1
|
Xu K, Feng H, Zhao R, Huang Y. Targeting Tetraspanins at Cell Interfaces: Functional Modulation and Exosome-Based Drug Delivery for Precise Disease Treatment. ChemMedChem 2025; 20:e202400664. [PMID: 39415492 DOI: 10.1002/cmdc.202400664] [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: 08/26/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 10/18/2024]
Abstract
Tetraspanins are key players in various physiological and pathological processes, including malignancy, immune response, fertilization, and infectious disease. Affinity ligands targeting the interactions between tetraspanins and partner proteins are promising for modulating downstream signaling pathways, thus emerging as attractive candidates for interfering related biological functions. Due to the involvement in vesicle biogenesis and cargo trafficking, tetraspanins are also regarded as exosome markers, and become molecular targets for drug loading and delivery. Given the rapid development in these areas, this minireview focuses on recent advances in design and engineering of affinity binders toward tetraspanins including CD63, CD81, and CD9. Their mechanism of actions in modulating protein interactions at cell interfaces and treatment of malignant diseases are discussed. Strategies for constructing exosome-based drug delivery platforms are also reviewed, with emphasis on the important roles of tetraspanins and the affinity ligands. Finally, challenges and future development of tetraspanin-targeting therapy and exosomal drug delivery platforms are also discussed.
Collapse
Affiliation(s)
- Kun Xu
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huixia Feng
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Rui Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanyan Huang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
2
|
Lestari DY, Mastutik G, Mukono IS. Betulinic acid and oleanolic acid modulate CD81 expression and induce apoptosis in triple-negative breast cancer cells through ROS generation. Med Oncol 2024; 42:24. [PMID: 39644426 DOI: 10.1007/s12032-024-02574-4] [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: 10/05/2024] [Accepted: 11/22/2024] [Indexed: 12/09/2024]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer characterized by a lack of hormones receptors and the HER2 receptor, making it unresponsive to targeted therapy. Triterpenoids such as betulinic acid (BA) and oleanolic acid (OA) have anticancer effects by inducing apoptosis in TNBC cells. CD81 is a tetraspanin that affects the growth and metastasis of cancer cells. To examine the effect of BA and OA on the viability of TNBC cell line (MDA-MB 231) by analyzing the CD81 expression, intracellular ROS, and apoptosis. The MDA-MB 231 cells was cultured and treated by BA and OA. The viability cell was evaluated by the CCK8 assay. This study analyzed the binding of BA and OA with CD81 using molecular docking and evaluated CD81 expression, intracellular ROS, and apoptosis by flow cytometry. The result showed that BA and OA inhibited viability of MDA-MB-231 cells. BA and OA bind to CD81 in silico, with binding affinities of 9.0 kcal/mol for BA and 7.2 kcal/mol for OA. Flow cytometry results revealed that BA can downregulate CD81 expression. BA and OA also increased intracellular ROS levels and induced apoptosis. These findings suggest that BA and OA, especially BA, can modulate CD81 expression and promote apoptosis in TNBC cells through the generation of ROS, thereby offering a potential therapeutic strategy for the treatment of TNBC.
Collapse
Affiliation(s)
- Dian Yuliartha Lestari
- Doctoral Program of Medical Science, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Medical Faculty, University of Muhammadiyah Malang, Malang, Indonesia
| | - Gondo Mastutik
- Department of Pathology Anatomy, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Indri Safitri Mukono
- Department of Physiology and Medical Biochemistry, Faculty of Medicine, Univesitas Airlangga, Surabaya, Indonesia.
| |
Collapse
|
3
|
Hüser L, Chhabra Y, Gololobova O, Wang V, Liu G, Dixit A, Rocha MR, Harper EI, Fane ME, Marino-Bravante GE, Zabransky DJ, Cai KQ, Utikal J, Slusher BS, Walston J, Lipson EJ, Witwer KW, Weeraratna AT. Aged fibroblast-derived extracellular vesicles promote angiogenesis in melanoma. Cell Rep 2024; 43:114721. [PMID: 39255061 PMCID: PMC11835374 DOI: 10.1016/j.celrep.2024.114721] [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: 11/13/2023] [Revised: 07/09/2024] [Accepted: 08/20/2024] [Indexed: 09/12/2024] Open
Abstract
Advancing age is a negative prognostic factor for cutaneous melanoma. However, the role of extracellular vesicles (EVs) within the melanoma tumor microenvironment (TME) has remained unexplored in the context of aging. While the size and morphology of the EVs isolated from young vs. aged fibroblasts remained unaltered, the contents of the protein cargo were changed. Aging reduced the expression of the tetraspanin CD9 in both the dermal fibroblasts and released EVs. CD9 is a crucial regulator of EV cargo sorting. Modulating the CD9 expression in fibroblasts was sufficient to alter its levels in EVs. Mass spectrometry analysis of EVs released by CD9 knockdown (KD) vs. control cells revealed a significant increase in angiopoietin-like protein 2 (ANGPTL2), an angiogenesis promoter. Analysis of primary endothelial cells confirmed increased sprouting under CD9 KD conditions. Together, our data indicate that aged EVs play an important role in promoting a tumor-permissive microenvironment.
Collapse
Affiliation(s)
- Laura Hüser
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA; Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Yash Chhabra
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA; Research Program Cancer Signaling and Microenvironment, Fox Chase Institute for Cancer Research, Philadelphia, PA, USA
| | - Olesia Gololobova
- Department of Molecular and Comparative Pathobiology, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Vania Wang
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Guanshu Liu
- Department of Radiology and Radiological Science, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Agrani Dixit
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Murilo Ramos Rocha
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Elizabeth I Harper
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Mitchell E Fane
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA; Research Program Cancer Signaling and Microenvironment, Fox Chase Institute for Cancer Research, Philadelphia, PA, USA
| | - Gloria E Marino-Bravante
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Daniel J Zabransky
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kathy Q Cai
- Research Program Cancer Signaling and Microenvironment, Fox Chase Institute for Cancer Research, Philadelphia, PA, USA
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Barbara S Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry and Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeremy Walston
- Department of Medicine - Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Oncology - Hematologic Malignancies, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Evan J Lipson
- Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA; Bloomberg Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, USA
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ashani T Weeraratna
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
4
|
Martorana E, Raciti G, Giuffrida R, Bruno E, Ficarra V, Ludovico GM, Suardi NR, Iraci N, Leggio L, Bussolati B, Grange C, Lorico A, Leonardi R, Forte S. A Novel Liquid Biopsy Method Based on Specific Combinations of Vesicular Markers Allows Us to Discriminate Prostate Cancer from Hyperplasia. Cells 2024; 13:1286. [PMID: 39120316 PMCID: PMC11311686 DOI: 10.3390/cells13151286] [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: 04/23/2024] [Revised: 07/19/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Prostate cancer is the second most common cancer in males worldwide, and its incidence is rising. Early detection is crucial for improving the outcomes, but the current screening methods have limitations. While prostate-specific antigen (PSA) testing is the most widely used screening tool, it has poor specificity, leading to a high rate of false positives and unnecessary biopsies. The existing biopsy techniques are invasive and are associated with complications. The liquid biopsy methods that analyze the biomarkers in blood or other bodily fluids offer a non-invasive and more accurate alternative for detecting and characterizing prostate tumors. METHODS Here, we present a novel liquid biopsy method for prostate cancer based on the identification of specific proteins in the extracellular vesicles isolated from the blood of patients with prostate cancer. RESULTS We observed that a specific combination of sEV proteins is a sensitive indicator of prostate cancer. Indeed, we found that the number of clusters expressed by specific combinations of either intra-vesicular (STAT3 and CyclinD1) or surface proteins (ERBB3, ALK, and CD81) allowed us to significantly discriminate the patients with prostate cancer from the individuals with hyperplasia. CONCLUSION This new liquid biopsy method has the potential to improve prostate cancer screening by providing a non-invasive and more accurate diagnostic tool.
Collapse
Affiliation(s)
- Emanuele Martorana
- IOM Ricerca Srl, Viagrande, 95029 Catania, Italy; (E.M.); (G.R.); (R.G.); (A.L.)
| | - Gabriele Raciti
- IOM Ricerca Srl, Viagrande, 95029 Catania, Italy; (E.M.); (G.R.); (R.G.); (A.L.)
- Department of Biomedical, Dental and Morphological and Functional Imaging Sciences, University of Messina, 98122 Messina, Italy
| | - Raffaella Giuffrida
- IOM Ricerca Srl, Viagrande, 95029 Catania, Italy; (E.M.); (G.R.); (R.G.); (A.L.)
| | - Elena Bruno
- Department of Physic and Astronomy “Ettore Majorana”, University of Catania, 95123 Catania, Italy;
| | - Vincenzo Ficarra
- Azienda Ospedaliera Policlinico Universitario “G. Martino”, Dipartimento di Patologia Umana dell’Adulto e dell’Età Evolutiva, 98124 Messina, Italy;
| | - Giuseppe Mario Ludovico
- Ospedale Generale Regionale “F. Miulli”, Divisione di Urologia, Acquaviva Delle Fonti, 70021 Bari, Italy;
| | - Nazareno Roberto Suardi
- Azienda Ospedaliera Policlinico Universitario Di Genova, Divisione di Urologia, 16132 Genova, Italy;
| | - Nunzio Iraci
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (N.I.); (L.L.)
| | - Loredana Leggio
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (N.I.); (L.L.)
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10124 Turin, Italy;
| | - Cristina Grange
- Department of Medical Sciences, University of Turin, 10124 Turin, Italy;
| | - Aurelio Lorico
- IOM Ricerca Srl, Viagrande, 95029 Catania, Italy; (E.M.); (G.R.); (R.G.); (A.L.)
- College of Osteopathic Medicine, Touro University Nevada, Henderson, NV 89014, USA
| | | | - Stefano Forte
- IOM Ricerca Srl, Viagrande, 95029 Catania, Italy; (E.M.); (G.R.); (R.G.); (A.L.)
| |
Collapse
|
5
|
Li W, Pan L, Hong W, Ginhoux F, Zhang X, Xiao C, Li X. A single-cell pan-cancer analysis to show the variability of tumor-infiltrating myeloid cells in immune checkpoint blockade. Nat Commun 2024; 15:6142. [PMID: 39034339 PMCID: PMC11271490 DOI: 10.1038/s41467-024-50478-8] [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: 12/03/2023] [Accepted: 07/12/2024] [Indexed: 07/23/2024] Open
Abstract
Myeloid cells are vital components of the immune system and have pivotal functions in orchestrating immune responses. Understanding their functions within the tumor microenvironment and their interactions with tumor-infiltrating lymphocytes presents formidable challenges across diverse cancer types, particularly with regards to cancer immunotherapies. Here, we explore tumor-infiltrating myeloid cells (TIMs) by conducting a pan-cancer analysis using single-cell transcriptomics across eight distinct cancer types, encompassing a total of 192 tumor samples from 129 patients. By examining gene expression patterns and transcriptional activities of TIMs in different cancer types, we discern notable alterations in abundance of TIMs and kinetic behaviors prior to and following immunotherapy. We also identify specific cell-cell interaction targets in immunotherapy; unique and shared regulatory profiles critical for treatment response; and TIMs associated with survival outcomes. Overall, our study illuminates the heterogeneity of TIMs and improves our understanding of tissue-specific and cancer-specific myeloid subsets within the context of tumor immunotherapies.
Collapse
Affiliation(s)
- Weiyuan Li
- School of Medicine, Yunnan University, Kunming, Yunnan, 650091, China
- Department of Reproductive Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan, 650032, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650031, China
| | - Lu Pan
- Institute of Environmental Medicine, Karolinska Institutet, Solna, 171 65, Sweden
| | - Weifeng Hong
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, 310005, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310005, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, 310005, China
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
- Institut Gustave Roussy, INSERM U1015, Bâtiment de Médecine Moléculaire 114 rue Edouard Vaillant, 94800, Villejuif, France
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xuan Zhang
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Chunjie Xiao
- School of Medicine, Yunnan University, Kunming, Yunnan, 650091, China.
| | - Xuexin Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, Liaoning, China.
- Institute of Health Sciences, China Medical University, Shenyang, 110122, Liaoning, China.
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, 171 65, Sweden.
| |
Collapse
|
6
|
Dharan R, Sorkin R. Tetraspanin proteins in membrane remodeling processes. J Cell Sci 2024; 137:jcs261532. [PMID: 39051897 DOI: 10.1242/jcs.261532] [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] [Indexed: 07/27/2024] Open
Abstract
Membrane remodeling is a fundamental cellular process that is crucial for physiological functions such as signaling, membrane fusion and cell migration. Tetraspanins (TSPANs) are transmembrane proteins of central importance to membrane remodeling events. During these events, TSPANs are known to interact with themselves and other proteins and lipids; however, their mechanism of action in controlling membrane dynamics is not fully understood. Since these proteins span the membrane, membrane properties such as rigidity, curvature and tension can influence their behavior. In this Review, we summarize recent studies that explore the roles of TSPANs in membrane remodeling processes and highlight the unique structural features of TSPANs that mediate their interactions and localization. Further, we emphasize the influence of membrane curvature on TSPAN distribution and membrane domain formation and describe how these behaviors affect cellular functions. This Review provides a comprehensive perspective on the multifaceted function of TSPANs in membrane remodeling processes and can help readers to understand the intricate molecular mechanisms that govern cellular membrane dynamics.
Collapse
Affiliation(s)
- Raviv Dharan
- School of Chemistry , Raymond & Beverly Sackler Faculty of Exact Sciences , Tel Aviv University, 6997801, Tel Aviv, Israel
- Center for Physics and Chemistry of Living Systems , Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Raya Sorkin
- School of Chemistry , Raymond & Beverly Sackler Faculty of Exact Sciences , Tel Aviv University, 6997801, Tel Aviv, Israel
- Center for Physics and Chemistry of Living Systems , Tel Aviv University, 6997801, Tel Aviv, Israel
| |
Collapse
|
7
|
Wang P, Pan Y, Zhang Y, Chen C, Hu J, Wang X. Role of interferon-induced transmembrane protein family in cancer progression: a special focus on pancreatic cancer. Med Oncol 2024; 41:85. [PMID: 38472606 DOI: 10.1007/s12032-024-02308-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 01/19/2024] [Indexed: 03/14/2024]
Abstract
Human interferon-induced transmembrane protein family (IFITMs) consists of five main proteins. IFITM1, IFITM2, and IFITM3 can be induced by interferon, while IFITM5 and IFITM10 are insensitive to interferon. IFITMs has various functions, including well-researched antiviral effects. As a molecule whose expression is significantly increased by interferon in the immune microenvironment, IFITMs has drawn growing interest in recent years for their role in the cancer progression. Unlike antiviral effects, the role and mechanism of IFITMs in cancer progression have not been clearly studied, especially the role and molecular mechanism of IFITMs in pancreatic cancer are rarely reported in the literature. This article focuses on the role and potential mechanism of IFITMs in pancreatic cancer progression by analyzing the function and mechanism of IFITM1-3 in other cancers and conducting bioinformatics analysis using the databases, so as to provide a new target for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Peipei Wang
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, 3-17 Renmin South Rd, Chengdu, 610041, Sichuan, China
| | - Yan Pan
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, 3-17 Renmin South Rd, Chengdu, 610041, Sichuan, China
| | - Yu Zhang
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, 3-17 Renmin South Rd, Chengdu, 610041, Sichuan, China
| | - Congliang Chen
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, 3-17 Renmin South Rd, Chengdu, 610041, Sichuan, China
| | - Junmei Hu
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, 3-17 Renmin South Rd, Chengdu, 610041, Sichuan, China
| | - Xia Wang
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, 3-17 Renmin South Rd, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
8
|
Shao S, Bu Z, Xiang J, Liu J, Tan R, Sun H, Hu Y, Wang Y. The role of Tetraspanins in digestive system tumor development: update and emerging evidence. Front Cell Dev Biol 2024; 12:1343894. [PMID: 38389703 PMCID: PMC10882080 DOI: 10.3389/fcell.2024.1343894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
Digestive system malignancies, including cancers of the esophagus, pancreas, stomach, liver, and colorectum, are the leading causes of cancer-related deaths worldwide due to their high morbidity and poor prognosis. The lack of effective early diagnosis methods is a significant factor contributing to the poor prognosis for these malignancies. Tetraspanins (Tspans) are a superfamily of 4-transmembrane proteins (TM4SF), classified as low-molecular-weight glycoproteins, with 33 Tspan family members identified in humans to date. They interact with other membrane proteins or TM4SF members to form a functional platform on the cytoplasmic membrane called Tspan-enriched microdomain and serve multiple functions including cell adhesion, migration, propagation and signal transduction. In this review, we summarize the various roles of Tspans in the progression of digestive system tumors and the underlying molecular mechanisms in recent years. Generally, the expression of CD9, CD151, Tspan1, Tspan5, Tspan8, Tspan12, Tspan15, and Tspan31 are upregulated, facilitating the migration and invasion of digestive system cancer cells. Conversely, Tspan7, CD82, CD63, Tspan7, and Tspan9 are downregulated, suppressing digestive system tumor cell metastasis. Furthermore, the connection between Tspans and the metastasis of malignant bone tumors is reviewed. We also summarize the potential role of Tspans as novel immunotherapy targets and as an approach to overcome drug resistance. Finally, we discuss the potential clinical value and therapeutic targets of Tspans in the treatments of digestive system malignancies and provide some guidance for future research.
Collapse
Affiliation(s)
- Shijie Shao
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Zhen Bu
- Department of General Surgery, Xinyi People's Hospital, Xinyi, China
| | - Jinghua Xiang
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jiachen Liu
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Rui Tan
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Han Sun
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yuanwen Hu
- Department of Gastroenterology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Yimin Wang
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
9
|
Zhao Q, Li F, Li J, Xia Y, Wang J, Chen L. An inflammatory response-related gene signature can predict the prognosis and impact the immune infiltration of multiple myeloma. Clin Exp Med 2024; 24:16. [PMID: 38280104 PMCID: PMC10821848 DOI: 10.1007/s10238-023-01277-w] [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: 11/06/2023] [Accepted: 11/25/2023] [Indexed: 01/29/2024]
Abstract
Multiple myeloma (MM) is a highly heterogeneous and incurable disease. Inflammation plays a vital role in cancer genesis and progression. However, the relationship between inflammatory response-related genes (IRRGs) and the prognosis of MM patients remains unknown. We constructed a IRRGs prognosis model by least absolute shrinkage and selection operator regression analysis. Moreover, clinical multivariate regression was performed to identify clinical implications. Gene set enrichment analysis was implemented to conduct its biological properties. CIBERSORT deconvolution algorithm was utilized to calculate the immune cell infiltration in different risk groups. The flow cytometry was utilized to perform protein expression of prognostic gene. A Six-IRRGs (VCAM1, RGS1, KIT, CD81, BLNK, and BIRC3) prognostic risk model was successfully constructed and validated. The risk model was an independent predictor for overall survival. Enrichment analysis revealed autophagy and PI3K-Akt signaling pathways were enriched in the high-risk group. Furthermore, we found CD81 widely impacted on the infiltration of immune cells, especially on monocytes and macrophages2. At last, the role of CD81 in MM was confirmed to be an adverse prognostic factor in clinical. Our study explores the potential application value of IRRGs in MM. These findings may provide new insights into the treatment for MM patients.
Collapse
Affiliation(s)
- Qian Zhao
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210003, China
- Department of Hematology, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, China
| | - Feng Li
- Department of Hematology, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, China
| | - Jing Li
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210003, China
| | - Yuan Xia
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210003, China
| | - Jing Wang
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210003, China
| | - Lijuan Chen
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210003, China.
| |
Collapse
|
10
|
Nie X, Wang D, Pan Y, Hua Y, Lü P, Yang Y. Discovery, classification and application of the CPISPR-Cas13 system. Technol Health Care 2024; 32:525-544. [PMID: 37545273 DOI: 10.3233/thc-230258] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
BACKGROUND The clustered regularly interspaced short palindromic repeats (CRISPR)-Cas system is an acquired immune system of bacteria and archaea. Continued research has resulted in the identification of other Cas13 proteins. OBJECTIVE This review briefly describes the discovery, classification, and application of the CRISPR-Cas13 system, including recent technological advances in addition to factors affecting system performance. METHODS Cas13-based molecular therapy of human, animal, and plant transcriptomes was discussed, including regulation of gene expression to combat pathogenic RNA viruses. In addition, the latest progress, potential shortcomings, and challenges of the CRISPR-Cas system for treatment of animal and plant diseases are reviewed. RESULTS The CRISPR-Cas system VI is characterized by two RNA-guided higher eukaryotes and prokaryotes nucleotide-binding domains. CRISPR RNA can cleave specific RNA through the interaction between the stem-loop rich chain of uracil residues and the Cas13a protein. The CRISPR-Cas13 system has been applied for gene editing in animal and plant cells, in addition to biological detection via accurate targeting of single-stranded RNA. CONCLUSION The CRISPR-Cas13 system offers a high-throughput and convenient technology for detection of viruses and potentially the development of anti-cancer drugs in the near future.
Collapse
Affiliation(s)
- Xiaojuan Nie
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Dandan Wang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ye Pan
- School of Experimental Animal Center, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ye Hua
- Institute of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Peng Lü
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yanhua Yang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
11
|
Abu-Saleh N, Kuo CC, Jiang W, Levy R, Levy S. The molecular mechanism of CD81 antibody inhibition of metastasis. Proc Natl Acad Sci U S A 2023; 120:e2305042120. [PMID: 37339209 PMCID: PMC10293848 DOI: 10.1073/pnas.2305042120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/17/2023] [Indexed: 06/22/2023] Open
Abstract
Metastases are reduced in CD81KO mice. In addition, a unique anti-CD81 antibody, 5A6, inhibits metastasis in vivo and invasion and migration in vitro. Here, we probed the structural components of CD81 required for the antimetastatic activity induced by 5A6. We found that the removal of either cholesterol or the intracellular domains of CD81 did not affect inhibition by the antibody. We show that the uniqueness of 5A6 is due not to increased affinity but rather to its recognition of a specific epitope on the large extracellular loop of CD81. Finally, we present a number of CD81 membrane-associated partners that may play a role in mediating the 5A6 antimetastatic attributes, including integrins and transferrin receptors.
Collapse
Affiliation(s)
- Niroz Abu-Saleh
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Chiung-Chi Kuo
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Wei Jiang
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Ronald Levy
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Shoshana Levy
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| |
Collapse
|
12
|
Bailly C, Thuru X. Targeting of Tetraspanin CD81 with Monoclonal Antibodies and Small Molecules to Combat Cancers and Viral Diseases. Cancers (Basel) 2023; 15:cancers15072186. [PMID: 37046846 PMCID: PMC10093296 DOI: 10.3390/cancers15072186] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/04/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023] Open
Abstract
Tetraspanin CD81 plays major roles in cell-cell interactions and the regulation of cellular trafficking. This cholesterol-embarking transmembrane protein is a co-receptor for several viruses, including HCV, HIV-1 and Chikungunya virus, which exploits the large extracellular loop EC2 for cell entry. CD81 is also an anticancer target implicated in cancer cell proliferation and mobility, and in tumor metastasis. CD81 signaling contributes to the development of solid tumors (notably colorectal, liver and gastric cancers) and has been implicated in the aggressivity of B-cell lymphomas. A variety of protein partners can interact with CD81, either to regulate attachment and uptake of viruses (HCV E2, claudin-1, IFIM1) or to contribute to tumor growth and dissemination (CD19, CD44, EWI-2). CD81-protein interactions can be modulated with molecules targeting the extracellular domain of CD81, investigated as antiviral and/or anticancer agents. Several monoclonal antibodies anti-CD81 have been developed, notably mAb 5A6 active against invasion and metastasis of triple-negative breast cancer cells. CD81-EC2 can also be targeted with natural products (trachelogenin and harzianoic acids A-B) and synthetic compounds (such as benzothiazole-quinoline derivatives). They are weak CD81 binders but offer templates for the design of new compounds targeting the open EC2 loop. There is no anti-CD81 compound in clinical development at present, but this structurally well-characterized tetraspanin warrants more substantial considerations as a drug target.
Collapse
Affiliation(s)
- Christian Bailly
- OncoWitan, Scientific Consulting Office, F-59290 Lille, France
- Institut de Chimie Pharmaceutique Albert Lespagnol (ICPAL), Faculty of Pharmacy, University of Lille, F-59006 Lille, France
- CNRS, Inserm, CHU Lille, UMR9020-U1277-Canther-Cancer Heterogeneity Plasticity and Resistance to Therapies, OncoLille Institut, University of Lille, F-59000 Lille, France
| | - Xavier Thuru
- CNRS, Inserm, CHU Lille, UMR9020-U1277-Canther-Cancer Heterogeneity Plasticity and Resistance to Therapies, OncoLille Institut, University of Lille, F-59000 Lille, France
| |
Collapse
|
13
|
Anaganti N, Chattopadhyay A, Di Filippo M, Hussain MM. New CRISPR Technology for Creating Cell Models of Lipoprotein Assembly and Secretion. Curr Atheroscler Rep 2023; 25:209-217. [PMID: 36913170 DOI: 10.1007/s11883-023-01095-1] [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] [Accepted: 02/22/2023] [Indexed: 03/14/2023]
Abstract
PURPOSE OF REVIEW This review is aimed at providing an overview of new developments in gene editing technology, including examples of how this technology has been used to develop cell models for studying the effects of gene ablation or missense mutations on lipoprotein assembly and secretion. RECENT FINDINGS CRISPR/Cas9-mediated gene editing is superior to other technologies because of its ease, sensitivity, and low off-target effects. This technology has been used to study the importance of microsomal triglyceride transfer protein in the assembly and secretion of apolipoprotein B-containing lipoproteins, as well as to establish causal effects of APOB gene missense mutations on lipoprotein assembly and secretion. CRISPR/Cas9 technology is anticipated to provide unprecedented flexibility in studying protein structure and function in cells and animals and to yield mechanistic insights into variants in the human genome.
Collapse
Affiliation(s)
- Narasimha Anaganti
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY, 11501, USA
| | - Atrayee Chattopadhyay
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY, 11501, USA
| | - Mathilde Di Filippo
- Department of Biochemistry and Molecular Biology, Hospices Civils de Lyon, Lyon, France
| | - M Mahmood Hussain
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY, 11501, USA.
- VA New York Harbor Healthcare System, Brooklyn, NY, 11209, USA.
| |
Collapse
|
14
|
Friedlová N, Zavadil Kokáš F, Hupp TR, Vojtěšek B, Nekulová M. IFITM protein regulation and functions: Far beyond the fight against viruses. Front Immunol 2022; 13:1042368. [PMID: 36466909 PMCID: PMC9716219 DOI: 10.3389/fimmu.2022.1042368] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/27/2022] [Indexed: 07/30/2023] Open
Abstract
Interferons (IFNs) are important cytokines that regulate immune responses through the activation of hundreds of genes, including interferon-induced transmembrane proteins (IFITMs). This evolutionarily conserved protein family includes five functionally active homologs in humans. Despite the high sequence homology, IFITMs vary in expression, subcellular localization and function. The initially described adhesive and antiproliferative or pro-oncogenic functions of IFITM proteins were diluted by the discovery of their antiviral properties. The large set of viruses that is inhibited by these proteins is constantly expanding, as are the possible mechanisms of action. In addition to their beneficial antiviral effects, IFITM proteins are often upregulated in a broad spectrum of cancers. IFITM proteins have been linked to most hallmarks of cancer, including tumor cell proliferation, therapeutic resistance, angiogenesis, invasion, and metastasis. Recent studies have described the involvement of IFITM proteins in antitumor immunity. This review summarizes various levels of IFITM protein regulation and the physiological and pathological functions of these proteins, with an emphasis on tumorigenesis and antitumor immunity.
Collapse
Affiliation(s)
- Nela Friedlová
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Filip Zavadil Kokáš
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Ted R. Hupp
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Bořivoj Vojtěšek
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Marta Nekulová
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| |
Collapse
|
15
|
Gonzalez-Salinas F, Martinez-Amador C, Trevino V. Characterizing genes associated with cancer using the CRISPR/Cas9 system: A systematic review of genes and methodological approaches. Gene 2022; 833:146595. [PMID: 35598687 DOI: 10.1016/j.gene.2022.146595] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/22/2022] [Accepted: 05/16/2022] [Indexed: 12/24/2022]
Abstract
The CRISPR/Cas9 system enables a versatile set of genomes editing and genetic-based disease modeling tools due to its high specificity, efficiency, and accessible design and implementation. In cancer, the CRISPR/Cas9 system has been used to characterize genes and explore different mechanisms implicated in tumorigenesis. Different experimental strategies have been proposed in recent years, showing dependency on various intrinsic factors such as cancer type, gene function, mutation type, and technical approaches such as cell line, Cas9 expression, and transfection options. However, the successful methodological approaches, genes, and other experimental factors have not been analyzed. We, therefore, initially considered more than 1,300 research articles related to CRISPR/Cas9 in cancer to finally examine more than 400 full-text research publications. We summarize findings regarding target genes, RNA guide designs, cloning, Cas9 delivery systems, cell enrichment, and experimental validations. This analysis provides valuable information and guidance for future cancer gene validation experiments.
Collapse
Affiliation(s)
- Fernando Gonzalez-Salinas
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Morones Prieto avenue 3000, Monterrey, Nuevo Leon 64710, Mexico
| | - Claudia Martinez-Amador
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Morones Prieto avenue 3000, Monterrey, Nuevo Leon 64710, Mexico
| | - Victor Trevino
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Morones Prieto avenue 3000, Monterrey, Nuevo Leon 64710, Mexico; Tecnologico de Monterrey, The Institute for Obesity Research, Eugenio Garza Sada avenue 2501, Monterrey, Nuevo Leon 64849, México.
| |
Collapse
|
16
|
Vences-Catalán F, Rajapaksa R, Kuo CC, Miller CL, Lee A, Ramani VC, Jeffrey SS, Levy R, Levy S. Targeting the tetraspanin CD81 reduces cancer invasion and metastasis. Proc Natl Acad Sci U S A 2021; 118:e2018961118. [PMID: 34099563 PMCID: PMC8214710 DOI: 10.1073/pnas.2018961118] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Tetraspanins are an evolutionary conserved family of proteins involved in multiple aspects of cell physiology, including proliferation, migration and invasion, protein trafficking, and signal transduction; yet their detailed mechanism of action is unknown. Tetraspanins have no known natural ligands, but their engagement by antibodies has begun to reveal their role in cell biology. Studies of tetraspanin knockout mice and of germline mutations in humans have highlighted their role under normal and pathological conditions. Previously, we have shown that mice deficient in the tetraspanin CD81 developed fewer breast cancer metastases compared to their wild-type (WT) counterparts. Here, we show that a unique anti-human CD81 antibody (5A6) effectively halts invasion of triple-negative breast cancer (TNBC) cell lines. We demonstrate that 5A6 induces CD81 clustering at the cell membrane and we implicate JAM-A protein in the ability of this antibody to inhibit tumor cell invasion and migration. Furthermore, in a series of in vivo studies we demonstrate that this antibody inhibits metastases in xenograft models, as well as in syngeneic mice bearing a mouse tumor into which we knocked in the human CD81 epitope recognized by the 5A6 antibody.
Collapse
Affiliation(s)
- Felipe Vences-Catalán
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Ranjani Rajapaksa
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Chiung-Chi Kuo
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Caitlyn L Miller
- Department of Bioengineering, Stanford University, Stanford, CA 94305
| | - Anderson Lee
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Vishnu C Ramani
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Stefanie S Jeffrey
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Ronald Levy
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305; s
| | - Shoshana Levy
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305; s
| |
Collapse
|
17
|
Assessment of TSPAN Expression Profile and Their Role in the VSCC Prognosis. Int J Mol Sci 2021; 22:ijms22095015. [PMID: 34065085 PMCID: PMC8125994 DOI: 10.3390/ijms22095015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/25/2021] [Accepted: 05/07/2021] [Indexed: 01/16/2023] Open
Abstract
The role and prognostic value of tetraspanins (TSPANs) in vulvar squamous cell carcinoma (VSCC) remain poorly understood. We sought to primarily determine, at both the molecular and tissue level, the expression profile of the TSPANs CD9, CD63, CD81, and CD82 in archived VSCC samples (n = 117) and further investigate their clinical relevance as prognostic markers. Our studies led us to identify CD63 as the most highly expressed TSPAN, at the gene and protein levels. Multicomparison studies also revealed that the expression of CD9 was associated with tumor size, whereas CD63 upregulation was associated with histological diagnosis and vascular invasion. Moreover, low expression of CD81 and CD82 was associated with worse prognosis. To determine the role of TSPANs in VSCC at the cellular level, we assessed the mRNA levels of CD63 and CD82 in established metastatic (SW962) and non-metastatic (SW954) VSCC human cell lines. CD82 was found to be downregulated in SW962 cells, thus supporting its metastasis suppressor role. However, CD63 was significantly upregulated in both cell lines. Silencing of CD63 by siRNA led to a significant decrease in proliferation of both SW954 and SW962. Furthermore, in SW962 particularly, CD63-siRNA also remarkably inhibited cell migration. Altogether, our data suggest that the differential expression of TSPANs represents an important feature for prognosis of VSCC patients and indicates that CD63 and CD82 are likely potential therapeutic targets in VSCC.
Collapse
|
18
|
Wang S, Wei H, Huang Z, Wang X, Shen R, Wu Z, Lin J. Epidermal growth factor receptor promotes tumor progression and contributes to gemcitabine resistance in osteosarcoma. Acta Biochim Biophys Sin (Shanghai) 2021; 53:317-324. [PMID: 33432347 DOI: 10.1093/abbs/gmaa177] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Indexed: 12/18/2022] Open
Abstract
Osteosarcoma (OS) is the most common type of primary malignant tumors that originate in the bone. Resistance to chemotherapy confers a poor prognosis on OS patients. Dysregulation of the epidermal growth factor receptor (EGFR) signaling has been reported in sarcomas. However, the functional contribution of EGFR hyperactivation to the tumor biology and chemoresistance remains largely unexplored in OS. In this study, we aimed to investigate the role of EGFR in OS progression and in the response of OS to gemcitabine treatment. The EGFR expression was found to be upregulated in fibroblastic OS cell lines. EGFR knockdown suppressed OS cell proliferation, migration, and invasion in vitro and tumor formation in vivo. Conversely, EGFR overexpression promoted the growth and motility of OS cells. In terms of mechanism, the levels of phospho-Akt and phospho-ERK were decreased upon EGFR knockdown but increased as a result of EGFR overexpression, implying a possible involvement of PI3K/Akt and ERK pathways in mediating the effects of EGFR on OS cells. Moreover, the level of phospho-EGFR was increased in OS cells when exposed to gemcitabine treatment. A more profound proliferative inhibition and a higher rate of apoptosis were obtained in OS cells via inducing cell cycle arrest at G1 phase upon gemcitabine treatment combined with EGFR knockdown, as compared to gemcitabine alone. On the contrary, EGFR overexpression counteracted the growth-inhibiting and pro-apoptotic effects of gemcitabine in OS cells. The present study suggests that EGFR promotes tumor progression and contributes to gemcitabine resistance in OS.
Collapse
Affiliation(s)
- Shenglin Wang
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Hongxiang Wei
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Zhen Huang
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Xinwen Wang
- Department of Orthopedics, The People’s Hospital of Jiangmen City, Jiangmen 529051, China
| | - Rongkai Shen
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Zhaoyang Wu
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Jianhua Lin
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| |
Collapse
|
19
|
Liu W, Wang S, Lin B, Zhang W, Ji G. Applications of CRISPR/Cas9 in the research of malignant musculoskeletal tumors. BMC Musculoskelet Disord 2021; 22:149. [PMID: 33546657 PMCID: PMC7866880 DOI: 10.1186/s12891-021-04020-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 01/26/2021] [Indexed: 12/05/2022] Open
Abstract
Background Malignant tumors of the musculoskeletal system, especially osteosarcoma, Ewing sarcoma and rhabdomyosarcoma, pose a major threat to the lives and health of adolescents and children. Current treatments for musculoskeletal tumors mainly include surgery, chemotherapy, and radiotherapy. The problems of chemotherapy resistance, poor long-term outcome of radiotherapy, and the inherent toxicity and side effects of chemical drugs make it extremely urgent to seek new treatment strategies. Main text As a potent gene editing tool, the rapid development of CRISPR/Cas9 technology in recent years has prompted scientists to apply it to the study of musculoskeletal tumors. This review summarizes the application of CRISPR/Cas9 technology for the treatment of malignant musculoskeletal tumors, focusing on its essential role in the field of basic research. Conclusion CRISPR, has demonstrated strong efficacy in targeting tumor-related genes, and its future application in the clinical treatment of musculoskeletal tumors is promising.
Collapse
Affiliation(s)
- Wei Liu
- Department of Orthopaedics, Xiang'an Hospital, School of Medicine, Xiamen University, No. 2000 East Xiang'an Road, Xiang'an District, Xiamen, 361102, China
| | - Shubin Wang
- Department of Orthopaedics, Xiang'an Hospital, School of Medicine, Xiamen University, No. 2000 East Xiang'an Road, Xiang'an District, Xiamen, 361102, China
| | - Binhui Lin
- Department of Orthopaedics, Xiang'an Hospital, School of Medicine, Xiamen University, No. 2000 East Xiang'an Road, Xiang'an District, Xiamen, 361102, China
| | - Wei Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Guangrong Ji
- Department of Orthopaedics, Xiang'an Hospital, School of Medicine, Xiamen University, No. 2000 East Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| |
Collapse
|
20
|
Mao X, Yang X, Chen X, Yu S, Yu S, Zhang B, Ji Y, Chen Y, Ouyang Y, Luo W. Single-cell transcriptome analysis revealed the heterogeneity and microenvironment of gastrointestinal stromal tumors. Cancer Sci 2021; 112:1262-1274. [PMID: 33393143 PMCID: PMC7935798 DOI: 10.1111/cas.14795] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/15/2020] [Accepted: 12/31/2020] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal stromal tumor (GIST) is the most common mesenchymal tumor of the human gastrointestinal tract. In this study, we performed single-cell RNA sequencing (RNA-seq) on intra- and peri-tumor tissues from GIST patients with the aim of discovering the heterogeneity of tumor cells in GIST and their interactions with other cells. We found four predominating cell types in GIST tumor tissue, including T cells, macrophages, tumor cells, and NK cells. Tumor cells could be clustered into two groups: one was highly proliferating and associated with high risk of metastasis, the other seemed "resting" and associated with low risk. Their clinical relevance and prognostic values were confirmed by RNA-seq of 65 GIST samples. T cells were the largest cell type in our single-cell data. Two groups of CD8+ effector memory (EM) cells were in the highest clonal expansion and performed the highest cytotoxicity but were also the most exhausted among all T cells. A group of macrophages were found polarized to possess both M1 and M2 signatures, and increased along with tumor progression. Cell-to-cell interaction analysis revealed that adipose endothelial cells had high interactions with tumor cells to facilitate their progression. Macrophages were at the center of the tumor microenvironment, recruiting immune cells to the tumor site and having most interactions with both tumor and nontumor cells. In conclusion, we obtained an overview of the GIST microenvironment and revealed the heterogeneity of each cell type and their relevance to risk classifications, which provided a novel theoretical basis for learning and curing GISTs.
Collapse
Affiliation(s)
- Xiaofan Mao
- Clinical Research Institute, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China.,Medical Engineering Technology Research and Development Center of Immune Repertoire in Foshan, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China
| | - Xuezhu Yang
- Gastroenterology, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China
| | - Xiangping Chen
- Clinical Research Institute, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China.,Medical Engineering Technology Research and Development Center of Immune Repertoire in Foshan, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China
| | - Sifei Yu
- Clinical Research Institute, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China.,Medical Engineering Technology Research and Development Center of Immune Repertoire in Foshan, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China
| | - Si Yu
- Gastroenterology, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China
| | - Beiying Zhang
- Clinical Research Institute, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China.,Medical Engineering Technology Research and Development Center of Immune Repertoire in Foshan, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China
| | - Yong Ji
- Gastroenterology, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China
| | - Yihao Chen
- Clinical Research Institute, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China.,Medical Engineering Technology Research and Development Center of Immune Repertoire in Foshan, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China
| | - Ying Ouyang
- Clinical Research Institute, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China.,Medical Engineering Technology Research and Development Center of Immune Repertoire in Foshan, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China
| | - Wei Luo
- Clinical Research Institute, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China.,Medical Engineering Technology Research and Development Center of Immune Repertoire in Foshan, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China
| |
Collapse
|
21
|
Miao Y, Liu G, Liu L. Histone methyltransferase SUV39H2 regulates LSD1-dependent CDH1 expression and promotes epithelial mesenchymal transition of osteosarcoma. Cancer Cell Int 2021; 21:2. [PMID: 33397384 PMCID: PMC7784292 DOI: 10.1186/s12935-020-01636-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 11/02/2020] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Osteosarcoma (OS) is a malignant tumor characterized by the direct production of bone or osteoid tissues by proliferating tumor cells. Suppressor of variegation 3-9 homolog 2 (SUV39H2) is implicated in the occurrence of OS. Therefore, we designed this study to investigate effects of SUV39H2 in OS meditated by the lysine specific demethylase-1/E-cadherin (LSD1/CDH1) axis. METHODS Clinical OS tissues and paracancerous tissues were collected for analysis of SUV39H2, LSD1 and CDH1 expression, and Kaplan-Meier survival analysis was applied to test the relationship between SUV39H2 expression and overall survival. Loss- and gain-of-function assays were conducted to determine the roles of SUV39H2, LSD1 and CDH1 in OS epithelial mesenchymal transition (EMT) and migration in OS cells, with quantitation of relevant proteins by immunofluorescence. We confirmed the effects of modulating the SUV39H2/CDH1 axis in a mouse OS tumor model. RESULTS SUV39H2 and LSD1 were highly expressed, while CDH1 was downregulated in OS tissues and cells. SUV39H2 expression correlated inversely with overall survival of patients with OS. SUV39H2 positively regulated LSD1 expression, while LSD1 negatively regulated CDH1 expression. SUV39H2 or LSD1 overexpression, or CDH1 silencing promoted migration and EMT, as indicated by reduced E-cadherin and dramatically upregulated Vimentin and N-cadherin of OS cells. SUV39H2 expedited the progression of OS, which was reversed by CDH1 repression in the setting of OS in vitro and in vivo. CONCLUSIONS Collectively, our results demonstrate highly expressed SUV39H2 in OS elevates the expression of LSD1 to downregulate CDH1 expression, thereby aggravating OS, providing a potential therapeutic target for treatment of OS.
Collapse
Affiliation(s)
- Yingying Miao
- Department of Anesthesiology, Union Hospital of Jilin University, Changchun, 130033 People’s Republic of China
| | - Guifeng Liu
- Department of Anesthesiology, Union Hospital of Jilin University, Changchun, 130033 People’s Republic of China
- Department of Radiology, Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun, 130033 Jilin People’s Republic of China
| | - Lin Liu
- Department of Anesthesiology, Union Hospital of Jilin University, Changchun, 130033 People’s Republic of China
- Department of Radiology, Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun, 130033 Jilin People’s Republic of China
| |
Collapse
|
22
|
Liliac IM, Popescu EL, Văduva IA, Pirici D, Mogoşanu GD, Streba CT, Busuioc CJ, Bejenaru LE, Bejenaru C, Crăciunoiu N, Dumitru I, Elayan H, Mogoantă L. Nanoparticle-functionalized dressings for the treatment of third-degree skin burns - histopathological and immunohistochemical study. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY = REVUE ROUMAINE DE MORPHOLOGIE ET EMBRYOLOGIE 2021; 62:159-168. [PMID: 34609418 PMCID: PMC8597381 DOI: 10.47162/rjme.62.1.15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 09/27/2021] [Indexed: 02/05/2023]
Abstract
Skin burns are one of the most common injuries associated with increased morbidity and mortality, especially in the children and the elderlies. Severe burns, especially, result in a systemic immune and inflammatory response, which may reflect in multiple organ insufficiency, and a fast and effective local restorative process is essential for functionality recovering, as well as for interrupting the generalized systemic response. We have aimed here to assess the effect of different wound dressings in what it regards the morphology and clinical restoration after a skin burn. On a rat animal model, we have evaluated the macroscopic and histopathological features of controlled third degree skin burns in control animals versus treatments with local dressings of silver sulfadiazine (SDA) cream, simple gel (G), gel + silver nanoparticles (AgNPs) (G+NPS), gel + exosomes (G+EXO) and gel + AgNPs + exosomes (Gel+NPS+EXO), at 14 days and, respectively, 21 days after the lesion. Tissue fragments were harvested and processed for histopathology and immunohistochemistry. Immunofluorescence was utilized to evaluate the maturity of underlaying granulation tissue based on double stainings for smooth muscle actin (SMA) and cluster of differentiation 31 (CD31). Our study showed variability in what it regards the vessel density and immunoexpression of SMA between the treatments, and image analysis revealed that most SMA reduction and blood vessel density reduction in the maturing granulation tissue occurred for the G+NPS and G+NPS+EXO treatments. A complete re-epithelization was also observed for the G+NPS+EXO treatment. Overall, our results show that improved topic treatments promote faster re-epithelization and reparation of the dermis after skin burn lesions, providing thus an avenue for new treatments that aim both local recuperation and systemic infection prevention.
Collapse
|
23
|
Immunotherapy for osteosarcoma: Fundamental mechanism, rationale, and recent breakthroughs. Cancer Lett 2020; 500:1-10. [PMID: 33359211 DOI: 10.1016/j.canlet.2020.12.024] [Citation(s) in RCA: 297] [Impact Index Per Article: 59.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 02/06/2023]
Abstract
Osteosarcoma (OS) is the most common primary malignancy of the bone and has a high propensity for local invasion and metastasis. Although combining surgery with chemotherapy has immensely improved the outcomes of osteosarcoma patients, the prognosis of metastatic or recurrent osteosarcomas is still unsatisfactory. Immunotherapy has proven to be a promising therapeutic strategy against human malignancies and improved understanding of the immune response to OS, and biomarker development has increased the number of patients who benefit from immunotherapies in recent years. Here, we review recent advances in immunotherapy in osteosarcoma and discuss the mechanisms and status of immunotherapies in both preclinical and clinical trials as well as future therapies on the horizon. These advances may pave the way for novel treatments requisite for patients with osteosarcoma in need of new therapies.
Collapse
|
24
|
Tumor cell MT1-MMP is dispensable for osteosarcoma tumor growth, bone degradation and lung metastasis. Sci Rep 2020; 10:19138. [PMID: 33154487 PMCID: PMC7645741 DOI: 10.1038/s41598-020-75995-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 10/16/2020] [Indexed: 02/03/2023] Open
Abstract
The membrane-anchored matrix metalloprotease MT1-MMP is a potent collagenolytic enzyme with a well-established role in extracellular matrix turnover and cellular invasion into collagen-rich tissues. MT1-MMP is highly expressed in various types of cancer and has been demonstrated to be directly involved in several stages of tumor progression, including primary tumor growth, angiogenesis, invasion and metastasis. Osteosarcoma is the most common type of primary bone cancer. This disease is characterized by invasive tumor growth, leading to extensive bone destruction, and metastasis to the lungs. The tumor cells in human osteosarcoma display a strong expression of MT1-MMP, but the role of MT1-MMP in osteosarcoma progression is currently unknown. In this study, we investigated the role of MT1-MMP during various stages of osteosarcoma development. We utilized an optimized orthotopic murine osteosarcoma model and human osteosarcoma cells in which the MT1-MMP gene was knocked out using CRISPR/Cas9. We observed a strong expression of MT1-MMP in wildtype cells of both primary tumors and lung metastases, but, surprisingly, MT1-MMP deficiency did not affect primary tumor growth, bone degradation or the formation and growth of lung metastases. We therefore propose that, unlike findings reported in other cancers, tumor-expressed MT1-MMP is dispensable for all stages of osteosarcoma progression.
Collapse
|
25
|
Guan X, Xu Y, Zheng J. Long non‑coding RNA PCAT6 promotes the development of osteosarcoma by increasing MDM2 expression. Oncol Rep 2020; 44:2465-2474. [PMID: 33125146 PMCID: PMC7610325 DOI: 10.3892/or.2020.7813] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022] Open
Abstract
Osteosarcoma is a severe malignant tumor. Several studies indicated that lncRNA prostate cancer-associated transcript 6 (PCAT6) promoted the development of multiple types of cancers. Studies have also revealed that MDM2 could aggravate tumor symptoms inhibiting P53 expression. However, whether lncRNA PCAT6 could affect the proliferation and metastasis of osteosarcoma cells by regulating P53 expression is unclear. The present study established lncRNA PCAT6-overexpressing osteosarcoma cells. Cell Counting Kit-8, wound healing and Transwell assays were performed to detect the change in proliferation, migration and invasion of these cells, respectively. Subsequently, E3 ubiquitin-protein ligase Mdm2 (MDM2), P53 and P21 expression were determined using western blotting. Finally, MDM2 expression was inhibited and the proliferation, migration and invasion of these cells was determined again. The present study found that the proliferation, migration and invasion of osteosarcoma cells increased following overexpression of lncRNA PCAT6. MDM2 expression was upregulated while the levels of P53 and P21 decreased following overexpression of lncRNA PCAT6. However, the proliferation, migration and invasion of osteosarcoma cells were inhibited following MDM2 knockdown. Additionally, P53 and P21 was rescued following MDM2 knockdown. To conclude, lncRNA PCAT6 promoted the proliferation, migration and invasion of osteosarcoma cells by promoting the expression of MDM2 and suppressing the expression of P53 and P21.
Collapse
Affiliation(s)
- Xiliang Guan
- Department of Orthopaedic Surgery, People's Hospital of Rizhao, Rizhao, Shandong 276826, P.R. China
| | - Yufen Xu
- Department of Oncology, The First Hospital of Jiaxing and The Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314001, P.R. China
| | - Jufen Zheng
- The Department of Bone, Zhejiang Hospital, Hangzhou, Zhejiang 310030, P.R. China
| |
Collapse
|