1
|
Nasif S, Colombo M, Uldry AC, Schröder M, de Brot S, Mühlemann O. Inhibition of nonsense-mediated mRNA decay reduces the tumorigenicity of human fibrosarcoma cells. NAR Cancer 2023; 5:zcad048. [PMID: 37681034 PMCID: PMC10480688 DOI: 10.1093/narcan/zcad048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/08/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
Nonsense-mediated mRNA decay (NMD) is a eukaryotic RNA decay pathway with roles in cellular stress responses, differentiation, and viral defense. It functions in both quality control and post-transcriptional regulation of gene expression. NMD has also emerged as a modulator of cancer progression, although available evidence supports both a tumor suppressor and a pro-tumorigenic role, depending on the model. To further investigate the role of NMD in cancer, we knocked out the NMD factor SMG7 in the HT1080 human fibrosarcoma cell line, resulting in suppression of NMD function. We then compared the oncogenic properties of the parental cell line, the SMG7-knockout, and a rescue cell line in which we re-introduced both isoforms of SMG7. We also tested the effect of a drug inhibiting the NMD factor SMG1 to distinguish NMD-dependent effects from putative NMD-independent functions of SMG7. Using cell-based assays and a mouse xenograft tumor model, we showed that suppression of NMD function severely compromises the oncogenic phenotype. Molecular pathway analysis revealed that NMD suppression strongly reduces matrix metalloprotease 9 (MMP9) expression and that MMP9 re-expression partially rescues the oncogenic phenotype. Since MMP9 promotes cancer cell migration and invasion, metastasis and angiogenesis, its downregulation may contribute to the reduced tumorigenicity of NMD-suppressed cells. Collectively, our results highlight the potential value of NMD inhibition as a therapeutic approach.
Collapse
Affiliation(s)
- Sofia Nasif
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Switzerland
| | - Martino Colombo
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Switzerland
| | - Anne-Christine Uldry
- Proteomics & Mass Spectrometry Core Facility, Department for BioMedical Research, University of Bern, Switzerland
| | - Markus S Schröder
- NCCR RNA & Disease Bioinformatics Support,Department of Biology, ETH Zürich, Switzerland
| | - Simone de Brot
- COMPATH, Institute of Animal Pathology, University of Bern, Switzerland
| | - Oliver Mühlemann
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Switzerland
| |
Collapse
|
2
|
Yang Q, Liu J, Wu B, Wang X, Jiang Y, Zhu D. Role of extracellular vesicles in osteosarcoma. Int J Med Sci 2022; 19:1216-1226. [PMID: 35928720 PMCID: PMC9346389 DOI: 10.7150/ijms.74137] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
Osteosarcoma is a malignant bone tumor characterized by the direct production of osteoid tissue from tumor cells. Extracellular vesicles are membranous vesicles released by cells into the extracellular matrix, which exist widely in various body fluids and cell supernatants, and stably carry some important signaling molecules. They are involved in cell communication, cell migration, angiogenesis and tumor cell growth. Increasing evidence has shown that extracellular vesicles play a significant role in osteosarcoma development, progression, and metastatic process, indicating that extracellular vesicles can be use as biomarker vehicles in the diagnosis and prognosis of osteosarcoma. This review discusses the basic biological characteristics of extracellular vesicles and focuses on their application in osteosarcoma.
Collapse
Affiliation(s)
- Qifan Yang
- Department of Orthopaedics, the First Hospital of Jilin University, Changchun, Street Xinmin 71, China
| | - Jing Liu
- The first clinical medical college of Bin Zhou Medical College, Street Huanghe 661, China
| | - Bo Wu
- Department of Orthopaedics, the First Hospital of Jilin University, Changchun, Street Xinmin 71, China
| | - Xinyu Wang
- Department of Orthopaedics, the First Hospital of Jilin University, Changchun, Street Xinmin 71, China
| | - Ye Jiang
- Department of Orthopaedics, the First Hospital of Jilin University, Changchun, Street Xinmin 71, China
| | - Dong Zhu
- Department of Orthopaedics, the First Hospital of Jilin University, Changchun, Street Xinmin 71, China
| |
Collapse
|
3
|
El Kharbili M, Cario M, Béchetoille N, Pain C, Boucheix C, Degoul F, Masse I, Berthier-Vergnes O. Tspan8 Drives Melanoma Dermal Invasion by Promoting ProMMP-9 Activation and Basement Membrane Proteolysis in a Keratinocyte-Dependent Manner. Cancers (Basel) 2020; 12:cancers12051297. [PMID: 32455575 PMCID: PMC7281247 DOI: 10.3390/cancers12051297] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/15/2020] [Accepted: 05/16/2020] [Indexed: 12/12/2022] Open
Abstract
Melanoma is the most aggressive skin cancer with an extremely challenging therapy. The dermal-epidermal junction (DEJ) degradation and subsequent dermal invasion are the earliest steps of melanoma dissemination, but the mechanisms remain elusive. We previously identified Tspan8 as a key actor in melanoma invasiveness. Here, we investigated Tspan8 mechanisms of action during dermal invasion, using a validated skin-reconstruct-model that recapitulates melanoma dermal penetration through an authentic DEJ. We demonstrate that Tspan8 is sufficient to induce melanoma cells’ translocation to the dermis. Mechanistically, Tspan8+ melanoma cells cooperate with surrounding keratinocytes within the epidermis to promote keratinocyte-originated proMMP-9 activation process, collagen IV degradation and dermal colonization. This concurs with elevated active MMP-3 and low TIMP-1 levels, known to promote MMP-9 activity. Finally, a specific Tspan8-antibody reduces proMMP-9 activation and dermal invasion. Overall, our results provide new insights into the role of keratinocytes in melanoma dermal colonization through a cooperative mechanism never reported before, and establish for the first time the pro-invasive role of a tetraspanin family member in a cell non-autonomous manner. This work also displays solid arguments for the use of Tspan8-blocking antibodies to impede early melanoma spreading and therefore metastasis.
Collapse
Affiliation(s)
- Manale El Kharbili
- Centre de Génétique et de Physiologie Moléculaires et Cellulaires, CNRS UMR5534, Université de Lyon, F-69003 Lyon, France; (M.E.K.); (O.B.-V.)
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Muriel Cario
- National Reference Center for Rare Skin Disease, Department of Dermatology, University Hospital, INSERM 1035, F-33000 Bordeaux, France; (M.C.); (C.P.)
- AquiDerm, University Bordeaux, F-33076 Bordeaux, France
| | | | - Catherine Pain
- National Reference Center for Rare Skin Disease, Department of Dermatology, University Hospital, INSERM 1035, F-33000 Bordeaux, France; (M.C.); (C.P.)
| | - Claude Boucheix
- INSERM U935, Université Paris-Sud, F-94800 Villejuif, France;
| | - Françoise Degoul
- INSERM U1240, Université Clermont Auvergne, Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont Ferrand, France;
| | - Ingrid Masse
- Centre de Génétique et de Physiologie Moléculaires et Cellulaires, CNRS UMR5534, Université de Lyon, F-69003 Lyon, France; (M.E.K.); (O.B.-V.)
- Centre de Recherche en Cancérologie de Lyon, CNRS-UMR5286, INSERM U1052, Université de Lyon, F-69008 Lyon, France
- Correspondence:
| | - Odile Berthier-Vergnes
- Centre de Génétique et de Physiologie Moléculaires et Cellulaires, CNRS UMR5534, Université de Lyon, F-69003 Lyon, France; (M.E.K.); (O.B.-V.)
- US7INSERM /UMS3453 UCBL SFR Santé Lyon-Est, F-69372 Lyon, France
| |
Collapse
|
4
|
Leung KT, Zhang C, Chan KYY, Li K, Cheung JTK, Ng MHL, Zhang XB, Sit T, Lee WYW, Kang W, To KF, Yu JWS, Man TKF, Wang H, Tsang KS, Cheng FWT, Lam GKS, Chow TW, Leung AWK, Leung TF, Yuen PMP, Ng PC, Li CK. CD9 blockade suppresses disease progression of high-risk pediatric B-cell precursor acute lymphoblastic leukemia and enhances chemosensitivity. Leukemia 2019; 34:709-720. [DOI: 10.1038/s41375-019-0593-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 08/13/2019] [Accepted: 08/15/2019] [Indexed: 12/12/2022]
|
5
|
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: 22] [Impact Index Per Article: 3.1] [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
|
6
|
Lan M, Zhu XP, Cao ZY, Liu JM, Lin Q, Liu ZL. Extracellular vesicles-mediated signaling in the osteosarcoma microenvironment: Roles and potential therapeutic targets. J Bone Oncol 2018; 12:101-104. [PMID: 30155405 PMCID: PMC6111053 DOI: 10.1016/j.jbo.2018.07.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 07/17/2018] [Accepted: 07/18/2018] [Indexed: 12/19/2022] Open
Abstract
Osteosarcoma (OS) is the most common non-hematologic malignant tumor of bone in children. It is usually characterized by a high risk of developing lung metastasis and poor prognosis. Extracellular vesicles (EVs) are cell-derived nanoparticles with a small size of 50–200 nm in diameter. As a communicator, the contents of the EVs secreted via either fusing with lysosomes for degradation and recycling or fusing with the cell plasma membrane into the extracellular environment, which play an important role in regulating the tumor microenvironment of OS and mediating the Wnt/β-catenin and TGF-β signalings. Increasing evidences suggest that EVs have significant role in OS growth, progression, metastasis and drug resistance. In this study, the roles of EVs in the physiology and pathogenesis of OS and the potential attractive therapeutic target for the treatment of OS were reviewed.
Collapse
Affiliation(s)
- Min Lan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Xiao-Ping Zhu
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Zhi-Yuan Cao
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Jia-Ming Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Qing Lin
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Zhi-Li Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| |
Collapse
|
7
|
Vences-Catalán F, Levy S. Immune Targeting of Tetraspanins Involved in Cell Invasion and Metastasis. Front Immunol 2018; 9:1277. [PMID: 29946318 PMCID: PMC6006414 DOI: 10.3389/fimmu.2018.01277] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 05/22/2018] [Indexed: 01/15/2023] Open
Abstract
Metastasis is the ultimate consequence of cancer progression and the cause of patients’ death across different cancer types. Patients with initial diagnosis of distant disease have a worst 5-year survival compared to patients with localized disease. Therapies that target primary tumors fail to eradicate distant dissemination of cancer. Recently, immunotherapies have improved the survival of patients with metastatic disease, such as melanoma and lung cancer. However, only a fraction of patients responds to immunotherapy modalities that target the host immune system. The need to identify new druggable targets that inhibit or prevent metastasis is, therefore, much needed. Tetraspanins have emerged as key players in regulating cell migration, invasion, and metastasis. By serving as molecular adaptors that cluster adhesion receptors, signaling molecules, and cell surface receptors; tetraspanins are involved in all steps of the metastatic cascade. They regulate cell proliferation, participate in EMT transition, modulate integrin-mediated cell adhesion, and participate in angiogenesis and invasion processes. Tetraspanins have also been shown to modulate metastasis indirectly through exosomes and by regulating cellular interactions in the immune system. Importantly, targeting individual tetraspanin with antibodies has impacted tumor progression. This review will focus on the contribution of tetraspanins to the metastatic process and their potential as therapeutic tumor targets.
Collapse
Affiliation(s)
- Felipe Vences-Catalán
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Shoshana Levy
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
8
|
Lin Q, Peng S, Yang Y. Inhibition of CD9 expression reduces the metastatic capacity of human hepatocellular carcinoma cell line MHCC97-H. Int J Oncol 2018; 53:266-274. [PMID: 29749468 DOI: 10.3892/ijo.2018.4381] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 04/17/2018] [Indexed: 11/06/2022] Open
Abstract
Metastasis is a characteristic of malignant tumors and may be a fatal clinical factor for many patients with cancer. Hepatocellular carcinoma (HCC) cells are highly metastatic; the mechanism of metastasis is complicated and may be influenced by a number of factors. Membrane proteins may block receptors or inhibit important enzymes, thus inhibiting tumor progression, and may be potential therapeutic targets for tumor prognosis and treatment. The present study aimed to use proteomics to analyze the dynamic changes of membrane proteins in HCC cells, to improve our understanding of membrane protein functions and to clarify the important components of the mechanisms of HCC metastasis. The present study used the highly metastatic MHCC97-H and the lowly metastatic MHCC97-L HCC cell lines, and the isobaric tags for relative and absolute quantitation (iTRAQ) approach was used for high-throughput screening of metastasis-related membrane proteins. A total of 22 membrane proteins were identified as differentially expressed between the MHCC97-H and MHCC97-L cell lines; these results were verified by reverse transcription-quantitative polymerase chain reaction and western blotting. A number of the identified proteins were revealed to be related to tumor metastasis, including the tetraspan in transmembrane protein CD9. CD9 was demonstrated to be highly expressed in MHCC97-H cells compared with MHCC97-L cells. The functional role of CD9 was characterized by inhibiting its expression using a small interfering RNAs, which demonstrated that reduced CD9 expression inhibited cell migration and metastasis, as determined by wound-healing and invasion assays. Results from the present study demonstrated that CD9 was highly expressed in the highly metastatic HCC cells and promoted HCC cell migration. This protein may be a novel target for regulating the invasive phenotype in HCC.
Collapse
Affiliation(s)
- Qing Lin
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Shifang Peng
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yixuan Yang
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
9
|
Sechler M, Parrish JK, Birks DK, Jedlicka P. The histone demethylase KDM3A, and its downstream target MCAM, promote Ewing Sarcoma cell migration and metastasis. Oncogene 2017; 36:4150-4160. [PMID: 28319067 PMCID: PMC5519422 DOI: 10.1038/onc.2017.44] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 01/27/2017] [Accepted: 02/02/2017] [Indexed: 12/15/2022]
Abstract
Ewing Sarcoma is the second most common solid pediatric malignant neoplasm of bone and soft tissue. Driven by EWS/Ets, or rarely variant, oncogenic fusions, Ewing Sarcoma is a biologically and clinically aggressive disease with a high propensity for metastasis. However, the mechanisms underpinning Ewing Sarcoma metastasis are currently not well understood. In the present study, we identify and characterize a novel metastasis-promotional pathway in Ewing Sarcoma, involving the histone demethylase KDM3A, previously identified by our laboratory as a new cancer-promoting gene in this disease. Using global gene expression profiling, we show that KDM3A positively regulates genes and pathways implicated in cell migration and metastasis, and demonstrate, using functional assays, that KDM3A promotes migration in vitro and experimental, post-intravasation, metastasis in vivo. We further identify the melanoma cell adhesion molecule (MCAM) as a novel KDM3A target gene in Ewing Sarcoma, and an important effector of KDM3A pro-metastatic action. Specifically, we demonstrate that MCAM depletion, like KDM3A depletion, inhibits cell migration in vitro and experimental metastasis in vivo, and that MCAM partially rescues impaired migration due to KDM3A knock-down. Mechanistically, we show that KDM3A regulates MCAM expression both through a direct mechanism, involving modulation of H3K9 methylation at the MCAM promoter, and an indirect mechanism, via the Ets1 transcription factor. Finally, we identify an association between high MCAM levels in patient tumors and poor survival, in two different Ewing Sarcoma clinical cohorts. Taken together, our studies uncover a new metastasis-promoting pathway in Ewing Sarcoma, with therapeutically targetable components.
Collapse
Affiliation(s)
- Marybeth Sechler
- Cancer Biology Graduate Training Program
- University of Colorado Denver, Anschutz Medical Campus, Aurora CO
| | - Janet K. Parrish
- Department of Pathology
- University of Colorado Denver, Anschutz Medical Campus, Aurora CO
| | - Diane K. Birks
- Department of Neurosurgery
- University of Colorado Denver, Anschutz Medical Campus, Aurora CO
| | - Paul Jedlicka
- Cancer Biology Graduate Training Program
- Department of Pathology
- University of Colorado Denver, Anschutz Medical Campus, Aurora CO
| |
Collapse
|
10
|
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
|
11
|
Rappa G, Green TM, Karbanová J, Corbeil D, Lorico A. Tetraspanin CD9 determines invasiveness and tumorigenicity of human breast cancer cells. Oncotarget 2016; 6:7970-91. [PMID: 25762645 PMCID: PMC4480729 DOI: 10.18632/oncotarget.3419] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 02/02/2015] [Indexed: 01/10/2023] Open
Abstract
Interaction of breast cancer cells (BCCs) with stromal components is critical for tumor growth and metastasis. Here, we assessed the role of CD9 in adhesion, migration and invasiveness of BCCs. We used co-cultures of BCCs and bone marrow-derived multipotent mesenchymal stromal cells (MSCs), and analyzed their behavior and morphology by dynamic total internal reflection fluorescence, confocal and scanning electron microscopy. 83, 16 and 10% of contacts between MDA-MB-231 (MDA), MA-11 or MCF-7 cells and MSCs, respectively, resulted in MSC invasion. MDA cells developed long magnupodia, lamellipodia and dorsal microvilli, whereas long microvilli emerged from MA-11 cells. MCF-7 cells displayed large dorsal ruffles. CD9 knockdown and antibody blockage in MDA cells inhibited MSC invasion by 95 and 70%, respectively, suggesting that CD9 is required for this process. Remarkably, CD9-deficient MDA cells displayed significant alteration of their plasma membrane, harboring numerous peripheral and dorsal membrane ruffles instead of intact magnupodium/lamellipodium and microvillus, respectively. Such modification might explain the delayed adhesion, and hence MSC invasion. In agreement with this hypothesis, CD9-knockdown suppressed the metastatic capacity of MDA cells in mouse xenografts. Our data indicate that CD9 is implicated in BCC invasiveness and metastases by cellular mechanisms that involve specific CD9+ plasma membrane protrusions of BCCs.
Collapse
Affiliation(s)
- Germana Rappa
- Cancer Research Center, Roseman University of Health Sciences with Roseman University College of Medicine, Las Vegas, Nevada, USA
| | - Toni M Green
- Cancer Research Center, Roseman University of Health Sciences with Roseman University College of Medicine, Las Vegas, Nevada, USA
| | - Jana Karbanová
- Tissue Engineering Laboratories (BIOTEC) and DFG Research Center and Cluster of Excellence for Regenerative Therapies Dresden, Technische Universität Dresden, Tatzberg, Dresden, Germany
| | - Denis Corbeil
- Tissue Engineering Laboratories (BIOTEC) and DFG Research Center and Cluster of Excellence for Regenerative Therapies Dresden, Technische Universität Dresden, Tatzberg, Dresden, Germany
| | - Aurelio Lorico
- Cancer Research Center, Roseman University of Health Sciences with Roseman University College of Medicine, Las Vegas, Nevada, USA
| |
Collapse
|
12
|
YU SEONMI, KIM SONGJA. Salinomycin causes migration and invasion of human fibrosarcoma cells by inducing MMP-2 expression via PI3-kinase, ERK-1/2 and p38 kinase pathways. Int J Oncol 2016; 48:2686-92. [DOI: 10.3892/ijo.2016.3448] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/08/2016] [Indexed: 11/06/2022] Open
|
13
|
Huan J, Gao Y, Xu J, Sheng W, Zhu W, Zhang S, Cao J, Ji J, Zhang L, Tian Y. Overexpression of CD9 correlates with tumor stage and lymph node metastasis in esophageal squamous cell carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:3054-3061. [PMID: 26045817 PMCID: PMC4440126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 02/26/2015] [Indexed: 06/04/2023]
Abstract
OBJECTIVE Esophageal squamous cell carcinoma (ESCC) is one of the leading causes of cancer deaths worldwide. CD9 has been reported to play a critical role in cell motility, growth and metastasis of multiple cancers. The present study investigated the clinicopathological features of CD9, and its biological characteristics in ESCC. METHODS Fifteen normal esophageal tissue specimens, fifty-three ESCC adjacent tissues and one hundred and four ESCC tissues were included in this study. Using immunohistochemistry (IHC), the expression levels of CD9 were evaluated among different samples. And its clinicopathological parameters and its prognostic factors were analyzed. Western blotting was used to measure CD9 expression and colony formation was performed to determine the effect of CD9 on cell growth in ESCC TE-1 cells. RESULTS Compared with normal esophageal tissues and tumor adjacent tissues, CD9 expression level is significantly higher in ESCC tissues. CD9 expression correlated with tumor stage (P=0.022) and lymph node metastasis (P=0.019) in ESCC patients. Furthermore, the small interfering RNA-mediated silencing of CD9 expression in TE-1 cells resulted in increased proliferation as evidenced by increased colony number and colony size. CONCLUSION CD9 expression is upregulated in ESCC tissues and its expression is correlated with tumor stage and lymph node metastasis in ESCC patients. CD9 suppresses the proliferation of TE-1 cells. CD9 may present a potential in tumor progression in ESCC.
Collapse
Affiliation(s)
- Jian Huan
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow UniversitySuzhou 215004, China
| | - Yi Gao
- Department of Gastroenterology, The Affiliated Jiangyin Hospital of Southeast UniversityJiangyin 214400, China
| | - Jing Xu
- School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow UniversitySuzhou 215123, China
| | - Wenjiong Sheng
- School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow UniversitySuzhou 215123, China
| | - Wei Zhu
- School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow UniversitySuzhou 215123, China
| | - Shuyu Zhang
- School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow UniversitySuzhou 215123, China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow UniversitySuzhou 215123, China
| | - Jianping Cao
- School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow UniversitySuzhou 215123, China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow UniversitySuzhou 215123, China
| | - Jiang Ji
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow UniversitySuzhou 215004, China
| | - Liyuan Zhang
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow UniversitySuzhou 215004, China
| | - Ye Tian
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow UniversitySuzhou 215004, China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow UniversitySuzhou 215123, China
| |
Collapse
|
14
|
Tetraspanin CD9 regulates cell contraction and actin arrangement via RhoA in human vascular smooth muscle cells. PLoS One 2014; 9:e106999. [PMID: 25184334 PMCID: PMC4153684 DOI: 10.1371/journal.pone.0106999] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 08/07/2014] [Indexed: 02/07/2023] Open
Abstract
The most prevalent cardiovascular diseases arise from alterations in vascular smooth muscle cell (VSMC) morphology and function. Tetraspanin CD9 has been previously implicated in regulating vascular pathologies; however, insight into how CD9 may regulate adverse VSMC phenotypes has not been provided. We utilized a human model of aortic smooth muscle cells to understand the consequences of CD9 deficiency on VSMC phenotypes. Upon knocking down CD9, the cells developed an abnormally small and rounded morphology. We determined that this morphological change was due to a lack of typical parallel actin arrangement. We also found similar total RhoA but decreased GTP-bound (active) RhoA levels in CD9 deficient cells. As a result, cells lacking a full complement of CD9 were less contractile than their control treated counterparts. Upon restoration of RhoA activity in the CD9 deficient cells, the phenotype was reversed and cell contraction was restored. Conversely, inhibition of RhoA activity in the control cells mimicked the CD9-deficient cell phenotype. Thus, alteration in CD9 expression was sufficient to profoundly disrupt cellular actin arrangement and endogenous cell contraction by interfering with RhoA signaling. This study provides insight into how CD9 may regulate previously described vascular smooth muscle cell pathophysiology.
Collapse
|
15
|
Extracellular Membrane Vesicles Derived from 143B Osteosarcoma Cells Contain Pro-Osteoclastogenic Cargo: A Novel Communication Mechanism in Osteosarcoma Bone Microenvironment. Transl Oncol 2014; 7:331-40. [PMID: 25180057 PMCID: PMC4145399 DOI: 10.1016/j.tranon.2014.04.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 01/21/2014] [Accepted: 03/03/2014] [Indexed: 12/21/2022] Open
Abstract
The bone microenvironment (BME) is the main hub of all skeletal related pathological events in osteosarcoma leading to tumor induced bone destruction, and decreasing overall bone quality and bone strength. The role of extra-cellular membrane vesicles (EMVs) as mediators of intercellular communication in modulating osteosarcoma-BME is unknown, and needs to be investigated. It is our hypothesis that osteosarcoma-EMVs contain pro-osteoclastogenic cargo which increases osteoclastic activity, and dysregulated bone remodeling in the osteosarcoma-BME. In this study, EMVs were isolated from the conditioned media of 143B and HOS human osteosarcoma cell cultures using differential ultracentrifugation. Nano-particle tracking analysis determined EMVs in the size range of 50-200 nm in diameter. The EMV yield from 143B cells was relatively higher compared to HOS cells. Transmission electron microscopy confirmed the ultrastructure of 143B-EMVs and detected multivesicular bodies. Biochemical characterization of 143B-EMVs detected the expression of bioactive pro-osteoclastic cargo including matrix metalloproteinases-1 and -13 (MMP-1, -13), transforming growth factor-β (TGF-β), CD-9, and receptor activator of nuclear factor kappa-β ligand (RANKL). Detection of a protein signature that is uniquely pro-osteoclastic in 143B-EMVs is a novel finding, and is significant as EMVs represent an interesting mechanism for potentially mediating bone destruction in the osteosarcoma-BME. This study further demonstrates that 143B cells actively mobilize calcium in the presence of ionomycin, and forskolin, and induce cytoskeleton rearrangements leading to vesicular biogenesis. In conclusion, this study demonstrates that 143B osteosarcoma cells generate EMVs mainly by mechanisms involving increased intracellular calcium or cAMP levels, and contain pro-osteoclastic cargo.
Collapse
|
16
|
Herr MJ, Longhurst CM, Baker B, Homayouni R, Speich HE, Kotha J, Jennings LK. Tetraspanin CD9 modulates human lymphoma cellular proliferation via histone deacetylase activity. Biochem Biophys Res Commun 2014; 447:616-20. [PMID: 24747564 DOI: 10.1016/j.bbrc.2014.04.046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 04/09/2014] [Indexed: 11/26/2022]
Abstract
Non-Hodgkin Lymphoma (NHL) is a type of hematological malignancy that affects two percent of the overall population in the United States. Tetraspanin CD9 is a cell surface protein that has been thoroughly demonstrated to be a molecular facilitator of cellular phenotype. CD9 expression varies in two human lymphoma cell lines, Raji and BJAB. In this report, we investigated the functional relationship between CD9 and cell proliferation regulated by histone deacetylase (HDAC) activity in these two cell lines. Introduction of CD9 expression in Raji cells resulted in significantly increased cell proliferation and HDAC activity compared to Mock transfected Raji cells. The increase in CD9-Raji cell proliferation was significantly inhibited by HDAC inhibitor (HDACi) treatment. Pretreatment of BJAB cells with HDAC inhibitors resulted in a significant decrease in endogenous CD9 mRNA and cell surface expression. BJAB cells also displayed decreased cell proliferation after HDACi treatment. These results suggest a significant relationship between CD9 expression and cell proliferation in human lymphoma cells that may be modulated by HDAC activity.
Collapse
Affiliation(s)
- Michael J Herr
- Vascular Biology Center of Excellence, The University of Tennessee Health Science Center, Memphis, TN 38163, United States; Department of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, United States; Department of Molecular Sciences, The University of Tennessee Health Science Center, Memphis, TN 38163, United States; Department of Surgery, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Celia M Longhurst
- Vascular Biology Center of Excellence, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Benjamin Baker
- Vascular Biology Center of Excellence, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Ramin Homayouni
- Department of Biology, Bioinformatics Program, University of Memphis, Memphis, TN 38152, United States
| | - Henry E Speich
- Vascular Biology Center of Excellence, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Jayaprakash Kotha
- Vascular Biology Center of Excellence, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Lisa K Jennings
- Vascular Biology Center of Excellence, The University of Tennessee Health Science Center, Memphis, TN 38163, United States; Department of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, United States; Department of Molecular Sciences, The University of Tennessee Health Science Center, Memphis, TN 38163, United States; Department of Surgery, The University of Tennessee Health Science Center, Memphis, TN 38163, United States; Department of Biology, Bioinformatics Program, University of Memphis, Memphis, TN 38152, United States.
| |
Collapse
|
17
|
Herr MJ, Mabry SE, Jameson JF, Jennings LK. Pro-MMP-9 upregulation in HT1080 cells expressing CD9 is regulated by epidermal growth factor receptor. Biochem Biophys Res Commun 2013; 442:99-104. [PMID: 24246676 DOI: 10.1016/j.bbrc.2013.11.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 11/04/2013] [Indexed: 11/26/2022]
Abstract
Degradation of the surrounding extracellular matrix (ECM) by matrix metalloproteinases (MMPs) drives invasion and metastasis of cancer cells. We previously demonstrated that tetraspanin CD9 expression upregulates pro-MMP-9 expression and release and promotes cellular invasion in a human fibrosarcoma cell line (HT1080). These events were dependent upon the highly functional second extracellular loop of CD9. We report here that the epidermal growth factor receptor (EGFR) tyrosine kinase expression and activity are involved in the CD9-mediated increase in pro-MMP-9 release and cellular invasion. Pro-MMP-9 expression was significantly decreased in a dose-dependent manner using first a broad spectrum receptor tyrosine kinase inhibitor and multiple specific EGFR inhibitors in CD9-HT1080 cells. Furthermore, gefitinib treatment of CD9-HT1080 cells reduced invasion through matrigel. EGFR knockdown using short interfering RNA resulted in decreased pro-MMP-9 expression and release into the media and subsequent cellular invasion without affecting CD9 expression or localization. Conclusively, this study points to EGFR as a key mediator between CD9-mediated pro-MMP-9 release and cellular invasion of HT1080 cells.
Collapse
Affiliation(s)
- Michael J Herr
- The Vascular Biology Center of Excellence, Department of Internal Medicine, USA; Department of Microbiology, Immunology, and Biochemistry, USA
| | | | | | | |
Collapse
|