/2022] Open
Abstract
Tetraspanin family of proteins participates in numerous fundamental signaling pathways involved in viral transmission, virus-specific immunity, and virus-mediated vesicular trafficking. Studies in the identification of novel therapeutic candidates and strategies to target West Nile virus, dengue and Zika viruses are highly warranted due to the failure in development of vaccines. Recent evidences have shown that the widely distributed tetraspanin proteins may provide a platform for the development of novel therapeutic approaches. In this review, we discuss the diversified and important functions of tetraspanins in exosome/extracellular vesicle biology, virus-host interactions, virus-mediated vesicular trafficking, modulation of immune mechanism(s), and their possible role(s) in host antiviral defense mechanism(s) through interactions with noncoding RNAs. We also highlight the role of tetraspanins in the development of novel therapeutics to target arthropod-borne flaviviral diseases.
Collapse
Affiliation(s)
- Waqas Ahmed
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States
| | - Girish Neelakanta
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States.,Center for Molecular Medicine, Old Dominion University, Norfolk, VA, United States
| | - Hameeda Sultana
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States.,Center for Molecular Medicine, Old Dominion University, Norfolk, VA, United States
| |
Collapse
|
18
|
Sun G, Chen J, Ding Y, Wren JD, Xu F, Lu L, Wang Y, Wang DW, Zhang XA. A Bioinformatics Perspective on the Links Between Tetraspanin-Enriched Microdomains and Cardiovascular Pathophysiology. Front Cardiovasc Med 2021; 8:630471. [PMID: 33860000 PMCID: PMC8042132 DOI: 10.3389/fcvm.2021.630471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/15/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Tetraspanins and integrins are integral membrane proteins. Tetraspanins interact with integrins to modulate the dynamics of adhesion, migration, proliferation, and signaling in the form of membrane domains called tetraspanin-enriched microdomains (TEMs). TEMs also contain other cell adhesion proteins like immunoglobulin superfamily (IgSF) proteins and claudins. Cardiovascular functions of these TEM proteins have emerged and remain to be further revealed. Objectives: The aims of this study are to explore the roles of these TEM proteins in the cardiovascular system using bioinformatics tools and databases and to highlight the TEM proteins that may functionally associate with cardiovascular physiology and pathology. Methods: For human samples, three databases-GTEx, NCBI-dbGaP, and NCBI-GEO-were used for the analyses. The dbGaP database was used for GWAS analysis to determine the association between target genes and human phenotypes. GEO is an NCBI public repository that archives genomics data. GTEx was used for the analyses of tissue-specific mRNA expression levels and eQTL. For murine samples, GeneNetwork was used to find gene-phenotype correlations and gene-gene correlations of expression levels in mice. The analysis of cardiovascular data was the focus of this study. Results: Some integrins and tetraspanins, such as ITGA8 and Cd151, are highly expressed in the human cardiovascular system. TEM components are associated with multiple cardiovascular pathophysiological events in humans. GWAS and GEO analyses showed that human Cd82 and ITGA9 are associated with blood pressure. Data from mice also suggest that various cardiovascular phenotypes are correlated with integrins and tetraspanins. For instance, Cd82 and ITGA9, again, have correlations with blood pressure in mice. Conclusion: ITGA9 is related to blood pressure in both species. KEGG analysis also linked ITGA9 to metabolism and MAPK signaling pathway. This work provides an example of using integrated bioinformatics approaches across different species to identify the connections of structurally and/or functionally related molecules to certain categories of diseases.
Collapse
Affiliation(s)
- Ge Sun
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Junxiong Chen
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Yingjun Ding
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Jonathan D. Wren
- Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Fuyi Xu
- University of Tennessee Health Science Center, Memphis, TN, United States
| | - Lu Lu
- University of Tennessee Health Science Center, Memphis, TN, United States
| | - Yan Wang
- Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Dao-wen Wang
- Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Xin A. Zhang
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
19
|
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: 29] [Impact Index Per Article: 7.3] [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
|
20
|
Murru L, Ponzoni L, Longatti A, Mazzoleni S, Giansante G, Bassani S, Sala M, Passafaro M. Lateral habenula dysfunctions in Tm4sf2 -/y mice model for neurodevelopmental disorder. Neurobiol Dis 2021; 148:105189. [PMID: 33227491 PMCID: PMC7840593 DOI: 10.1016/j.nbd.2020.105189] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/30/2020] [Accepted: 11/17/2020] [Indexed: 12/25/2022] Open
Abstract
Mutations in the TM4SF2 gene, which encodes TSPAN7, cause a severe form of intellectual disability (ID) often comorbid with autism spectrum disorder (ASD). Recently, we found that TM4SF2 loss in mice affects cognition. Here, we report that Tm4sf2-/y mice, beyond an ID-like phenotype, display altered sociability, increased repetitive behaviors, anhedonic- and depressive-like states. Cognition relies on the integration of information from several brain areas. In this context, the lateral habenula (LHb) is strategically positioned to coordinate the brain regions involved in higher cognitive functions. Furthermore, in Tm4sf2-/y mice we found that LHb neurons present hypoexcitability, aberrant neuronal firing pattern and altered sodium and potassium voltage-gated ion channels function. Interestingly, we also found a reduced expression of voltage-gated sodium channel and a hyperactivity of the PKC-ERK pathway, a well-known modulator of ion channels activity, which might explain the functional phenotype showed by Tm4sf2-/y mice LHb neurons. These findings support Tm4sf2-/y mice as useful in modeling some ASD-like symptoms. Additionally, we can speculate that LHb functional alteration in Tm4sf2-/y mice might play a role in the disease pathophysiology.
Collapse
Affiliation(s)
- Luca Murru
- Institute of Neuroscience, CNR, Milan 20129, Italy; NeuroMI Milan Center for Neuroscience, Università Milano-Bicocca, Milan 20126, Italy.
| | - Luisa Ponzoni
- Department of Medical Biotechnology and Translational Medicine, Università di Milano, Segrate, MI 20090, Italy
| | | | - Sara Mazzoleni
- Institute of Neuroscience, CNR, Milan 20129, Italy; Department of Medical Biotechnology and Translational Medicine, Università di Milano, Segrate, MI 20090, Italy
| | | | - Silvia Bassani
- Institute of Neuroscience, CNR, Milan 20129, Italy; NeuroMI Milan Center for Neuroscience, Università Milano-Bicocca, Milan 20126, Italy
| | - Mariaelvina Sala
- Institute of Neuroscience, CNR, Milan 20129, Italy; NeuroMI Milan Center for Neuroscience, Università Milano-Bicocca, Milan 20126, Italy
| | - Maria Passafaro
- Institute of Neuroscience, CNR, Milan 20129, Italy; NeuroMI Milan Center for Neuroscience, Università Milano-Bicocca, Milan 20126, Italy.
| |
Collapse
|
21
|
Abstract
A novel tool for the presentation of peptides and small proteins on the surface of human cells has been developed. Our tANCHOR system utilizes tetraspanin anchors containing heterologous amino acid sequences inserted instead of the large extracellular loop. This technology allows a highly effective extracellular display of epitopes for antibody binding studies and many other potential applications.
Collapse
|
22
|
Pierce CF, Brown VR, Olsen SC, Boggiatto P, Pedersen K, Miller RS, Speidel SE, Smyser TJ. Loci Associated With Antibody Response in Feral Swine ( Sus scrofa) Infected With Brucella suis. Front Vet Sci 2020; 7:554674. [PMID: 33324693 PMCID: PMC7724110 DOI: 10.3389/fvets.2020.554674] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 10/23/2020] [Indexed: 11/13/2022] Open
Abstract
Feral swine (Sus scrofa) are a destructive invasive species widespread throughout the United States that disrupt ecosystems, damage crops, and carry pathogens of concern for the health of domestic stock and humans including Brucella suis-the causative organism for swine brucellosis. In domestic swine, brucellosis results in reproductive failure due to abortions and infertility. Contact with infected feral swine poses spillover risks to domestic pigs as well as humans, companion animals, wildlife, and other livestock. Genetic factors influence the outcome of infectious diseases; therefore, genome wide association studies (GWAS) of differential immune responses among feral swine can provide an understanding of disease dynamics and inform management to prevent the spillover of brucellosis from feral swine to domestic pigs. We sought to identify loci associated with differential antibody responses among feral swine naturally infected with B. suis using a case-control GWAS. Tissue, serum, and genotype data (68,516 bi-allelic single nucleotide polymorphisms) collected from 47 feral swine were analyzed in this study. The 47 feral swine were culture positive for Brucella spp. Of these 47, 16 were antibody positive (cases) whereas 31 were antibody negative (controls). Single-locus GWAS were performed using efficient mixed-model association eXpedited (EMMAX) methodology with three genetic models: additive, dominant, and recessive. Eight loci associated with seroconversion were identified on chromosome 4, 8, 9, 10, 12, and 18. Subsequent bioinformatic analyses revealed nine putative candidate genes related to immune function, most notably phagocytosis and induction of an inflammatory response. Identified loci and putative candidate genes may play an important role in host immune responses to B. suis infection, characterized by a detectable bacterial presence yet a differential antibody response. Given that antibody tests are used to evaluate brucellosis infection in domestic pigs and for disease surveillance in invasive feral swine, additional studies are needed to fully understand the genetic component of the response to B. suis infection and to more effectively translate estimates of Brucella spp. antibody prevalence among feral swine to disease control management action.
Collapse
Affiliation(s)
- Courtney F. Pierce
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, National Wildlife Research Center, Fort Collins, CO, United States
- Department of Animal Sciences, Colorado State University, Fort Collins, CO, United States
| | - Vienna R. Brown
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, National Feral Swine Damage Management Program, Fort Collins, CO, United States
| | - Steven C. Olsen
- United States Department of Agriculture, Agricultural Research Service, Infectious Bacterial Diseases, National Animal Disease Center, Ames, IA, United States
| | - Paola Boggiatto
- United States Department of Agriculture, Agricultural Research Service, Infectious Bacterial Diseases, National Animal Disease Center, Ames, IA, United States
| | - Kerri Pedersen
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, Raleigh, NC, United States
| | - Ryan S. Miller
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, Center for Epidemiology and Animal Health, Fort Collins, CO, United States
| | - Scott E. Speidel
- Department of Animal Sciences, Colorado State University, Fort Collins, CO, United States
| | - Timothy J. Smyser
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, National Wildlife Research Center, Fort Collins, CO, United States
| |
Collapse
|
23
|
Tetraspanins: useful multifunction proteins for the possible design and development of small-molecule therapeutic tools. Drug Discov Today 2020; 26:56-68. [PMID: 33137483 DOI: 10.1016/j.drudis.2020.10.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/21/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023]
Abstract
Tetraspanins constitute a well-conserved superfamily of four-span small membrane proteins (TM4SF), with >30 members in humans, with important roles in numerous mechanisms of cell biology. Moreover, tetraspanins associate with either specific partner proteins or another tetraspanin, generating a network of interactions involved in cell and membrane compartmentalization and having a role in cellular development, proliferation, activation, motility, and membrane fusions. Therefore, tetraspanins are considered regulators of cellular signaling and are often depicted as 'molecular facilitators'. In view of these many physiological functions, it is likely that these molecules are important actors in pathological processes. In this review, we present the main characteristics of this superfamily, providing a more detailed description of some significant representatives and discuss their relevance as potential targets for the design and development of small-molecule therapeutics in different pathologies.
Collapse
|
24
|
Tetraspanins, More than Markers of Extracellular Vesicles in Reproduction. Int J Mol Sci 2020; 21:ijms21207568. [PMID: 33066349 PMCID: PMC7589920 DOI: 10.3390/ijms21207568] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/08/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023] Open
Abstract
The participation of extracellular vesicles in many cellular processes, including reproduction, is unquestionable. Although currently, the tetraspanin proteins found in extracellular vesicles are mostly applied as markers, increasing evidence points to their role in extracellular vesicle biogenesis, cargo selection, cell targeting, and cell uptake under both physiological and pathological conditions. In this review, we bring other insight into the involvement of tetraspanin proteins in extracellular vesicle physiology in mammalian reproduction. We provide knowledge regarding the involvement of extracellular vesicle tetraspanins in these processes in somatic cells. Furthermore, we discuss the future direction towards an understanding of their functions in the tissues and fluids of the mammalian reproductive system in gamete maturation, fertilization, and embryo development; their involvement in mutual cell contact and communication in their complexity.
Collapse
|
25
|
Akella M, Malla R. Molecular modeling and in vitro study on pyrocatechol as potential pharmacophore of CD151 inhibitor. J Mol Graph Model 2020; 100:107681. [PMID: 32738620 DOI: 10.1016/j.jmgm.2020.107681] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/25/2020] [Accepted: 06/24/2020] [Indexed: 11/24/2022]
Abstract
CD151 has been recognized as a prognostic marker, the therapeutic target of breast cancers, but less explored for small molecule inhibitors due to lack of a validated model. The 3-D structure of CD151 large extracellular loop (LEL) was modeled using the LOMETS server and validated by the Ramachandran plot. The validated structure was employed for molecular docking and structure-based pharmacophore analysis. Druglikeness was evaluated by the ADMET description protocol. Antiproliferative activity was evaluated by MTT, BrdU incorporation, flow cytometry, and cell death ELISAPLUS assay. This study predicted the best model for CD151-LEL with 94.1% residues in favored regions and Z score -2.79 kcal/mol using the threading method. The web-based receptor cavity method identified one functional target site, which was suitable for the binding of aromatic and heterocyclic compounds. Molecular docking study identified pyrocatechol (PCL) and 5-fluorouracil (FU) as potential leads of CD151-LEL. The pharmacophore model identified interaction points of modeled CD151-LEL with PCL and FU. Also, the analysis of ADMET properties revealed the drug-likeness of PCL and FU. The viability of MDA-MB 231 cells was significantly reduced with PCL and FU but less affected MCF-12A, normal healthy breast epithelial cell line. With 50% toxic concentration, both PCL and FU significantly inhibited 82.46 and 87.12% proliferation, respectively, of MDA-MB 231 cells by altering morphology and inducing G1 cell cycle arrest and apoptosis. In addition, PCL and FU inhibited the CD151 expression by 4.5-and 4.8-folds, respectively. This study suggests the further assessment of pyrocatechol as a potential lead of CD151 in breast cancer at the molecular level.
Collapse
Affiliation(s)
- Manasa Akella
- Cancer Biology Lab, Dept. of Biochemistry and Bioinformatics, Institute of Science, GITAM (Deemed to Be University), Visakhapatnam, 530045, Andhra Pradesh, India
| | - RamaRao Malla
- Cancer Biology Lab, Dept. of Biochemistry and Bioinformatics, Institute of Science, GITAM (Deemed to Be University), Visakhapatnam, 530045, Andhra Pradesh, India.
| |
Collapse
|
26
|
Dunlock VE. Tetraspanin CD53: an overlooked regulator of immune cell function. Med Microbiol Immunol 2020; 209:545-552. [PMID: 32440787 PMCID: PMC7395052 DOI: 10.1007/s00430-020-00677-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/02/2020] [Indexed: 11/25/2022]
Abstract
Tetraspanins are membrane organizing proteins that play a role in organizing the cell surface through the formation of subcellular domains consisting of tetraspanins and their partner proteins. These complexes are referred to as tetraspanin enriched microdomains (TEMs) or the tetraspanin web. The formation of TEMs allows for the regulation of a variety of cellular processes such as adhesion, migration, signaling, and cell fusion. Tetraspanin CD53 is a member of the tetraspanin superfamily expressed exclusively within the immune compartment. Amongst others, B cells, CD4+ T cells, CD8+ T cells, dendritic cells, macrophages, and natural killer cells have all been found to express high levels of this protein on their surface. Almost three decades ago it was reported that patients who lacked CD53 suffered from an increased susceptibility to pathogens resulting in the clinical manifestation of recurrent viral, bacterial, and fungal infections. This clearly suggests a vital and non-redundant role for CD53 in immune function. Yet, despite this striking finding, the specific functional roles of CD53 within the immune system have remained elusive. This review aims to provide a concise overview of the published literature concerning CD53 and reflect on the underappreciated role of this protein in immune cell regulation and function.
Collapse
Affiliation(s)
- V E Dunlock
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
27
|
Zhang Y, Qian H, Xu A, Yang G. Increased expression of CD81 is associated with poor prognosis of prostate cancer and increases the progression of prostate cancer cells in vitro. Exp Ther Med 2019; 19:755-761. [PMID: 31885712 DOI: 10.3892/etm.2019.8244] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 10/30/2019] [Indexed: 12/27/2022] Open
Abstract
CD81, a member of the tetraspanin family, has been revealed to be upregulated and associated with prognosis in several types of cancer; however, this relationship has not been explored in prostate cancer. The present study aimed to investigate the prognostic significance and functional role of CD81 in prostate cancer. The expression of CD81 in prostate cancer tissues and cell lines was evaluated using qRT-PCR analysis. Kaplan-Meier survival analysis and Cox regression analysis were conducted to explore the prognostic significance of CD81. Cell experiments were used to explore the effects of CD81 on cell proliferation, migration, and invasion in prostate cell lines in vitro. The expression of CD81 was increased in both prostate cancer tissues and cell lines. Upregulation of CD81 was significantly associated with lymph node metastasis and TNM stage. Moreover, patients with high CD81 levels had poorer overall survival than those with lower levels. Additionally, tumor cell proliferation, migration, and invasion were inhibited by knockdown of CD81. The present results indicated that CD81 plays an oncogenic role in prostate cancer. Overexpression of CD81 may serve as a prognostic biomarker and therapeutic target and is involved in the progression of prostate cancer.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Urology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Haining Qian
- Department of Urology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - An Xu
- Department of Urology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Ganggang Yang
- Department of Urology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| |
Collapse
|
28
|
Li P, Dong M, Wang Z. Downregulation of TSPAN13 by miR-369-3p inhibits cell proliferation in papillary thyroid cancer (PTC). Bosn J Basic Med Sci 2019; 19:146-154. [PMID: 30114378 DOI: 10.17305/bjbms.2018.2865] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 01/30/2018] [Indexed: 01/07/2023] Open
Abstract
Previous studies demonstrated dysregulation of different microRNAs in thyroid cancer. Tetraspanins (TSPANs) are cell surface proteins with critical roles in many cellular processes, and implications in tumor development. Here we investigated the role of miR-369-3p in papillary thyroid cancer (PTC) and its association with TSPAN13. miR-369-3p and the TSPAN13 gene expression profiles of 513 thyroid cancer and 59 normal thyroid tissues were downloaded from the Cancer Genome Atlas database. Thyroid cancer tissues were classified according to the histological type, grouped based on low and high median miR-369-3p and TSPAN13 expression, and analyzed in relation to overall survival (OS) of patients. Human PTC cell lines (TPC-1 and GLAG-66) and human embryonic kidney 293T (HEK293T) cells were used for in vitro analysis. Transfection experiments were performed with synthetic miRNA mimics for miR-369-3p and small interfering RNAs for TSPAN13. Relative expression of miR-369-3p and TSPAN13 mRNA was determined by RT-qPCR. Protein levels of TSPAN13 were determined by western blotting. Cell proliferation (CCK-8 assay), colony formation, and apoptosis (flow cytometry) were analyzed in transfected cells. Binding sites of miR-369-3p in TSPAN13 mRNA were determined by bioinformatics analysis and dual luciferase reporter assay. miR-369-3p was downregulated and TSPAN13 upregulated in PTC, follicular thyroid cancer, and tall cell variant tissues. Both low expression of miR-369-3p and high expression of TSPAN13 were associated with shorter OS in thyroid cancer patients. Overexpression of miR-369-3p significantly suppressed proliferation and promoted apoptosis in PTC cells. TSPAN13 was a direct target of miR-369-3p, and silencing of TSPAN13 phenocopied the effect of miR-369-3p mimics in PTC cells. Overall, the downregulation of miR-369-3p and consequent upregulation of its target TSPAN13 appear to be involved in pathophysiology of PTC.
Collapse
Affiliation(s)
- Peng Li
- Department of Emergency, Jingmen NO.1 People's Hospital, Jingmen, Hubei, China.
| | | | | |
Collapse
|
29
|
Zhao W, Tao XL, Liu N, Lu HZ, Zheng CH, Li XY, Yu M, Li YG. Host protein CD63 promotes viral RNA replication by interacting with human astrovirus non-structural protein nsP1a/4. J Gen Virol 2019; 100:616-628. [DOI: 10.1099/jgv.0.001231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Wei Zhao
- 1College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou 121200, Liaoning, PR China
- 2Biological Anthropology institute, Jinzhou Medical University, Jinzhou 121200, Liaoning, PR China
| | - Xiao Li Tao
- 1College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou 121200, Liaoning, PR China
| | - Nian Liu
- 1College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou 121200, Liaoning, PR China
- 2Biological Anthropology institute, Jinzhou Medical University, Jinzhou 121200, Liaoning, PR China
| | - Heng Zhang Lu
- 1College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou 121200, Liaoning, PR China
| | - Chun Hong Zheng
- 1College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou 121200, Liaoning, PR China
| | - Xiang Yu Li
- 1College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou 121200, Liaoning, PR China
| | - Man Yu
- 1College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou 121200, Liaoning, PR China
| | - Yong Gang Li
- 1College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou 121200, Liaoning, PR China
| |
Collapse
|
30
|
Biogenesis of Extracellular Vesicles during Herpes Simplex Virus 1 Infection: Role of the CD63 Tetraspanin. J Virol 2019; 93:JVI.01850-18. [PMID: 30355691 DOI: 10.1128/jvi.01850-18] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 10/16/2018] [Indexed: 12/20/2022] Open
Abstract
Herpes simplex virus 1 (HSV-1) infections afflict more than 80% of the population worldwide. The virus primarily infects mucoepithelial cells and establishes latent reservoirs in neurons in sensory ganglia. Frequent reactivation has been linked to severe diseases, especially in immunocompromised individuals. Earlier, we reported that viral and host factors are packaged in extracellular vesicles (EVs) and delivered to uninfected cells, where they activate antiviral responses and restrict virus infection. Here, we interrogated the effect of HSV-1 infection on EV biogenesis. We found that HSV-1 infection causes a decrease in the amount of intracellular CD63 protein with a concomitant increase in extracellular CD63. This observation correlates with our previous finding that infected cells release more CD63-positive EVs than uninfected cells. The stimulation of CD63 exocytosis requires virus replication. CD63 is a member of the tetraspanin family of proteins that traffics between the plasma membrane and endosomal compartments and has a role in sorting cargo into the EVs. Previously, we reported that in cells depleted of CD63, HSV-1 virus yields increased, and here we provide data showing that in cells overexpressing CD63, HSV-1 virus yields decreased. Taken together, our data indicate that CD63 negatively impacts HSV-1 infection and that the CD63-positive EVs could control the dissemination of the virus in the host. Perhaps EV release by HSV-1-infected cells is a mechanism that controls virus dissemination.IMPORTANCE Intercellular communication, especially in neurons, largely relies on EVs, and modulation of EVs is known to impact physiological processes. Here, we present evidence that HSV-1 infection causes major alterations in the biogenesis of EVs, including an increase in their number and an increase in the CD63-positive population of EVs. These alterations result in an enrichment of the milieu of infection with EVs carrying signatures from infected cells. In addition to changes in the origin and type, EVs released by infected cells have differences in cargo, as they carry viral and host factors determined by the virus. The tetraspanin CD63 negatively impacts the infection, as demonstrated by CD63-knockdown and overexpression assays. A proposed mechanism involves the activation of antiviral responses in cells receiving CD63-positive EVs released by infected cells. Overall, HSV-1 causes major alterations in EVs that could contribute to HSV-1 persistence and pathogenesis.
Collapse
|
31
|
Mu W, Wang Z, Zöller M. Ping-Pong-Tumor and Host in Pancreatic Cancer Progression. Front Oncol 2019; 9:1359. [PMID: 31921628 PMCID: PMC6927459 DOI: 10.3389/fonc.2019.01359] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022] Open
Abstract
Metastasis is the main cause of high pancreatic cancer (PaCa) mortality and trials dampening PaCa mortality rates are not satisfying. Tumor progression is driven by the crosstalk between tumor cells, predominantly cancer-initiating cells (CIC), and surrounding cells and tissues as well as distant organs, where tumor-derived extracellular vesicles (TEX) are of major importance. A strong stroma reaction, recruitment of immunosuppressive leukocytes, perineural invasion, and early spread toward the peritoneal cavity, liver, and lung are shared with several epithelial cell-derived cancer, but are most prominent in PaCa. Here, we report on the state of knowledge on the PaCIC markers Tspan8, alpha6beta4, CD44v6, CXCR4, LRP5/6, LRG5, claudin7, EpCAM, and CD133, which all, but at different steps, are engaged in the metastatic cascade, frequently via PaCIC-TEX. This includes the contribution of PaCIC markers to TEX biogenesis, targeting, and uptake. We then discuss PaCa-selective features, where feedback loops between stromal elements and tumor cells, including distorted transcription, signal transduction, and metabolic shifts, establish vicious circles. For the latter particularly pancreatic stellate cells (PSC) are responsible, furnishing PaCa to cope with poor angiogenesis-promoted hypoxia by metabolic shifts and direct nutrient transfer via vesicles. Furthermore, nerves including Schwann cells deliver a large range of tumor cell attracting factors and Schwann cells additionally support PaCa cell survival by signaling receptor binding. PSC, tumor-associated macrophages, and components of the dysplastic stroma contribute to perineural invasion with signaling pathway activation including the cholinergic system. Last, PaCa aggressiveness is strongly assisted by the immune system. Although rich in immune cells, only immunosuppressive cells and factors are recovered in proximity to tumor cells and hamper effector immune cells entering the tumor stroma. Besides a paucity of immunostimulatory factors and receptors, immunosuppressive cytokines, myeloid-derived suppressor cells, regulatory T-cells, and M2 macrophages as well as PSC actively inhibit effector cell activation. This accounts for NK cells of the non-adaptive and cytotoxic T-cells of the adaptive immune system. We anticipate further deciphering the molecular background of these recently unraveled intermingled phenomena may turn most lethal PaCa into a curatively treatable disease.
Collapse
Affiliation(s)
- Wei Mu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Wei Mu
| | - Zhe Wang
- Department of Oncology, The First Affiliated Hospital of Guangdong, Pharmaceutical University, Guangzhou, China
| | - Margot Zöller
- Department of Oncology, The First Affiliated Hospital of Guangdong, Pharmaceutical University, Guangzhou, China
| |
Collapse
|
32
|
Zhao K, Wang Z, Hackert T, Pitzer C, Zöller M. Tspan8 and Tspan8/CD151 knockout mice unravel the contribution of tumor and host exosomes to tumor progression. J Exp Clin Cancer Res 2018; 37:312. [PMID: 30541597 PMCID: PMC6292129 DOI: 10.1186/s13046-018-0961-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/14/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The tetraspanins Tspan8 and CD151 promote metastasis, exosomes (Exo) being suggested to be important in the crosstalk between tumor and host. The contribution of Tspan8 and CD151 to host versus tumor-derived exosome (TEX) activities being not defined, we approached the questions using 3-methylcholanthrene-induced (MCA) tumors from wt, Tspan8ko, CD151ko and Tspan8/CD151 (db)ko mice, implanted into tetraspanin-competent and deficient hosts. METHODS Tumor growth and dissemination, hematopoiesis and angiogenesis were surveyed in wild type (wt), Tspan8ko, CD151ko and dbko mice bearing tetraspanin-competent and -deficient MCA tumors. In vitro studies using tumor cells, bone marrow cells (BMC) and endothelial cells (EC) elaborated the mechanism of serum (s)Exo- and TEX-induced target modulation. RESULTS Tumors grew in autochthonous and syngeneic hosts differing in Tspan8- and/or CD151-competence. However, Tspan8ko- and/or CD151ko-tumor cell dissemination and settlement in metastatic organs was significantly reduced in the autochthonous host, and less severely in the wt-host. Impaired wt-MCA tumor dissemination in the ko-host confirmed a contribution of host- and tumor-Tspan8/-CD151 to tumor cell dissemination, delivery of sExo and TEX being severely impaired by a Tspan8ko/CD151ko. Coculturing tumor cells, BMC and EC with sExo and TEX revealed minor defects in epithelial mesenchymal transition and apoptosis resistance of ko tumors. Strongly reduced migratory and invasive capacity of Tspan8ko/CD151ko-MCA relies on distorted associations with integrins and CAM and missing Tspan8/CD151-promoted recruitment of proteases. The defects, differing between Tspan8ko- and CD151ko-MCA, were rescued by wt-TEX and, less efficiently Tspan8ko- and CD151ko-TEX. Minor defects in hematopoietic progenitor maturation were based on the missing association of hematopoietic growth factors /- receptors with CD151 and, less pronounced, Tspan8. Rescue of impaired angiogenesis in ko mice by wt-sExo and promotion of angiogenesis by TEX depended on the association of Tspan8 and CD151 with GPCR and RTK in EC and tumor cells. CONCLUSIONS Tspan8-/CD151-TEX play central roles in tumor progression. Tspan8-/CD151-sExo and TEX contribute by stimulating angiogenesis. Tspan8 and CD151 fulfill these tasks by associating with function-relevant proteins, the additive impact of Tspan8 and CD151 relying on differences in preferred associations. The distinct Tspan8 and CD151 contributions suggest a blockade of TEX-Tspan8 and -CD151 promising for therapeutic intervention.
Collapse
Affiliation(s)
- Kun Zhao
- Pancreas Section, University Hospital of Surgery, Ruprecht-Karls-University, Heidelberg, Germany
| | - Zhe Wang
- Pancreas Section, University Hospital of Surgery, Ruprecht-Karls-University, Heidelberg, Germany
- Present Address: Department of Oncology, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong, China
| | - Thilo Hackert
- Pancreas Section, University Hospital of Surgery, Ruprecht-Karls-University, Heidelberg, Germany
| | - Claudia Pitzer
- Interdisciplinary Neurobehavioral Core, Institute of Pharmacology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Margot Zöller
- Pancreas Section, University Hospital of Surgery, Ruprecht-Karls-University, Heidelberg, Germany
| |
Collapse
|
33
|
Machado-Pineda Y, Cardeñes B, Reyes R, López-Martín S, Toribio V, Sánchez-Organero P, Suarez H, Grötzinger J, Lorenzen I, Yáñez-Mó M, Cabañas C. CD9 Controls Integrin α5β1-Mediated Cell Adhesion by Modulating Its Association With the Metalloproteinase ADAM17. Front Immunol 2018; 9:2474. [PMID: 30455686 PMCID: PMC6230984 DOI: 10.3389/fimmu.2018.02474] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 10/08/2018] [Indexed: 12/21/2022] Open
Abstract
Integrin α5β1 is a crucial adhesion molecule that mediates the adherence of many cell types to the extracellular matrix through recognition of its classic ligand fibronectin as well as to other cells through binding to an alternative counter-receptor, the metalloproteinase ADAM17/TACE. Interactions between integrin α5β1 and ADAM17 may take place both in trans (between molecules expressed on different cells) or in cis (between molecules expressed on the same cell) configurations. It has been recently reported that the cis association between α5β1 and ADAM17 keeps both molecules inactive, whereas their dissociation results in activation of their adhesive and metalloproteinase activities. Here we show that the tetraspanin CD9 negatively regulates integrin α5β1-mediated cell adhesion by enhancing the cis interaction of this integrin with ADAM17 on the cell surface. Additionally we show that, similarly to CD9, the monoclonal antibody 2A10 directed to the disintegrin domain of ADAM17 specifically inhibits integrin α5β1-mediated cell adhesion to its ligands fibronectin and ADAM17.
Collapse
Affiliation(s)
- Yesenia Machado-Pineda
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Beatriz Cardeñes
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Raquel Reyes
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Soraya López-Martín
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.,Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| | - Víctor Toribio
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.,Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| | - Paula Sánchez-Organero
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Henar Suarez
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.,Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| | - Joachim Grötzinger
- Institute of Biochemistry, Christian-Albrechts University, Kiel, Germany
| | - Inken Lorenzen
- Department of Structural Biology, Institute of Zoology, Kiel, Germany
| | - María Yáñez-Mó
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.,Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Biología Molecular Severo Ochoa, Instituto de Investigación Sanitaria la Princesa (IIS-IP), Madrid, Spain
| | - Carlos Cabañas
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.,Departamento de Inmunología, Oftalmología y OTR, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| |
Collapse
|
34
|
The peptide mimicking small extracellular loop domain of CD82 inhibits tumor cell migration, adhesion and induces apoptosis by inhibiting integrin mediated signaling. Biochem Biophys Res Commun 2018; 503:2206-2211. [DOI: 10.1016/j.bbrc.2018.06.139] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 06/25/2018] [Indexed: 12/21/2022]
|
35
|
Murru L, Moretto E, Martano G, Passafaro M. Tetraspanins shape the synapse. Mol Cell Neurosci 2018; 91:76-81. [DOI: 10.1016/j.mcn.2018.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/29/2018] [Accepted: 04/01/2018] [Indexed: 01/01/2023] Open
|
36
|
Zhang N, Zuo L, Zheng H, Li G, Hu X. Increased Expression of CD81 in Breast Cancer Tissue is Associated with Reduced Patient Prognosis and Increased Cell Migration and Proliferation in MDA-MB-231 and MDA-MB-435S Human Breast Cancer Cell Lines In Vitro. Med Sci Monit 2018; 24:5739-5747. [PMID: 30117494 PMCID: PMC6109364 DOI: 10.12659/msm.911612] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Background CD81, a member of the tetraspanin family, is overexpressed in several tumor types, but its role in breast cancer remains unknown. The present study aimed to investigate the effects of increased CD81 expression on cell migration and proliferation in breast cancer cell lines, MDA-MB-231 and MDA-MB-435S in vitro, and the effects of increased CD81 expression in breast cancer tissue microarrays on patient prognosis. Material/Methods The expression of CD81 was evaluated using immunohistochemistry in a human breast cancer tissue microarray containing 140 tumor tissues and a microarray containing 77 normal breast tissues. The effects of increased CD81 expression on cell proliferation and migration in breast cancer cell lines, MDA-MB-231 and MDA-MB-435S, were evaluated by proliferation, transwell migration, and cell counting kit-8 (CCK-8) assays. CD81-expressing plasmid transfection upregulated CD81 expression, and short hairpin RNA (shRNA) lentivirus silenced CD81 expression in vitro. Results CD81 expression was significantly increased in breast cancer tissues compared with normal breast tissues (P<0.05). Increased expression of CD81 was significantly associated with lymph node metastasis (P<0.05), clinical stage (P<0.05) and with reduced overall survival (OS) in patients with breast cancer (P<0.05). Increased CD81 expression in MDA-MB-231 and MDA-MB-435S cells promoted cell proliferation and migration, which were inhibited by CD81 silencing. Conclusions The findings of the study showed that CD81 might be a potential prognostic biomarker associated with poor patient prognosis in breast cancer. These findings should be investigated further with large-scale controlled prospective studies in patients with breast cancer.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China (mainland).,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (mainland).,Department of Oncology, Fudan University Shanghai Cancer Center Minhang Branch, Shanghai, China (mainland)
| | - Li Zuo
- Department of Oncology, Fudan University Shanghai Cancer Center Minhang Branch, Shanghai, China (mainland)
| | - Hong Zheng
- Department of Oncology, Fudan University Shanghai Cancer Center Minhang Branch, Shanghai, China (mainland)
| | - Gang Li
- Department of Oncology, Fudan University Shanghai Cancer Center Minhang Branch, Shanghai, China (mainland)
| | - Xichun Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China (mainland).,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (mainland).,Department of Oncology, Fudan University Shanghai Cancer Center Minhang Branch, Shanghai, China (mainland)
| |
Collapse
|
37
|
Murru L, Vezzoli E, Longatti A, Ponzoni L, Falqui A, Folci A, Moretto E, Bianchi V, Braida D, Sala M, D'Adamo P, Bassani S, Francolini M, Passafaro M. Pharmacological Modulation of AMPAR Rescues Intellectual Disability-Like Phenotype in Tm4sf2-/y Mice. Cereb Cortex 2018; 27:5369-5384. [PMID: 28968657 PMCID: PMC5939231 DOI: 10.1093/cercor/bhx221] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 08/02/2017] [Indexed: 01/02/2023] Open
Abstract
Intellectual disability affects 2–3% of the world's population and typically begins during childhood, causing impairments in social skills and cognitive abilities. Mutations in the TM4SF2 gene, which encodes the TSPAN7 protein, cause a severe form of intellectual disability, and currently, no therapy is able to ameliorate this cognitive impairment. We previously reported that, in cultured neurons, shRNA-mediated down-regulation of TSPAN7 affects AMPAR trafficking by enhancing PICK1–GluA2 interaction, thereby increasing the intracellular retention of AMPAR. Here, we found that loss of TSPAN7 function in mice causes alterations in hippocampal excitatory synapse structure and functionality as well as cognitive impairment. These changes occurred along with alterations in AMPAR expression levels. We also found that interfering with PICK1–GluA2 binding restored synaptic function in Tm4sf2−/y mice. Moreover, potentiation of AMPAR activity via the administration of the ampakine CX516 reverted the neurological phenotype observed in Tm4sf2−/y mice, suggesting that pharmacological modulation of AMPAR may represent a new approach for treating patients affected by TM4SF2 mutations and intellectual disability.
Collapse
Affiliation(s)
- Luca Murru
- CNR Institute of Neuroscience, 20129 Milano, Italy
| | - Elena Vezzoli
- Department of Medical Biotechnology and Translational Medicine, Università degli studi di Milano, Via Vanvitelli 32, 20129 Milano, Italy.,Department of Pharmacological and Biomolecular Sciences (DiSFeB), Università di Milano, Via Balzaretti 9, 20133 Milano, Italy.,Department of Biosciences and Centre for Stem Cell Research, University of Milan and Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi" Milan, Italy
| | - Anna Longatti
- CNR Institute of Neuroscience, 20129 Milano, Italy.,Department of Pharmacological and Biomolecular Sciences (DiSFeB), Università di Milano, Via Balzaretti 9, 20133 Milano, Italy
| | - Luisa Ponzoni
- Department of Medical Biotechnology and Translational Medicine, Università degli studi di Milano, Via Vanvitelli 32, 20129 Milano, Italy.,Fondazione Umberto Veronesi, Piazza Velasca 5, 20122 Milan, Italy
| | - Andrea Falqui
- Biological and Environmental Sciences and Engineering Division, King Abdullah University for Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| | | | | | - Veronica Bianchi
- Division of Neuroscience, IRCSS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Daniela Braida
- Department of Medical Biotechnology and Translational Medicine, Università degli studi di Milano, Via Vanvitelli 32, 20129 Milano,Italy
| | | | - Patrizia D'Adamo
- Division of Neuroscience, IRCSS San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | - Maura Francolini
- Department of Medical Biotechnology and Translational Medicine, Università degli studi di Milano, Via Vanvitelli 32, 20129 Milano,Italy
| | | |
Collapse
|
38
|
Florin L, Lang T. Tetraspanin Assemblies in Virus Infection. Front Immunol 2018; 9:1140. [PMID: 29887866 PMCID: PMC5981178 DOI: 10.3389/fimmu.2018.01140] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 05/07/2018] [Indexed: 12/23/2022] Open
Abstract
Tetraspanins (Tspans) are a family of four-span transmembrane proteins, known as plasma membrane “master organizers.” They form Tspan-enriched microdomains (TEMs or TERMs) through lateral association with one another and other membrane proteins. If multiple microdomains associate with each other, larger platforms can form. For infection, viruses interact with multiple cell surface components, including receptors, activating proteases, and signaling molecules. It appears that Tspans, such as CD151, CD82, CD81, CD63, CD9, Tspan9, and Tspan7, coordinate these associations by concentrating the interacting partners into Tspan platforms. In addition to mediating viral attachment and entry, these platforms may also be involved in intracellular trafficking of internalized viruses and assist in defining virus assembly and exit sites. In conclusion, Tspans play a role in viral infection at different stages of the virus replication cycle. The present review highlights recently published data on this topic, with a focus on events at the plasma membrane. In light of these findings, we propose a model for how Tspan interactions may organize cofactors for viral infection into distinct molecular platforms.
Collapse
Affiliation(s)
- Luise Florin
- Department of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Thorsten Lang
- Department of Membrane Biochemistry, Life & Medical Sciences Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
39
|
Willms E, Cabañas C, Mäger I, Wood MJA, Vader P. Extracellular Vesicle Heterogeneity: Subpopulations, Isolation Techniques, and Diverse Functions in Cancer Progression. Front Immunol 2018; 9:738. [PMID: 29760691 PMCID: PMC5936763 DOI: 10.3389/fimmu.2018.00738] [Citation(s) in RCA: 651] [Impact Index Per Article: 93.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 03/26/2018] [Indexed: 12/14/2022] Open
Abstract
Cells release membrane enclosed nano-sized vesicles termed extracellular vesicles (EVs) that function as mediators of intercellular communication by transferring biological information between cells. Tumor-derived EVs have emerged as important mediators in cancer development and progression, mainly through transfer of their bioactive content which can include oncoproteins, oncogenes, chemokine receptors, as well as soluble factors, transcripts of proteins and miRNAs involved in angiogenesis or inflammation. This transfer has been shown to influence the metastatic behavior of primary tumors. Moreover, tumor-derived EVs have been shown to influence distant cellular niches, establishing favorable microenvironments that support growth of disseminated cancer cells upon their arrival at these pre-metastatic niches. It is generally accepted that cells release a number of major EV populations with distinct biophysical properties and biological functions. Exosomes, microvesicles, and apoptotic bodies are EV populations most widely studied and characterized. They are discriminated based primarily on their intracellular origin. However, increasing evidence suggests that even within these EV populations various subpopulations may exist. This heterogeneity introduces an extra level of complexity in the study of EV biology and function. For example, EV subpopulations could have unique roles in the intricate biological processes underlying cancer biology. Here, we discuss current knowledge regarding the role of subpopulations of EVs in cancer development and progression and highlight the relevance of EV heterogeneity. The position of tetraspanins and integrins therein will be highlighted. Since addressing EV heterogeneity has become essential for the EV field, current and novel techniques for isolating EV subpopulations will also be discussed. Further dissection of EV heterogeneity will advance our understanding of the critical roles of EVs in health and disease.
Collapse
Affiliation(s)
- Eduard Willms
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Carlos Cabañas
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.,Department of Microbiology I (Immunology), Faculty of Medicine, Universidad Complutense, Madrid, Spain
| | - Imre Mäger
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.,Institute of Technology, University of Tartu, Tartu, Estonia
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Pieter Vader
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.,Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, Netherlands.,Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
40
|
Frolikova M, Manaskova-Postlerova P, Cerny J, Jankovicova J, Simonik O, Pohlova A, Secova P, Antalikova J, Dvorakova-Hortova K. CD9 and CD81 Interactions and Their Structural Modelling in Sperm Prior to Fertilization. Int J Mol Sci 2018; 19:ijms19041236. [PMID: 29671763 PMCID: PMC5979608 DOI: 10.3390/ijms19041236] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/10/2018] [Accepted: 04/13/2018] [Indexed: 11/16/2022] Open
Abstract
Proteins CD9 and CD81 are members of the tetraspanin superfamily and were detected in mammalian sperm, where they are suspected to form an active tetraspanin web and to participate in sperm–egg membrane fusion. The importance of these two proteins during the early stages of fertilization is supported by the complete sterility of CD9/CD81 double null female mice. In this study, the putative mechanism of CD9/CD81 involvement in tetraspanin web formation in sperm and its activity prior to fertilization was addressed. Confocal microscopy and colocalization assay was used to determine a mutual CD9/CD81 localization visualised in detail by super-resolution microscopy, and their interaction was address by co-immunoprecipitation. The species-specific traits in CD9 and CD81 distribution during sperm maturation were compared between mice and humans. A mutual position of CD9/CD81 is shown in human spermatozoa in the acrosomal cap, however in mice, CD9 and CD81 occupy a distinct area. During the acrosome reaction in human sperm, only CD9 is relocated, compared to the relocation of both proteins in mice. The structural modelling of CD9 and CD81 homologous and possibly heterologous network formation was used to propose their lateral Cis as well as Trans interactions within the sperm membrane and during sperm–egg membrane fusion.
Collapse
Affiliation(s)
- Michaela Frolikova
- Group of Reproductive Biology, Institute of Biotechnology, Czech Academy of Sciences, v.v.i., BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic.
| | - Pavla Manaskova-Postlerova
- Group of Reproductive Biology, Institute of Biotechnology, Czech Academy of Sciences, v.v.i., BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic.
- Department of Veterinary Sciences, Faculty of Agrobiology, Food and Natural Resources, University of Life Sciences Prague, Kamycka 129, 165 00 Prague, Czech Republic.
| | - Jiri Cerny
- Laboratory of Structural Bioinformatics of Proteins, Institute of Biotechnology Czech Academy of Sciences, v.v.i., BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic.
| | - Jana Jankovicova
- Laboratory of Reproductive Physiology, Institute of Animal Biochemistry and Genetics Centre of Biosciences Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia.
| | - Ondrej Simonik
- Group of Reproductive Biology, Institute of Biotechnology, Czech Academy of Sciences, v.v.i., BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic.
- Department of Veterinary Sciences, Faculty of Agrobiology, Food and Natural Resources, University of Life Sciences Prague, Kamycka 129, 165 00 Prague, Czech Republic.
| | - Alzbeta Pohlova
- Group of Reproductive Biology, Institute of Biotechnology, Czech Academy of Sciences, v.v.i., BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic.
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030/8, 128 43 Prague, Czech Republic.
| | - Petra Secova
- Laboratory of Reproductive Physiology, Institute of Animal Biochemistry and Genetics Centre of Biosciences Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia.
| | - Jana Antalikova
- Laboratory of Reproductive Physiology, Institute of Animal Biochemistry and Genetics Centre of Biosciences Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia.
| | - Katerina Dvorakova-Hortova
- Group of Reproductive Biology, Institute of Biotechnology, Czech Academy of Sciences, v.v.i., BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic.
- Department of Zoology, Faculty of Science, Charles University, Vinicna 7, 128 44 Prague, Czech Republic.
| |
Collapse
|
41
|
Böker KO, Lemus-Diaz N, Rinaldi Ferreira R, Schiller L, Schneider S, Gruber J. The Impact of the CD9 Tetraspanin on Lentivirus Infectivity and Exosome Secretion. Mol Ther 2017; 26:634-647. [PMID: 29221804 DOI: 10.1016/j.ymthe.2017.11.008] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 10/24/2017] [Accepted: 11/11/2017] [Indexed: 12/18/2022] Open
Abstract
Efficient transduction tools are a hallmark for both research and therapy development. Here, we introduce new insights into the generation of lentiviral vectors with improved performance by utilizing producer cells with increased production rates of extracellular vesicles through CD9 overexpression. Most human cells secrete small vesicles from their surface (microvesicles) or intraluminal endosome-derived membranes (exosomes). In particular, enhanced levels of the tetraspanin CD9 result in significantly increased numbers of extracellular vesicles with exosome-like features that were secreted from four different human cell lines. Intriguingly, exosomes and their biogenesis route display similarities to lentivirus and we examined the impact of CD9 expression on release and infectivity of recombinant lentiviral vectors. Although the titers of released viral particles were not increased upon production in high CD9 cells, we observed improved performance in terms of both speed and efficiency of lentiviral gene delivery into numerous human cell lines, including HEK293, HeLa, SH-SY5Y, as well as B and T lymphocytes. Here, we demonstrate that enhanced CD9 enables lentiviral transduction in the absence of any pseudotyping viral glycoprotein or fusogenic molecule. Our findings indicate an important role of CD9 for lentiviral vector and exosome biogenesis and point out a remarkable function of this tetraspanin in membrane fusion, viral infectivity, and exosome-mediated horizontal information transfer.
Collapse
Affiliation(s)
- Kai O Böker
- Junior Research Group Medical RNA Biology, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany; Department for Trauma Surgery, Orthopaedics and Plastic Surgery, University Medical Center, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Nicolas Lemus-Diaz
- Junior Research Group Medical RNA Biology, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Rafael Rinaldi Ferreira
- Junior Research Group Medical RNA Biology, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Lara Schiller
- Junior Research Group Medical RNA Biology, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Stefan Schneider
- Junior Research Group Medical RNA Biology, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Jens Gruber
- Junior Research Group Medical RNA Biology, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany.
| |
Collapse
|
42
|
Quantitative membrane proteomics reveals a role for tetraspanin enriched microdomains during entry of human cytomegalovirus. PLoS One 2017; 12:e0187899. [PMID: 29121670 PMCID: PMC5679760 DOI: 10.1371/journal.pone.0187899] [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: 03/02/2017] [Accepted: 10/27/2017] [Indexed: 12/15/2022] Open
Abstract
Human cytomegalovirus (HCMV) depends on and modulates multiple host cell membrane proteins during each stage of the viral life cycle. To gain a global view of the impact of HCMV-infection on membrane proteins, we analyzed HCMV-induced changes in the abundance of membrane proteins in fibroblasts using stable isotope labeling with amino acids (SILAC), membrane fractionation and protein identification by two-dimensional liquid chromatography and tandem mass spectrometry. This systematic approach revealed that CD81, CD44, CD98, caveolin-1 and catenin delta-1 were down-regulated during infection whereas GRP-78 was up-regulated. Since CD81 downregulation was also observed during infection with UV-inactivated virus we hypothesized that this tetraspanin is part of the viral entry process. Interestingly, additional members of the tetraspanin family, CD9 and CD151, were also downregulated during HCMV-entry. Since tetraspanin-enriched microdomains (TEM) cluster host cell membrane proteins including known CMV receptors such as integrins, we studied whether TEMs are required for viral entry. When TEMs were disrupted with the cholesterol chelator methyl-β-cylcodextrin, viral entry was inhibited and this inhibition correlated with reduced surface levels of CD81, CD9 and CD151, whereas integrin levels remained unchanged. Furthermore, simultaneous siRNA-mediated knockdown of multiple tetraspanins inhibited viral entry whereas individual knockdown had little effect suggesting essential, but redundant roles for individual tetraspanins during entry. Taken together, our data suggest that TEM act as platforms for receptors utilized by HCMV for entry into cells.
Collapse
|
43
|
Zuidscherwoude M, Worah K, van der Schaaf A, Buschow SI, van Spriel AB. Differential expression of tetraspanin superfamily members in dendritic cell subsets. PLoS One 2017; 12:e0184317. [PMID: 28880937 PMCID: PMC5589240 DOI: 10.1371/journal.pone.0184317] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 08/22/2017] [Indexed: 11/18/2022] Open
Abstract
Dendritic cells (DCs), which are essential for initiating immune responses, are comprised of different subsets. Tetraspanins organize dendritic cell membranes by facilitating protein-protein interactions within the so called tetraspanin web. In this study we analyzed expression of the complete tetraspanin superfamily in primary murine (CD4+, CD8+, pDC) and human DC subsets (CD1c+, CD141+, pDC) at the transcriptome and proteome level. Different RNA and protein expression profiles for the tetraspanin genes across human and murine DC subsets were identified. Although RNA expression levels of CD37 and CD82 were not significantly different between human DC subsets, CD9 RNA was highly expressed in pDCs, while CD9 protein expression was lower. This indicates that relative RNA and protein expression levels are not always in agreement. Both murine CD8α+ DCs and its regarded human counterpart, CD141+ DCs, displayed relatively high protein levels of CD81. CD53 protein was highly expressed on human pDCs in contrast to the relatively low protein expression of most other tetraspanins. This study demonstrates that tetraspanins are differentially expressed by human and murine DC subsets which provides a valuable resource that will aid the understanding of tetraspanin function in DC biology.
Collapse
Affiliation(s)
- Malou Zuidscherwoude
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kuntal Worah
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Centre for Molecular and Biomolecular Informatics (CMBI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Alie van der Schaaf
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sonja I. Buschow
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Annemiek B. van Spriel
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- * E-mail:
| |
Collapse
|
44
|
Bulfone-Paus S, Nilsson G, Draber P, Blank U, Levi-Schaffer F. Positive and Negative Signals in Mast Cell Activation. Trends Immunol 2017; 38:657-667. [DOI: 10.1016/j.it.2017.01.008] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/17/2017] [Accepted: 01/27/2017] [Indexed: 01/05/2023]
|
45
|
Munkley J, McClurg UL, Livermore KE, Ehrmann I, Knight B, Mccullagh P, Mcgrath J, Crundwell M, Harries LW, Leung HY, Mills IG, Robson CN, Rajan P, Elliott DJ. The cancer-associated cell migration protein TSPAN1 is under control of androgens and its upregulation increases prostate cancer cell migration. Sci Rep 2017; 7:5249. [PMID: 28701765 PMCID: PMC5507901 DOI: 10.1038/s41598-017-05489-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 05/30/2017] [Indexed: 02/06/2023] Open
Abstract
Cell migration drives cell invasion and metastatic progression in prostate cancer and is a major cause of mortality and morbidity. However the mechanisms driving cell migration in prostate cancer patients are not fully understood. We previously identified the cancer-associated cell migration protein Tetraspanin 1 (TSPAN1) as a clinically relevant androgen regulated target in prostate cancer. Here we find that TSPAN1 is acutely induced by androgens, and is significantly upregulated in prostate cancer relative to both normal prostate tissue and benign prostate hyperplasia (BPH). We also show for the first time, that TSPAN1 expression in prostate cancer cells controls the expression of key proteins involved in cell migration. Stable upregulation of TSPAN1 in both DU145 and PC3 cells significantly increased cell migration and induced the expression of the mesenchymal markers SLUG and ARF6. Our data suggest TSPAN1 is an androgen-driven contributor to cell survival and motility in prostate cancer.
Collapse
Affiliation(s)
- Jennifer Munkley
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, UK.
| | - Urszula L McClurg
- Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK
| | - Karen E Livermore
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, UK
| | - Ingrid Ehrmann
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, UK
| | - Bridget Knight
- NIHR Exeter Clinical Research Facility, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Paul Mccullagh
- Department of Pathology, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - John Mcgrath
- Exeter Surgical Health Services Research Unit, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Malcolm Crundwell
- Department of Urology, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Lorna W Harries
- Institute of Biomedical and Clinical Sciences, University of Exeter, Devon, UK
| | - Hing Y Leung
- Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Ian G Mills
- Prostate Cancer Research Group, Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo and Oslo University Hospitals, Forskningsparken, Gaustadalléen 21, N-0349, Oslo, Norway
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital HE - Norwegian Radium Hospital, Montebello, Ian G. Mills, NO-0424, Oslo, Norway
- Movember/Prostate Cancer UK Centre of Excellence for Prostate Cancer Research, Centre for Cancer Research and Cell Biology (CCRCB), Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Craig N Robson
- Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK
| | - Prabhakar Rajan
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - David J Elliott
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, UK
| |
Collapse
|
46
|
Oligomerization of the Tetraspanin CD81 via the Flexibility of Its δ-Loop. Biophys J 2017; 110:2463-2474. [PMID: 27276264 DOI: 10.1016/j.bpj.2016.05.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 01/28/2023] Open
Abstract
Tetraspanins are master organizers in the plasma membrane, forming tetraspanin-enriched microdomains with one another and other surface molecules. Their rod-shaped structure includes a large extracellular loop (LEL) that plays a pivotal role in tetraspanin network formation. We performed comparative atomistic and coarse-grain molecular-dynamics simulations of the LEL in isolation and full-length CD81, and reproduced LEL flexibility patterns known from wet-lab experiments in which the LEL δ-loop region showed a pronounced flexibility. In a 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine lipid bilayer and a plasma membrane environment, the conformational flexibility of the δ-loop initiates CD81-CD81 contacts for oligomerization. Furthermore, in the plasma membrane, CD81-ganglioside bridges arising from preformed glycolipid patches cross-link the complexes. The data suggest that exposing a flexible domain enables binding to interaction partners by circumventing the restriction of orientation and conformational freedom of membrane proteins.
Collapse
|
47
|
Zuidscherwoude M, Dunlock VME, van den Bogaart G, van Deventer SJ, van der Schaaf A, van Oostrum J, Goedhart J, In 't Hout J, Hämmerling GJ, Tanaka S, Nadler A, Schultz C, Wright MD, Adjobo-Hermans MJW, van Spriel AB. Tetraspanin microdomains control localized protein kinase C signaling in B cells. Sci Signal 2017; 10:eaag2755. [PMID: 28487417 DOI: 10.1126/scisignal.aag2755] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Activation of B cells by the binding of antigens to the B cell receptor (BCR) requires the protein kinase C (PKC) family member PKCβ. Because PKCs must translocate to the plasma membrane to become activated, we investigated the mechanisms regulating their spatial distribution in mouse and human B cells. Through live-cell imaging, we showed that BCR-stimulated production of the second messenger diacylglycerol (DAG) resulted in the translocation of PKCβ from the cytosol to plasma membrane regions containing the tetraspanin protein CD53. CD53 was specifically enriched at sites of BCR signaling, suggesting that BCR-dependent PKC signaling was initiated at these tetraspanin microdomains. Fluorescence lifetime imaging microscopy studies confirmed the molecular recruitment of PKC to CD53-containing microdomains, which required the amino terminus of CD53. Furthermore, we showed that Cd53-deficient B cells were defective in the phosphorylation of PKC substrates. Consistent with this finding, PKC recruitment to the plasma membrane was impaired in both mouse and human CD53-deficient B cells compared to that in their wild-type counterparts. These data suggest that CD53 promotes BCR-dependent PKC signaling by recruiting PKC to the plasma membrane so that it can phosphorylate its substrates and that tetraspanin-containing microdomains can act as signaling hotspots in the plasma membrane.
Collapse
Affiliation(s)
- Malou Zuidscherwoude
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Vera-Marie E Dunlock
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Geert van den Bogaart
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Sjoerd J van Deventer
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Alie van der Schaaf
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Jenny van Oostrum
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Joachim Goedhart
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, 1098 XH Amsterdam, Netherlands
| | - Joanna In 't Hout
- Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, 6500 HB Nijmegen, Netherlands
| | - Günter J Hämmerling
- Department of Molecular Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Satoshi Tanaka
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Japan
| | - André Nadler
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Carsten Schultz
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Mark D Wright
- Department of Immunology, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria 3004, Australia
| | - Merel J W Adjobo-Hermans
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Annemiek B van Spriel
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands.
| |
Collapse
|
48
|
Tspan2: a tetraspanin protein involved in oligodendrogenesis and cancer metastasis. Biochem Soc Trans 2017; 45:465-475. [PMID: 28408487 PMCID: PMC5390497 DOI: 10.1042/bst20160022] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 02/06/2017] [Accepted: 02/09/2017] [Indexed: 12/14/2022]
Abstract
Tetraspanin 2 (Tspan2) is one of the less well-characterised members of the tetraspanin superfamily, and its precise function in different human tissue types remains to be explored. Initial studies have highlighted its possible association in neuroinflammation and carcinogenesis. In the central nervous system, Tspan2 may contribute to the early stages of the oligodendrocyte differentiation into myelin-forming glia. Furthermore, in human lung cancer, Tspan2 could be involved in the progression of the tumour metastasis by modulating cancer cell motility and invasion functions. In this review, we discuss the available evidence for the potential role of Tspan2 and introduce possible strategies for disease targeting.
Collapse
|
49
|
Quaynor SD, Bosley ME, Duckworth CG, Porter KR, Kim SH, Kim HG, Chorich LP, Sullivan ME, Choi JH, Cameron RS, Layman LC. Targeted next generation sequencing approach identifies eighteen new candidate genes in normosmic hypogonadotropic hypogonadism and Kallmann syndrome. Mol Cell Endocrinol 2016; 437:86-96. [PMID: 27502037 DOI: 10.1016/j.mce.2016.08.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 08/03/2016] [Accepted: 08/03/2016] [Indexed: 11/15/2022]
Abstract
The genetic basis is unknown for ∼60% of normosmic hypogonadotropic hypogonadism (nHH)/Kallmann syndrome (KS). DNAs from (17 male and 31 female) nHH/KS patients were analyzed by targeted next generation sequencing (NGS) of 261 genes involved in hypothalamic, pituitary, and/or olfactory pathways, or suggested by chromosome rearrangements. Selected variants were subjected to Sanger DNA sequencing, the gold standard. The frequency of Sanger-confirmed variants was determined using the ExAC database. Variants were classified as likely pathogenic (frameshift, nonsense, and splice site) or predicted pathogenic (nonsynonymous missense). Two novel FGFR1 mutations were identified, as were 18 new candidate genes including: AMN1, CCKBR, CRY1, CXCR4, FGF13, GAP43, GLI3, JAG1, NOS1, MASTL, NOTCH1, NRP2, PALM2, PDE3A, PLEKHA5, RD3, and TRAPPC9, and TSPAN11. Digenic and trigenic variants were found in 8/48 (16.7%) and 1/48 (2.1%) patients, respectively. NGS with confirmation by Sanger sequencing resulted in the identification of new causative FGFR1 gene mutations and suggested 18 new candidate genes in nHH/KS.
Collapse
Affiliation(s)
- Samuel D Quaynor
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, United States; University of Chicago, Department of Neurology, Chicago, IL, United States
| | - Maggie E Bosley
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Christina G Duckworth
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Kelsey R Porter
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Soo-Hyun Kim
- Molecular Cell Sciences Research Centre, St. George's Medical School, University of London, London, UK
| | - Hyung-Goo Kim
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, United States; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Lynn P Chorich
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, United States; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Megan E Sullivan
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, United States; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Jeong-Hyeon Choi
- Department of Biostatistics and Epidemiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Richard S Cameron
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Lawrence C Layman
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, United States; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States; Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States.
| |
Collapse
|
50
|
Hang Q, Isaji T, Hou S, Zhou Y, Fukuda T, Gu J. N-Glycosylation of integrin α5 acts as a switch for EGFR-mediated complex formation of integrin α5β1 to α6β4. Sci Rep 2016; 6:33507. [PMID: 27641064 PMCID: PMC5027594 DOI: 10.1038/srep33507] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 08/23/2016] [Indexed: 01/17/2023] Open
Abstract
N-Glycosylation of integrin α5β1 is involved in multiple cell behaviors. We previously reported that the N-glycosylations of the calf domain on integrin α5 (S3–5,10–14) are essential for its inhibitory effect on EGFR signaling in regulating cell proliferation. However, the importance of the individual N-glycosylation and the underlying mechanisms of inhibition remain unclear. Here, we characterize the S3–5,10–14 mutants in detail and found that the N-glycosylation of site-11 (Asn712) is key for cell growth. The restoration of site-11, unlike the other individual sites, significantly suppressed cell growth and EGFR signaling in a manner that was similar to that of wild-type (WT). Mechanistically, this N-glycosylation inhibited the response abilities upon EGF stimulation and EGFR dimerization. Interestingly, we found this N-glycosylation controlled the EGFR complex formation with integrin α5β1 or α6β4; i.e., the loss of site-11 switched EGFR-α5β1 to EGFR-α6β4, which is well known to promote cellular signaling for cell growth. Moreover, the site-11 N-glycan exhibited a more branching structure compared with other sites, which may be required for EGFR-α5β1 formation. Taken together, these data clearly demonstrate that the site-11 N-glycosylation on α5 is most important for its inhibitory effect on EGFR signaling, which may provide a novel regulatory mechanism for crosstalks between integrins and EGFR.
Collapse
Affiliation(s)
- Qinglei Hang
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan
| | - Tomoya Isaji
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan
| | - Sicong Hou
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan
| | - Ying Zhou
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan
| | - Tomohiko Fukuda
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan
| |
Collapse
|