1
|
Fernández-Nogueira P, Linzoain-Agos P, Cueto-Remacha M, De la Guia-Lopez I, Recalde-Percaz L, Parcerisas A, Gascon P, Carbó N, Gutierrez-Uzquiza A, Fuster G, Bragado P. Role of semaphorins, neuropilins and plexins in cancer progression. Cancer Lett 2024; 606:217308. [PMID: 39490515 DOI: 10.1016/j.canlet.2024.217308] [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: 08/02/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Progress in understanding nervous system-cancer interconnections has emphasized the functional role of semaphorins (SEMAs) and their receptors, neuropilins (NRPs) and plexins (PLXNs), in cancer progression. SEMAs are a conserved and extensive family of broadly expressed soluble and membrane-associated proteins that were first described as regulators of axon guidance and neural and vascular development. However, recent advances have shown that they can have a dual role in cancer progression, acting either as tumor promoters or suppressors. SEMAs effects result from their interaction with specific co-receptors/receptors NRPs/PLXNs, that have also been described to play a role in cancer progression. They can influence both cancer cells and tumor microenvironment components modulating various aspects of tumorigenesis such as oncogenesis, tumor growth, invasion and metastatic spread or treatment resistance. In this review we focus on the role of these axon guidance signals and their receptors and co-receptors in various aspects of cancer. Furthermore, we also highlight their potential application as novel approaches for cancer treatment in the future.
Collapse
Affiliation(s)
- P Fernández-Nogueira
- Department of Biomedicine, School of Medicine, Universitat de Barcelona, 08028, Barcelona, Spain; Biosciences Department, Faculty of Sciences, Technology and Engineering, University of Vic. Central University of Catalonia (UVic-UCC), 08500, Vic, Catalonia, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Institute of Biomedicine of the Universitat de Barcelona (IBUB), 08028, Barcelona, Spain
| | - P Linzoain-Agos
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040, Madrid, Spain; Health Research Institute of the Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - M Cueto-Remacha
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040, Madrid, Spain; Health Research Institute of the Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - I De la Guia-Lopez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040, Madrid, Spain; Health Research Institute of the Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - L Recalde-Percaz
- Department of Biomedicine, School of Medicine, Universitat de Barcelona, 08028, Barcelona, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Institute of Biomedicine of the Universitat de Barcelona (IBUB), 08028, Barcelona, Spain
| | - A Parcerisas
- Biosciences Department, Faculty of Sciences, Technology and Engineering, University of Vic. Central University of Catalonia (UVic-UCC), 08500, Vic, Catalonia, Spain; Tissue Repair and Regeneration Laboratory (TR2Lab), Institute of Research and Innovation of Life Sciences and Health, Catalunya Central (IRIS-CC), 08500, Vic, Catalonia, Spain
| | - P Gascon
- Department of Biomedicine, School of Medicine, Universitat de Barcelona, 08028, Barcelona, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Institute of Biomedicine of the Universitat de Barcelona (IBUB), 08028, Barcelona, Spain
| | - N Carbó
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Institute of Biomedicine of the Universitat de Barcelona (IBUB), 08028, Barcelona, Spain
| | - A Gutierrez-Uzquiza
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040, Madrid, Spain; Health Research Institute of the Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - G Fuster
- Department of Biomedicine, School of Medicine, Universitat de Barcelona, 08028, Barcelona, Spain; Biosciences Department, Faculty of Sciences, Technology and Engineering, University of Vic. Central University of Catalonia (UVic-UCC), 08500, Vic, Catalonia, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Institute of Biomedicine of the Universitat de Barcelona (IBUB), 08028, Barcelona, Spain; Tissue Repair and Regeneration Laboratory (TR2Lab), Institute of Research and Innovation of Life Sciences and Health, Catalunya Central (IRIS-CC), 08500, Vic, Catalonia, Spain.
| | - P Bragado
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040, Madrid, Spain; Health Research Institute of the Hospital Clínico San Carlos, 28040, Madrid, Spain.
| |
Collapse
|
2
|
Bica C, Tirpe A, Nutu A, Ciocan C, Chira S, Gurzau ES, Braicu C, Berindan-Neagoe I. Emerging roles and mechanisms of semaphorins activity in cancer. Life Sci 2023; 318:121499. [PMID: 36775114 DOI: 10.1016/j.lfs.2023.121499] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/08/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023]
Abstract
Semaphorins are regulatory molecules that are linked to the modulation of several cancer processes, such as angiogenesis, cancer cell invasiveness and metastasis, tumor growth, as well as cancer cell survival. Semaphorin (SEMA) activity depends on the cancer histotypes and their particularities. In broad terms, the effects of SEMAs result from their interaction with specific receptors/co-receptors - Plexins, Neuropilins and Integrins - and the subsequent effects upon the downstream effectors (e.g. PI3K/AKT, MAPK/ERK). The present article serves as an integrative review work, discussing the broad implications of semaphorins in cancer, focusing on cell proliferation/survival, angiogenesis, invasion, metastasis, stemness, and chemo-resistance/response whilst highlighting their heterogeneity as a family. Herein, we emphasized that semaphorins are largely implicated in cancer progression, interacting with the tumor microenvironment components. Whilst some SEMAs (e.g. SEMA3A, SEMA3B) function widely as tumor suppressors, others (e.g. SEMA3C) act as pro-tumor semaphorins. The differences observed in terms of the biological structure of SEMAs and the particularities of each cancer histotypes require that each semaphorin be viewed as a unique entity, and its roles must be researched accordingly. A more in-depth and comprehensive view of the molecular mechanisms that promote and sustain the malignant behavior of cancer cells is of utmost importance.
Collapse
Affiliation(s)
- Cecilia Bica
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania.
| | - Alexandru Tirpe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania; Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania.
| | - Andreea Nutu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania.
| | - Cristina Ciocan
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania.
| | - Sergiu Chira
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania.
| | - Eugen S Gurzau
- Cluj School of Public Health, College of Political, Administrative and Communication Sciences, Babes-Bolyai University, 7 Pandurilor Street, Cluj-Napoca, Romania; Environmental Health Center, 58 Busuiocului Street, 400240 Cluj-Napoca, Romania.
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania.
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania.
| |
Collapse
|
3
|
Zuo Q, Yang Y, Lyu Y, Yang C, Chen C, Salman S, Huang TYT, Wicks EE, Jackson W, Datan E, Qin W, Semenza GL. Plexin-B3 expression stimulates MET signaling, breast cancer stem cell specification, and lung metastasis. Cell Rep 2023; 42:112164. [PMID: 36857181 DOI: 10.1016/j.celrep.2023.112164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 12/21/2022] [Accepted: 02/09/2023] [Indexed: 03/02/2023] Open
Abstract
Intratumoral hypoxia is a microenvironmental feature that promotes breast cancer progression and is associated with cancer mortality. Plexin B3 (PLXNB3) is highly expressed in estrogen receptor-negative breast cancer, but the underlying mechanisms and consequences have not been thoroughly investigated. Here, we report that PLXNB3 expression is increased in response to hypoxia and that PLXNB3 is a direct target gene of hypoxia-inducible factor 1 (HIF-1) in human breast cancer cells. PLXNB3 expression is correlated with HIF-1α immunohistochemistry, breast cancer grade and stage, and patient mortality. Mechanistically, PLXNB3 is required for hypoxia-induced MET/SRC/focal adhesion kinase (FAK) and MET/SRC/STAT3/NANOG signaling as well as hypoxia-induced breast cancer cell migration, invasion, and cancer stem cell specification. PLXNB3 knockdown impairs tumor formation and lung metastasis in orthotopic breast cancer mouse models.
Collapse
Affiliation(s)
- Qiaozhu Zuo
- Armstrong Oxygen Biology Research Center and Vascular Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Yongkang Yang
- Armstrong Oxygen Biology Research Center and Vascular Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21205, USA
| | - Yajing Lyu
- Armstrong Oxygen Biology Research Center and Vascular Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chen Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Chelsey Chen
- Armstrong Oxygen Biology Research Center and Vascular Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shaima Salman
- Armstrong Oxygen Biology Research Center and Vascular Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tina Yi-Ting Huang
- Armstrong Oxygen Biology Research Center and Vascular Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elizabeth E Wicks
- Armstrong Oxygen Biology Research Center and Vascular Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Walter Jackson
- Armstrong Oxygen Biology Research Center and Vascular Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Emmanuel Datan
- Armstrong Oxygen Biology Research Center and Vascular Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Wenxin Qin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Gregg L Semenza
- Armstrong Oxygen Biology Research Center and Vascular Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21205, USA; Departments of Biological Chemistry, Medicine, Pediatrics, and Radiation Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
4
|
Hu J, Chen W, Shen L, Chen Z, Huang J. Crosstalk between the peripheral nervous system and breast cancer influences tumor progression. Biochim Biophys Acta Rev Cancer 2022; 1877:188828. [PMID: 36283598 DOI: 10.1016/j.bbcan.2022.188828] [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: 09/06/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/18/2022]
Abstract
Recent studies have shown that peripheral nerves play an important role in the progression of breast cancer. Breast cancer cells (BCCs) promote local peripheral nerve growth and branching by secreting neuroactive molecules, including neurotrophins and axon guidance molecules (AGMs). Sympathetic nerves promote breast cancer progression, while parasympathetic and sensory nerves mainly have anti-tumor effects in the progression of breast cancer. Specifically, peripheral nerves can influence the progression of breast cancer by secreting neurotransmitters not only directly binding to the corresponding receptors of BCCs, but also indirectly acting on immune cells to modulate anti-tumor immunity. In this review, we summarize the crosstalk between breast cancer and peripheral nerves and the roles of important neuroactive molecules in the progression of breast cancer. In addition, we summarize indicators, including nerve fiber density and perineural invasion (PNI), that may help determine the prognosis of breast cancer based on current research results, as well as potential therapeutic approaches, such as β-blockers and retroviral-mediated genetic neuroengineering techniques, that may enhance the prognosis of breast cancer. In addition, we propose suggestions for future research priorities based on a current lack of knowledge in this area.
Collapse
Affiliation(s)
- Jianming Hu
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Wuzhen Chen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Lesang Shen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Zhigang Chen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China..
| | - Jian Huang
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China..
| |
Collapse
|
5
|
Nojima S. Class IV semaphorins in disease pathogenesis. Pathol Int 2022; 72:471-487. [PMID: 36066011 DOI: 10.1111/pin.13270] [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: 05/16/2022] [Accepted: 08/16/2022] [Indexed: 12/01/2022]
Abstract
Semaphorins are a large family of secreted and/or transmembrane proteins, originally identified as proteins that function in axon guidance during neuronal development. However, semaphorins play crucial roles in other physiological and pathological processes, including immune responses, angiogenesis, maintenance of tissue homeostasis, and cancer progression. Class IV semaphorins may be present as transmembrane and soluble forms and are implicated in the pathogenesis of various diseases. This review discusses recent progress on the roles of class IV semaphorins determined by clinical and experimental pathology studies.
Collapse
Affiliation(s)
- Satoshi Nojima
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan
| |
Collapse
|
6
|
Kuhlmann L, Govindarajan M, Mejia-Guerrero S, Ignatchenko V, Liu LY, Grünwald BT, Cruickshank J, Berman H, Khokha R, Kislinger T. Glycoproteomics Identifies Plexin-B3 as a Targetable Cell Surface Protein Required for the Growth and Invasion of Triple-Negative Breast Cancer Cells. J Proteome Res 2022; 21:2224-2236. [PMID: 35981243 PMCID: PMC9442790 DOI: 10.1021/acs.jproteome.2c00332] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Driven by the lack of targeted therapies, triple-negative
breast cancers
(TNBCs) have the worst overall survival of all breast cancer subtypes.
Considering that cell surface proteins are favorable drug targets
and are predominantly glycosylated, glycoproteome profiling has significant
potential to facilitate the identification of much-needed drug targets
for TNBCs. Here, we performed N-glycoproteomics on
six TNBCs and five normal control (NC) cell lines using hydrazide-based
enrichment. Quantitative proteomics and integrative data mining led
to the discovery of Plexin-B3 (PLXNB3), a previously undescribed TNBC-enriched
cell surface protein. Furthermore, siRNA knockdown and CRISPR-Cas9
editing of in vitro and in vivo models show that PLXNB3 is required
for TNBC cell line growth, invasion, and migration. Altogether, we
provide insights into N-glycoproteome remodeling
associated with TNBCs and functional evaluation of an extracted target,
which indicate the surface protein PLXNB3 as a potential therapeutic
target for TNBCs.
Collapse
Affiliation(s)
- Laura Kuhlmann
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Meinusha Govindarajan
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Salvador Mejia-Guerrero
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Vladimir Ignatchenko
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Lydia Y Liu
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Barbara T Grünwald
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Jennifer Cruickshank
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Hal Berman
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario M5G 1L7, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Rama Khokha
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Thomas Kislinger
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| |
Collapse
|
7
|
Liang Y, Meng K, Qiu R. Circular RNA Circ_0013958 Functions as a Tumor Promoter in Ovarian Cancer by Regulating miR-637/PLXNB2 Axis. Front Genet 2021; 12:644451. [PMID: 34367233 PMCID: PMC8334736 DOI: 10.3389/fgene.2021.644451] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/30/2021] [Indexed: 01/22/2023] Open
Abstract
Background: Circular RNAs (circRNAs) have emerged as important regulators in diverse human malignancies, including ovarian cancer (OC). This study was performed to explore the function and regulatory mechanism underlying circ_0013958 in OC progression. Methods: Quantitative real-time polymerase chain reaction (qRT-PCR) or Western blot assay was applied to examine the expression of circ_0013958, microRNA-637 (miR-637), and Plexin B2 (PLXNB2). The target relationship between miR-637 and circ_0013958 or PLXNB2 was verified by dual-luciferase reporter assay or RNA immunoprecipitation (RIP) assay. Cell Counting Kit-8 (CCK-8) and colony formation assays were employed to detect cell viability and clonogenicity ability, respectively. Cell migration and invasion were analyzed by Transwell assay. Cell apoptosis was monitored by flow cytometry. The role of circ_0013958 in vivo was determined by xenograft tumor assay. Results: Circ_0013958 and PLXNB2 were upregulated, while miR-637 was downregulated in OC tissues and cells. Circ_0013958 acted as a sponge for miR-637 to regulate the expression of PLXNB2 in OC cells. The repression effects of circ_0013958 knockdown on cell proliferation, migration, invasion, and apoptosis in OC cells were partly attenuated by the miR-637 inhibitor. And miR-637 targeted PLXNB2 to suppress OC cell proliferation, migration, and invasion. Moreover, circ_0013958 silencing blocked OC tumor growth in vivo. Conclusion: Circ_0013958 knockdown impeded OC development through modulating the miR-637/PLXNB2 axis, highlighting a therapeutic target for OC.
Collapse
Affiliation(s)
- Yanfei Liang
- Department of Gynecology, The Second Nanning People's Hospital, Nanning, China
| | - Kaiyi Meng
- Department of Gynecology, The Second Nanning People's Hospital, Nanning, China
| | - Rui Qiu
- Department of Gynecology, The Second Nanning People's Hospital, Nanning, China
| |
Collapse
|
8
|
SEMA4D Knockdown Attenuates β-Catenin-Dependent Tumor Progression in Colorectal Cancer. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8507373. [PMID: 34337054 PMCID: PMC8321723 DOI: 10.1155/2021/8507373] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/14/2021] [Accepted: 07/08/2021] [Indexed: 01/06/2023]
Abstract
Semaphorin 4D (SEMA4D), a protein originally demonstrated to regulate the immune system and axonal growth cone collapse in the developing central nervous system, is overexpressed in various human malignancies, including colorectal cancer (CRC). This investigation was undertaken to examine the effects of SEMA4D silencing on the biological properties of the CRC cell line. SW48 cells were transfected with a siRNA-targeting SEMA4D. The mRNA expression of underlying pro- and antiapoptotic proteins including Bax, Bcl-2, P53, and caspase-3, cancer stem cell (CSC) markers, epithelial-mesenchymal transition (EMT) markers, MMP-2, and MMP-9 was examined using qRT-PCR. Further, the protein expression of E-cadherin and β-catenin was confirmed by Western blot. SW48 cell migration and MMP activity were detected using scratch and zymography analysis, respectively. Finally, the apoptosis rate was assessed via the flowcytometry test. SEMA4D knock-down was associated with a considerable suppression of in vitro cell viability, EMT-related genes, CSC markers, β-catenin signaling pathway, sphere-forming, cell migration, and MMP-2 activity as well as induction of apoptosis. This study identifies the inhibitory effects of SEMA4D gene silencing on tumor progression. Thereby, this might conclude a possible alternative to cancer therapy by targeting several prominent pathways involved in cancer through SEMA4D suppression.
Collapse
|
9
|
Sandström J, Balian A, Lockowandt R, Fornander T, Nordenskjöld B, Lindström L, Pérez-Tenorio G, Stål O. IP6K2 predicts favorable clinical outcome of primary breast cancer. Mol Clin Oncol 2021; 14:94. [PMID: 33767863 PMCID: PMC7976380 DOI: 10.3892/mco.2021.2256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/09/2021] [Indexed: 01/15/2023] Open
Abstract
The inositol hexakisphosphate kinase (IP6K) 1 and 2 genes are localized at 3p21.31, a highly altered gene-dense chromosomal region in cancer. The IP6Ks convert IP6 to IP7, which inhibits activation of the tumor-promoting PI3K/Akt/mTOR signaling pathway. IP6K2 has been suggested to be involved in p53-induced apoptosis, while IP6K1 may stimulate tumor growth and migration. The present study aimed to elucidate the role of the two IP6Ks in predicting outcome in patients with breast cancer. To the best of our knowledge, the role of IP6K was analyzed for the first time in tumors from three cohorts of patients with breast cancer; one Swedish low-risk cohort, one Dutch cohort and the TCGA dataset. Analyses of gene -and protein expression and subcellular localization were included. IP6K2 gene expression was associated with ER positivity and nuclear p-Akt. Improved prognosis was detected with high IP6K2 gene expression compared with low IP6K2 gene expression in systemically untreated patients in the Swedish low-risk and Dutch cohorts. In the TCGA dataset, IP6K2 prognostic value was significant when selecting for tumors with wild-type TP53. A multivariable analysis testing IP6K2 against other cancer-related genes at 3p.21.31, including IP6K1 and clinical biomarkers, revealed that IP6K2 was associated with decreased risk of distant recurrence. IP6K1 was associated with increased risk of distant recurrence in the multivariable test and protein analysis revealed trends of worse prognosis with high IP6K1 in the cytoplasm. The expression levels of IP6K1 and IP6K2 were associated to a high extent; however, a diverging prognostic value of the two genes was observed in breast cancer. The present data suggest that IP6K2 can be a favorable prognostic factor, while IP6K1 may not be.
Collapse
Affiliation(s)
- Josefine Sandström
- Department of Biomedical and Clinical Sciences and Department of Oncology, Linköping University, 581 83 Linköping, Sweden
| | - Alien Balian
- Department of Biomedical and Clinical Sciences and Department of Oncology, Linköping University, 581 83 Linköping, Sweden
| | - Rebecca Lockowandt
- Department of Biomedical and Clinical Sciences and Department of Oncology, Linköping University, 581 83 Linköping, Sweden
| | - Tommy Fornander
- Department of Oncology-Pathology, Karolinska Institute, 171 76 Stockholm, Sweden
| | - Bo Nordenskjöld
- Department of Biomedical and Clinical Sciences and Department of Oncology, Linköping University, 581 83 Linköping, Sweden
| | - Linda Lindström
- Department of Biosciences and Nutrition, Karolinska Institute, 141 83 Stockholm, Sweden
| | - Gizeh Pérez-Tenorio
- Department of Biomedical and Clinical Sciences and Department of Oncology, Linköping University, 581 83 Linköping, Sweden
| | - Olle Stål
- Department of Biomedical and Clinical Sciences and Department of Oncology, Linköping University, 581 83 Linköping, Sweden
| |
Collapse
|
10
|
Mastrantonio R, You H, Tamagnone L. Semaphorins as emerging clinical biomarkers and therapeutic targets in cancer. Theranostics 2021; 11:3262-3277. [PMID: 33537086 PMCID: PMC7847692 DOI: 10.7150/thno.54023] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/06/2020] [Indexed: 12/15/2022] Open
Abstract
Semaphorins are a large family of developmental regulatory signals, characterized by aberrant expression in human cancers. These molecules crucially control cell-cell communication, cell migration, invasion and metastasis, tumor angiogenesis, inflammatory and anti-cancer immune responses. Semaphorins comprise secreted and cell surface-exposed molecules and their receptors are mainly found in the Plexin and Neuropilin families, which are further implicated in a signaling network controlling the tumor microenvironment. Accumulating evidence indicates that semaphorins may be considered as novel clinical biomarkers for cancer, especially for the prediction of patient survival and responsiveness to therapy. Moreover, preclinical experimental studies have demonstrated that targeting semaphorin signaling can interfere with tumor growth and/or metastatic dissemination, suggesting their relevance as novel therapeutic targets in cancer; this has also prompted the development of semaphorin-interfering molecules for application in the clinic. Here we will survey, in diverse human cancers, the current knowledge about the relevance of semaphorin family members, and conceptualize potential lines of future research development in this field.
Collapse
|
11
|
Rashidi G, Rezaeepoor M, Mohammadi C, Solgi G, Najafi R. Inhibition of semaphorin 4D enhances chemosensitivity by increasing 5-fluorouracile-induced apoptosis in colorectal cancer cells. Mol Biol Rep 2020; 47:7017-7027. [PMID: 32888127 DOI: 10.1007/s11033-020-05761-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/28/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023]
Abstract
Overexpression of semaphorin 4D (SEMA4D), an immune semaphorin, is found in various human malignancies, including colorectal cancer (CRC). In this study, we explored the relationship between silencing SEMA4D expression and 5-fluorouracil (5-FU) response in the colorectal cancer cell line. SW48 cells were transfected with a short interfering RNA (siRNA) in order to silence SEMA4D gene expression and then exposed to 5-FU for 48 h. The down-regulation of SEMA4D expression was confirmed by qRT-PCR and the particular concentration of 5-FU was acquired using MTT assay. Flow cytometry and western blot were used to evaluate apoptosis rate and pro- and anti-apoptotic expression levels of proteins involved in apoptosis including Bax, Bcl-2, P53, and caspase-3. Other oncogenic activities including epithelial-mesenchymal transition (EMT) process, cancer stem cell (CSC) markers, and β-catenin pathway were investigated using qRT-PCR, and western blot. The proliferation was analyzed via colony formation test and cell invasion was assessed by transwell assay. Our data demonstrate that SEMA4D silencing results in strikingly elevated apoptosis in response to 5-FU treatment and leads to down-regulation of Bcl-2 and overexpression of Bax, P53, and caspase-3 in protein levels. Furthermore, the mRNA and protein expression levels of β-catenin, as well as transcript expressions of CSCs and EMT markers, were remarkably diminished. However, mRNA expression of E-cadherin as an epithelial marker was significantly increased in 5-FU treatment combined with siRNA SEMA4D. This study implicates that the silencing of SEMA4D by siRNA promotes the chemosensitivity of SW48 cells to 5-FU and it may be a potential therapeutic agent for colon cancer therapy.
Collapse
Affiliation(s)
- Golnaz Rashidi
- Department of Immunology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mahsa Rezaeepoor
- Department of Immunology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Chiman Mohammadi
- Department of Molecular Medicine and Genetics, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ghasem Solgi
- Department of Immunology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rezvan Najafi
- Department of Molecular Medicine and Genetics, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran. .,Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
12
|
Wang HM, Zhang XH, Ye LQ, Zhang K, Yang NN, Geng S, Chen J, Zhao SX, Yang KL, Fan FF. Insufficient CD100 shedding contributes to suppression of CD8 + T-cell activity in non-small cell lung cancer. Immunology 2020; 160:209-219. [PMID: 32149403 DOI: 10.1111/imm.13189] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/01/2020] [Accepted: 03/02/2020] [Indexed: 12/11/2022] Open
Abstract
CD100 is an immune semaphorin constitutively expressed on T-cells. Matrix metalloproteinase (MMP) is an important mediator of membrane-bound CD100 (mCD100) cleavage to generate soluble CD100 (sCD100), which has immunoregulatory activity in immune cell responses. The aim of the study was to investigate the level and role of sCD100 and mCD100 in modulating CD8+ T-cell function in non-small cell lung cancer (NSCLC). sCD100 and MMP-14 levels in the serum and bronchoalveolar lavage fluid (BALF), and mCD100 expression on peripheral and lung-resident CD8+ T-cells were analysed in NSCLC patients. The ability to induce sCD100 and the effect of MMP-14 on mCD100 shedding for the regulation of non-cytolytic and cytolytic functions of CD8+ T-cells were also analysed in direct and indirect contact co-culture systems. NSCLC patients had lower serum sCD100 and higher mCD100 levels on CD8+ T-cells compared with healthy controls. BALF from the tumour site also had decreased sCD100 and increased mCD100 on CD8+ T-cells compared with the non-tumour site. Recombinant CD100 stimulation enhanced non-cytolytic and cytolytic functions of CD8+ T-cells from NSCLC patients, whereas blockade of CD100 receptor CD72 attenuated CD8+ T-cell activity. NSCLC patients had lower MMP-14 in the serum and in BALF from the tumour site. Recombinant MMP-14 mediated mCD100 shedding from CD8+ T-cell membrane, and led to promotion of CD8+ T-cell response in NSCLC patients. Overall, decreased MMP-14 resulted in insufficient CD100 shedding, leading to suppression of peripheral and lung-resident CD8+ T-cell activity in NSCLC.
Collapse
Affiliation(s)
- Hong-Min Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiao-Hong Zhang
- Department of Respiratory Medicine, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Li-Qun Ye
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kai Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ning-Ning Yang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shen Geng
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Chen
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shun-Xin Zhao
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kang-Li Yang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fei-Fei Fan
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
13
|
Xu Q, Liu H, Yu B, Chen W, Zhai L, Li X, Fang Y. Long noncoding RNA ZEB2-AS1 facilitates laryngeal squamous cell carcinoma progression by miR-6840-3p/PLXNB1 axis. Onco Targets Ther 2019; 12:7337-7345. [PMID: 31564916 PMCID: PMC6735660 DOI: 10.2147/ott.s212749] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/31/2019] [Indexed: 02/01/2023] Open
Abstract
Purpose To investigate the role of zinc finger E‑box‑binding homeobox 2 antisense RNA 1 (ZEB2-AS1) in regulating laryngeal squamous cell carcinoma (LSCC) progression. Patients and methods In this retrospective study, we included all patients who underwent a surgical operation at The First Hospital of Qiqihaer City for LSCC. Then, we compared the expression of ZEB2-AS1 in LSCC tissues and paired healthy tissues. Besides, we also performed a series of functional assays, CCK8 assays, colony formation assays, and transwell assays to examine the functions of LSCC cells after knockdown of ZEB2-AS1. Through bioinformatics analysis, we predicted that ZEB2-AS1 binds to miR-6840-3p and targets PLXNB1. Results We indicated that the expression of ZEB2-AS1 was higher in LSCC tissues compared to the paired adjacent tissues, and ZEB2-AS1 was also highly expressed in LSCC cell lines. Furthermore, we discovered that ZEB2-AS1 promoted cell proliferation, migration and invasion and was associated with poor prognosis. To find the mechanism, we performed bioinformatics analysis. We identified that ZEB2-AS1 binds to miR-6840-3p and targets PLXNB1. Additionally, miR-6840-3p overexpression or knockdown of PLXNB1 decreased the abilities of cell migration and invasion. Conclusion These findings demonstrated that overexpression of ZEB2-AS1 promotes LSCC progression. Overexpression of miR-6840-3p or downregulation of PLXNB1 can abrogate ZEB2-AS1-mediated LSCC malignant development.
Collapse
Affiliation(s)
- Qiushi Xu
- Ear Nose and Throat Department, Affiliated Qiqihar Hospital, Southern Medical University, The First Hospital of Qiqihaer City, Guangzhou, Heilongjiang 161000, People's Republic of China
| | - Hongyu Liu
- Ear Nose and Throat Department, Affiliated Qiqihar Hospital, Southern Medical University, The First Hospital of Qiqihaer City, Guangzhou, Heilongjiang 161000, People's Republic of China
| | - Bing Yu
- Pathology Department, Affiliated Qiqihar Hospital, Southern Medical University, The First Hospital of Qiqihaer City, Guangzhou, Heilongjiang 161000, People's Republic of China
| | - Wenjing Chen
- Pathology Department, Affiliated Qiqihar Hospital, Southern Medical University, The First Hospital of Qiqihaer City, Guangzhou, Heilongjiang 161000, People's Republic of China
| | - Lili Zhai
- Pathology Department, Affiliated Qiqihar Hospital, Southern Medical University, The First Hospital of Qiqihaer City, Guangzhou, Heilongjiang 161000, People's Republic of China
| | - XueYing Li
- Ear Nose and Throat Department, Affiliated Qiqihar Hospital, Southern Medical University, The First Hospital of Qiqihaer City, Guangzhou, Heilongjiang 161000, People's Republic of China
| | - Yanchun Fang
- Pathology Department, Affiliated Qiqihar Hospital, Southern Medical University, The First Hospital of Qiqihaer City, Guangzhou, Heilongjiang 161000, People's Republic of China
| |
Collapse
|
14
|
Zhang Y, Shen S, Li P, Fan Y, Zhang L, Li W, Liu Y. PLEXIN-B2 promotes the osteogenic differentiation of human bone marrow mesenchymal stem cells via activation of the RhoA signaling pathway. Cell Signal 2019; 62:109343. [PMID: 31176746 DOI: 10.1016/j.cellsig.2019.06.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/30/2019] [Accepted: 06/05/2019] [Indexed: 01/07/2023]
Abstract
Plexin-B2 (PLXNB2), a transmembrane protein is found in various tissues. Recent studies have indicated the presence of PLXNB2 in large quantity in the growth plates of Sprague-Dawley rats and are believed to be potentially involved in their skeletal development. This study endeavored to analyze the effect of PLXNB2 on the osteogenic differentiation of BMSCs by using gene overexpression and knockdown assays. The results of our study revealed that PLXNB2 was upregulated during BMSCs differentiation into an osteoblastic lineage. By determining the expression levels of specific markers and mineral deposition, the study established that PLXNB2 promotes the osteogenic differentiation of human BMSCs through the activation of the RhoA signaling pathway. In conclusion, the study identified PLXNB2 as a novel regulator that enhanced the osteogenic differentiation of human BMSCs. The enhancing effect of PLXNB2 on osteogenesis of human BMSCs was mediated through activation of RhoA signaling. The results of our study imply that pharmacological targeting of PLXNB2 may initiate a possible improvement in bone formation.
Collapse
Affiliation(s)
- Ying Zhang
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital, Orthopedics Hospital of Henan Province, 82 Qiming South Road, Luoyang, Henan 471002, China
| | - Sheng Shen
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital, Orthopedics Hospital of Henan Province, 82 Qiming South Road, Luoyang, Henan 471002, China
| | - Peifeng Li
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital, Orthopedics Hospital of Henan Province, 82 Qiming South Road, Luoyang, Henan 471002, China
| | - Yanan Fan
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital, Orthopedics Hospital of Henan Province, 82 Qiming South Road, Luoyang, Henan 471002, China
| | - Leilei Zhang
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital, Orthopedics Hospital of Henan Province, 82 Qiming South Road, Luoyang, Henan 471002, China
| | - Wuyin Li
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital, Orthopedics Hospital of Henan Province, 82 Qiming South Road, Luoyang, Henan 471002, China.
| | - Youwen Liu
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital, Orthopedics Hospital of Henan Province, 82 Qiming South Road, Luoyang, Henan 471002, China.
| |
Collapse
|
15
|
Plasma levels of Semaphorin 4D are decreased by adjuvant tamoxifen but not aromatase inhibitor therapy in breast cancer patients. J Bone Oncol 2019; 16:100237. [PMID: 31011525 PMCID: PMC6461588 DOI: 10.1016/j.jbo.2019.100237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/01/2019] [Accepted: 04/03/2019] [Indexed: 01/28/2023] Open
Abstract
Background Semaphorin 4D (Sema4D) is a glycoprotein that inhibits bone formation and has been associated with cancer progression and the occurrence of bone metastases. Recently, Sema4D expression has been linked to estrogen signaling in breast cancer. Endocrine therapies like tamoxifen and aromatase inhibitors (AI) are a standard therapeutic approach in hormone receptor positive breast cancers. Tamoxifen exerts ER-agonistic effects on bone, whereas AI negatively affect bone health by increasing resorption and fracture risk. The effect of endocrine therapies on circulating Sema4D levels in breast cancer patients has not been investigated yet. Methods We measured circulating Sema4D plasma levels at primary diagnosis and in a follow-up sample 12 months after surgery in a cohort of 46 pre- and postmenopausal women with primary estrogen receptor positive breast cancer receiving adjuvant tamoxifen or AI. Results The mean baseline levels ± SD for Sema4D were 441.6 ± 143.4 pmol/l. No significant differences in total plasma Sema4D were observed when stratifying the patients according to age, menopausal status, tumor subtype, nodal and hormone receptor status, or tumor size. However, Sema4D levels were significantly reduced by 28% (p<0.001) in tamoxifen treated patients 12 months after surgery, whereas no alteration was observed in patients treated with AI. Conclusion This finding potentially represents an additional mechanism of the bone-protective properties of tamoxifen and further emphasizes a link between Sema4D and estrogen receptor signaling.
Collapse
|
16
|
Yang Y, Wang J, Li H, Liu L, Yao M, Xiao T. Association between prognosis and SEMA4D/Plexin-B1 expression in various malignancies: A meta-analysis. Medicine (Baltimore) 2019; 98:e13298. [PMID: 30762724 PMCID: PMC6407964 DOI: 10.1097/md.0000000000013298] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION SEMA4D and its high affinity receptor Plexin-B1 showed a promising prognosis prediction for carcinoma patients in recent studies, we performed a meta-analysis to evaluate the prognostic role of them in various malignancies. METHODS A systematic literature search was performed in PubMed, Embase, Web of Science, and CNKI from inception till July 2017. Eligible studies were identified by different reviewers. Hazard ratios (HRs)/related ratios (RRs) and their corresponding 95% confidence intervals (CIs) were extracted to investigate the relevance between malignancies prognosis and SEMA4D/Plexin-B1. RESULTS Around 2638 patients from 14 studies were included in this meta-analysis. High expression of SEMA4D was significantly associated with overall survival (OS) and disease-free survival/progression-free survival/recurrence-free survival (DFS/PFS/RFS) in tumors (respectively, HRos = 2.05, 95%CI: 1.68-2.50, P < .001; HRdfs/pfs/rfs = 1.59, 95%CI = 1.27-1.98, P < .001). However, the relationship between SEMA4D expression and prognosis of breast cancer patients was failed to find (HR = 0.76, 95%CI = 0.32-1.82, P = .539). Plexin-B1 level showed a significant positive correlation both with OS and DFS of Caucasian breast cancer patients (respectively, HRos = 0.56, 95%CI: 0.39-0.79, P = .001; HRdfs = 0.68, 95%CI = 0.51-0.90, P = .008) CONCLUSIONS:: SEMA4D could be a prospective biomarker for prognostic prediction of various malignancies except breast cancer. For Caucasian breast cancer patients, SEMA4D's high affinity receptor Plexin-B1 showed a significant positive correlation with survival.
Collapse
Affiliation(s)
- Yibo Yang
- Department of Sport Surgery and Sport Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University)
| | - Jing Wang
- Department of Sport Surgery and Sport Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University)
| | - Hui Li
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Lihong Liu
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Maojin Yao
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA
| | - Tao Xiao
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| |
Collapse
|
17
|
Butti R, Kumar TV, Nimma R, Kundu GC. Impact of semaphorin expression on prognostic characteristics in breast cancer. BREAST CANCER-TARGETS AND THERAPY 2018; 10:79-88. [PMID: 29910635 PMCID: PMC5987790 DOI: 10.2147/bctt.s135753] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Breast cancer is one of the major causes of cancer-related deaths among women worldwide. Aberrant regulation of various growth factors, cytokines, and other proteins and their receptors in cancer cells drives the activation of various oncogenic signaling pathways that lead to cancer progression. Semaphorins are a class of proteins which are differentially expressed in various types of cancer including breast cancer. Earlier, these proteins were known to have a major function in the nerve cell adhesion, migration, and development of the central nervous system. However, their role in the regulation of several aspects of tumor progression has eventually emerged. There are over 30 genes encoding the semaphorins, which are divided into eight subclasses. It has been reported that some members of semaphorin classes are antiangiogenic and antimetastatic in nature, whereas others act as proangiogenic and prometastatic genes. Because of their differential expression and role in angiogenesis and metastasis, semaphorins emerged as one of the important prognostic factors for appraising breast cancer progression.
Collapse
Affiliation(s)
- Ramesh Butti
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, Savitribai Phule Pune University, Pune, India
| | - Totakura Vs Kumar
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, Savitribai Phule Pune University, Pune, India
| | - Ramakrishna Nimma
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, Savitribai Phule Pune University, Pune, India
| | - Gopal C Kundu
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, Savitribai Phule Pune University, Pune, India
| |
Collapse
|
18
|
Abstract
Several neuronal guidance proteins, known as semaphorin molecules, function in the immune system. This dual tissue performance has led to them being defined as "neuroimmune semaphorins". They have been shown to regulate T cell activation by serving as costimulatory molecules. Similar to classical costimulatory molecules, neuroimmune semaphorins are either constitutively or inducibly expressed on immune cells. In contrast to the classical costimulatory molecule function, the action of neuroimmune semaphorins requires the presence of two signals, the first one provided by TCR/MHC engagement, and the second one provided by B7/CD28 interaction. Thus, neuroimmune semaphorins serve as a "signal three" for immune cell activation and regulate the overall intensity of immune response. The current knowledge on their structures, multiple receptors, specific cell/tissue/organ expression, and distinct functions in different diseases are summarized and discussed in this review.
Collapse
Affiliation(s)
- Svetlana P Chapoval
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA.
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA.
- Program in Oncology at the Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA.
- SemaPlex LLC, Ellicott City, MD, USA.
| |
Collapse
|
19
|
Lontos K, Adamik J, Tsagianni A, Galson DL, Chirgwin JM, Suvannasankha A. The Role of Semaphorin 4D in Bone Remodeling and Cancer Metastasis. Front Endocrinol (Lausanne) 2018; 9:322. [PMID: 29971044 PMCID: PMC6018527 DOI: 10.3389/fendo.2018.00322] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 05/28/2018] [Indexed: 12/20/2022] Open
Abstract
Semaphorin 4D (Sema4D; CD100) is a transmembrane homodimer 150-kDa glycoprotein member of the Semaphorin family. Semaphorins were first identified as chemorepellants that guide neural axon growth. Sema4D also possesses immune regulatory activity. Recent data suggest other Sema4D functions: inactivation of platelets, stimulation of angiogenesis, and regulation of bone formation. Sema4D is a coupling factor expressed on osteoclasts that inhibits osteoblast differentiation. Blocking Sema4D may, therefore, be anabolic for bone. Sema4D and its receptor Plexin-B1 are commonly dysregulated in cancers, suggesting roles in cancer progression, invasion, tumor angiogenesis, and skeletal metastasis. This review focuses on Sema4D in bone and cancer biology and the molecular pathways involved, particularly Sema4D-Plexin-B1 signaling crosstalk between cancer cells and the bone marrow microenvironment-pertinent areas since a humanized Sema4D-neutralizing antibody is now in early phase clinical trials in cancers and neurological disorders.
Collapse
Affiliation(s)
- Konstantinos Lontos
- Hematology-Oncology Division, Department of Medicine, UPMC Hillman Cancer Center, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Juraj Adamik
- Hematology-Oncology Division, Department of Medicine, UPMC Hillman Cancer Center, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Anastasia Tsagianni
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Deborah L. Galson
- Hematology-Oncology Division, Department of Medicine, UPMC Hillman Cancer Center, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - John M. Chirgwin
- Hematology and Oncology Division, Department of Medicine, Indiana University School of Medicine, Richard L. Roudebush VA Medical Center, Indianapolis, IN, United States
| | - Attaya Suvannasankha
- Hematology and Oncology Division, Department of Medicine, Indiana University School of Medicine, Richard L. Roudebush VA Medical Center, Indianapolis, IN, United States
- *Correspondence: Attaya Suvannasankha,
| |
Collapse
|
20
|
McColl B, Garg R, Riou P, Riento K, Ridley AJ. Rnd3-induced cell rounding requires interaction with Plexin-B2. J Cell Sci 2016; 129:4046-4056. [PMID: 27656111 PMCID: PMC5117210 DOI: 10.1242/jcs.192211] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/02/2016] [Indexed: 12/24/2022] Open
Abstract
Rnd proteins are atypical members of the Rho GTPase family that induce actin cytoskeletal reorganization and cell rounding. Rnd proteins have been reported to bind to the intracellular domain of several plexin receptors, but whether plexins contribute to the Rnd-induced rounding response is not known. Here we show that Rnd3 interacts preferentially with plexin-B2 of the three plexin-B proteins, whereas Rnd2 interacts with all three B-type plexins, and Rnd1 shows only very weak interaction with plexin-B proteins in immunoprecipitations. Plexin-B1 has been reported to act as a GAP for R-Ras and/or Rap1 proteins. We show that all three plexin-B proteins interact with R-Ras and Rap1, but Rnd proteins do not alter this interaction or R-Ras or Rap1 activity. We demonstrate that plexin-B2 promotes Rnd3-induced cell rounding and loss of stress fibres, and enhances the inhibition of HeLa cell invasion by Rnd3. We identify the amino acids in Rnd3 that are required for plexin-B2 interaction, and show that mutation of these amino acids prevents Rnd3-induced morphological changes. These results indicate that plexin-B2 is a downstream target for Rnd3, which contributes to its cellular function.
Collapse
Affiliation(s)
- Brad McColl
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Ritu Garg
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Philippe Riou
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Kirsi Riento
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Anne J Ridley
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| |
Collapse
|
21
|
Ikeya T, Maeda K, Nagahara H, Shibutani M, Iseki Y, Hirakawa K. The combined expression of Semaphorin4D and PlexinB1 predicts disease recurrence in colorectal cancer. BMC Cancer 2016; 16:525. [PMID: 27456345 PMCID: PMC4960918 DOI: 10.1186/s12885-016-2577-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 07/18/2016] [Indexed: 01/15/2023] Open
Abstract
Background Binding to Sema4D and PlexinB1 induce angiogenesis and invasive growth in colorectal cancer (CRC). The expression of Semaphorin4D (Sema4D) and PlexinB1 has been shown to be related to the prognosis of patients with various malignancies. However, the correlation between the expression of Sema4D and PlexinB1 and the relapse-free survival in patients with colorectal cancer remains controversial. Methods The study population included patients who underwent surgery for colorectal cancer (n = 226). The expression of Sema4D and PlexinB1 were analyzed by immunohistochemistry in tissue of stage I, II, and III colon cancers. Results The immunohistochemical staining of colorectal cancer tissue specimens revealed that 95 (42 %) and 105 (46.4 %) of the specimens were positive for Sema4D and PlexinB1. The expression of Sema4D and PlexinB1 respectively were both found to be significantly related to stage, depth of tumor invasion, lymph node metastasis, lymphatic invasion, and venous invasion, respectively. Sixty-three patients (27.9 %) expressed both Sema4D and PlexinB1. The positive expression of both Sema4D and PlexinB1 was found to be an independent risk factor for a worse survival (HR 1.079, CI 1.013–2.868; P = 0.044). Conclusion The combination of Sema4D and PlexinB1 protein detected by immunohistochemistry was therefore useful for predicting disease recurrence in CRC patients.
Collapse
Affiliation(s)
- Tetsuro Ikeya
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka, Japan.
| | - Kiyoshi Maeda
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka, Japan
| | - Hisashi Nagahara
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka, Japan
| | - Masatsune Shibutani
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka, Japan
| | - Yasuhito Iseki
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka, Japan
| | - Kosei Hirakawa
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka, Japan
| |
Collapse
|