1
|
Dhall A, Taniyama D, Elloumi F, Luna A, Varma S, Kumar S, Escobedo L, Aladjem MI, Redon CE, Roper N, Reinhold WC, Del Rivero J, Pommier Y. Bimodal genomic approach predicting Semaphorin 7A (SEMA7A) as prognostic biomarker in adrenocortical carcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.03.647086. [PMID: 40291702 PMCID: PMC12026583 DOI: 10.1101/2025.04.03.647086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Adrenocortical carcinoma (ACC) is a rare and aggressive endocrine malignancy with high mortality and poor prognosis. To elucidate the genetic underpinnings of ACCs, we have analyzed the transcriptome data of 112 ACC tumor samples from patients enrolled in the TCGA and NCI. Among 72 bimodally expressed genes stratifying patients into prognostic groups, we focused on SEMA7A , as it encodes a glycosylphosphatidylinositol-anchored membrane glycoprotein (Semaphorin 7a) regulating integrin-mediated signaling, cell migration and immune responses. We find that high SEMA7A gene expression is associated with poor prognosis (hazard ratio = 4.27; p-value < 0.001). In hormone-producing ACCs, SEMA7A expression is elevated and positively correlated with genes driving steroidogenesis, aldosterone and cortisol synthesis, including CYP17A1, CYP11A1, INHA, DLK1, NR5A1 and MC2R . Correlation analyses show that SEMA7A is co-expressed with the integrin-β1, FAK (focal adhesion kinase) and MAPK/ERK (mitogen-activated protein kinase/extracellular signal regulated kinases) signaling pathways. Immunohistochemistry (IHC) staining demonstrates the feasibility of evaluating SEMA7A in ACC tissues and shows significant correlation between gene expression (RNA-Seq) and protein expression (IHC). These findings suggest SEMA7A as a candidate for further research in ACC biology, a candidate for cancer therapy, as well as a potential prognosis biomarker for ACC patients. Translational relevance Adrenocortical cancer (ACC) remains a challenging disease primarily due to the scarcity of reliable biomarkers for predicting patient outcomes and informing innovative therapeutic strategies, as well as its rarity, which restricts the scope of clinical trials. In our study, we performed RNAseq and IHC analyses of ACC samples sourced from The Cancer Genome Atlas (TCGA), tissue microarray slide, and National Cancer Institute (NCI) cancer patient samples. Our findings indicate that a substantial proportion of ACC tumors exhibit expression of SEMA7A, a glycoprotein involved in Semaphorin cell surface signaling. Notably, elevated levels of SEMA7A were identified as a poor prognostic biomarker and were associated with activation of the integrin-ERK-MAPK kinase signaling pathways. These results suggest that ACC tumors with high SEMA7A expression should be considered at elevated risk, and SEMA7A may serve as a potential target for immunotherapeutic strategies, including antibody-drug conjugates, T-cell engagers, and/or small molecule inhibitors targeting the MAPK pathway.
Collapse
|
2
|
Yang L, Zhang S, Zheng L, Kong F, Dang W, Shen S, Li X, Jia L, Zhang X, Lu N. Pan-Cancer Analysis of the Prognostic and Immunological Role of SEMA7A. Int J Gen Med 2024; 17:6443-6461. [PMID: 39735166 PMCID: PMC11682669 DOI: 10.2147/ijgm.s499872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 12/20/2024] [Indexed: 12/31/2024] Open
Abstract
Background Semaphorin7A (SEMA7A) has been found to regulate both nerve and vessel homeostasis, but its specific role in pan-cancer remains uncertain. This research seeks to delve into the function and clinical relevance of SEMA7A in pan-cancer. Methods Through an analysis of gene expression omnibus and the cancer genome atlas datasets, we investigated the impact of SEMA7A on prognosis and immune regulation across 33 types of tumors. Variations in SEMA7A expression were observed between cancerous and adjacent normal tissues, with a notable correlation between SEMA7A levels and patient prognosis. Results Across most cancer types, SEMA7A expression was linked to the infiltration of immune cells, as well as immune checkpoints and other immune regulators. The findings were further confirmed through quantitative real-time polymerase chain reaction analysis of SEMA7A expression in breast cancer. Further, SEMA7A is positively associated with prognosis in different cancers. Additionally, SEMA7A expression was associated with TMB and MSI in some cancer types, while in 15 types of cancer, there was a correlation between SEMA7A expression and DNA methylation. SEMA7A was associated with the expression of multiple immune checkpoint genes and abundance of tumor-infiltrating immune cells across multiple types of cancer. Conclusion This inaugural pan-cancer examination of SEMA7A sheds light on its prognostic and immunological significance in diverse tumor types, suggesting its potential utility as a biomarker for predicting unfavorable outcomes and immune cell infiltration in cancer.
Collapse
Affiliation(s)
- Lixian Yang
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, Hebei, 054000, People’s Republic of China
| | - Shiyu Zhang
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, Hebei, 054000, People’s Republic of China
| | - Lei Zheng
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, Hebei, 054000, People’s Republic of China
| | - Fanting Kong
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, Hebei, 054000, People’s Republic of China
| | - Wei Dang
- Medical Research Center, Xingtai Medical College, Xingtai, Hebei, 054000, People’s Republic of China
| | - Shipeng Shen
- Medical Research Center, Xingtai Medical College, Xingtai, Hebei, 054000, People’s Republic of China
| | - Xiaowei Li
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, Hebei, 054000, People’s Republic of China
| | - Lining Jia
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, Hebei, 054000, People’s Republic of China
| | - Xiaoru Zhang
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, Hebei, 054000, People’s Republic of China
| | - Na Lu
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, Hebei, 054000, People’s Republic of China
| |
Collapse
|
3
|
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
|
4
|
Kamle S, Ma B, Schor G, Bailey M, Pham B, Cho I, Khan H, Azzoli C, Hofstetter M, Sadanaga T, Herbst R, Politi K, Lee CG, Elias JA. Chitinase 3-like-1 (CHI3L1) in the pathogenesis of epidermal growth factor receptor mutant non-small cell lung cancer. Transl Oncol 2024; 49:102108. [PMID: 39178575 PMCID: PMC11388375 DOI: 10.1016/j.tranon.2024.102108] [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: 03/18/2024] [Revised: 07/26/2024] [Accepted: 08/18/2024] [Indexed: 08/26/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for 85 % of all lung cancers. In NSCLC, 10-20 % of Caucasian patients and 30-50 % of Asian patients have tumors with activating mutations in the Epidermal Growth Factor Receptor (EGFR). A high percentage of these patients exhibit favorable responses to treatment with tyrosine kinase inhibitors (TKI). Unfortunately, a majority of these patients develop therapeutic resistance with progression free survival lasting 9-18 months. The mechanisms that underlie the tumorigenic effects of EGFR and the ability of NSCLC to develop resistance to TKI therapies, however, are poorly understood. Here we demonstrate that CHI3L1 is produced by EGFR activation of normal epithelial cells, transformed epithelial cells with wild type EGFR and cells with cancer-associated, activating EGFR mutations. We also demonstrate that CHI3L1 auto-induces itself and feeds back to stimulate EGFR and its ligands via a STAT3-dependent mechanism(s). Highly specific antibodies against CHI3L1 (anti-CHI3L1/FRG) and TKI, individually and in combination, abrogated the effects of EGFR activation on CHI3L1 and the ability of CHI3L1 to stimulate the EGFR axis. Anti-CHI3L1 also interacted with osimertinib to reverse TKI therapeutic resistance and induce tumor cell death and inhibit pulmonary metastasis while stimulating tumor suppressor genes including KEAP1. CHI3L1 is a downstream target of EGFR that feeds back to stimulate and activate the EGFR axis. Anti-CHI3L1 is an exciting potential therapeutic for EGFR mutant NSCLC, alone and in combination with osimertinib or other TKIs.
Collapse
Affiliation(s)
- Suchitra Kamle
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA; Legorreta Cancer Center, Brown University, Providence, RI, USA
| | - Bing Ma
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Gail Schor
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Madison Bailey
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Brianna Pham
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Inyoung Cho
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Hina Khan
- Medical Oncology, Department of Medicine, Warren Alpert Medical School Brown University, USA
| | - Christopher Azzoli
- Medical Oncology, Department of Medicine, Warren Alpert Medical School Brown University, USA
| | - Mara Hofstetter
- Department of Chemistry, Yale University, USA; University of Zurich, Switzerland
| | - Takayuki Sadanaga
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Roy Herbst
- Medical Oncology, Department of Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Katerina Politi
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Chun Geun Lee
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA; Legorreta Cancer Center, Brown University, Providence, RI, USA
| | - Jack A Elias
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA; Legorreta Cancer Center, Brown University, Providence, RI, USA; Departments of Medicine, Alpert Medical School, Brown University, Providence, RI, USA.
| |
Collapse
|
5
|
Zhang S, Kong F, Zheng L, Li X, Jia L, Yang L. SEMA7A as a Novel Prognostic Biomarker and Its Correlation with Immune Infiltrates in Breast Cancer. Int J Gen Med 2024; 17:4081-4099. [PMID: 39295856 PMCID: PMC11410036 DOI: 10.2147/ijgm.s474827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 09/12/2024] [Indexed: 09/21/2024] Open
Abstract
Background The role of Semaphorin 7a (SEMA7A) in the initiation and progression of different types of cancerous lesions has been extensively studied. However, the prognostic significance of SEMA7A, specifically in breast cancer (BC), lacks clarity. Methods We conducted an evaluation on the relationship between SEMA7A and the prognosis, immune invasion and tumor mutation burden in different types of cancer by analyzing data from The Cancer Genome Atlas database. The present study focused on investigating the expression level, mutation, immune correlation and coexpression of SEMA7A in BC, utilizing various databases such as the University of Alabama at Birmingham Cancer data analysis portal, cBioPortal and tumor immune estimation resource. Survival analysis was carried out using the Kaplan-Meier Plotter. Furthermore, we employed the R software package to generate receiver operating characteristic (ROC) curves and nomograms. Notably, P<0.05 was considered to indicate statistical significance. Results Using pancancer analysis, it has been observed that the expression of SEMA7A is elevated in various types of cancer and is strongly correlated with the prognosis of different cancer types. SEMA7A also exhibits a significant association with the tumor mutation burden of diverse types of cancer. Moreover, SEMA7A displays a notable increase in BC cases, and was indicated to have a substantial association with the abundance of immune infiltration. In-depth survival analysis demonstrated that elevated levels of SEMA7A expression are notably linked to shorter overall survival and distant metastasis-free survival among patients with BC. The efficiency of SEMA7A as a reliable prognostic biomarker for BC has been substantiated by the validation of ROC curves and nomograms. Conclusion SEMA7A has the potential to function as a prognostic indicator for BC, and its correlation with immune infiltration in BC is significant.
Collapse
Affiliation(s)
- Shiyu Zhang
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai, Hebei, 054000, People's Republic of China
| | - Fanting Kong
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai, Hebei, 054000, People's Republic of China
| | - Lei Zheng
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai, Hebei, 054000, People's Republic of China
| | - Xiaowei Li
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai, Hebei, 054000, People's Republic of China
| | - Lining Jia
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai, Hebei, 054000, People's Republic of China
| | - Lixian Yang
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai, Hebei, 054000, People's Republic of China
| |
Collapse
|
6
|
Chen T, Li S, Wang L. Semaphorins in tumor microenvironment: Biological mechanisms and therapeutic progress. Int Immunopharmacol 2024; 132:112035. [PMID: 38603857 DOI: 10.1016/j.intimp.2024.112035] [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: 01/08/2024] [Revised: 03/15/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024]
Abstract
Hallmark features of the tumor microenvironment include immune cells, stromal cells, blood vessels, and extracellular matrix (ECM), providing a conducive environment for the growth and survival of tumors. Recent advances in the understanding of cancer biology have highlighted the functional role of semaphorins (SEMAs). SEMAs are a large and diverse family of widely expressed secreted and membrane-binding proteins, which were initially implicated in axon guidance and neural development. However, it is now clear that they are widely expressed beyond the nervous system and participate in regulating immune responses and cancer progression. In fact, accumulating evidence disclosed that different SEMAs can either stimulate or restrict tumor progression, some of which act as important regulators of tumor angiogenesis. Conversely, limited information is known about the functional relevance of SEMA signals in TME. In this setting, we systematically elaborate the role SEMAs and their major receptors played in characterized components of TME. Furthermore, we provide a convergent view of current SEMAs pharmacological progress in clinical treatment and also put forward their potential application value and clinical prospects in the future.
Collapse
Affiliation(s)
- Tianyi Chen
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022, PR China
| | - Shazhou Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022, PR China
| | - Lufang Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022, PR China.
| |
Collapse
|
7
|
Salembier R, De Haes C, Bellemans J, Demeyere K, Van Den Broeck W, Sanders NN, Van Laere S, Lyons TR, Meyer E, Steenbrugge J. Chitin-mediated blockade of chitinase-like proteins reduces tumor immunosuppression, inhibits lymphatic metastasis and enhances anti-PD-1 efficacy in complementary TNBC models. Breast Cancer Res 2024; 26:63. [PMID: 38605414 PMCID: PMC11007917 DOI: 10.1186/s13058-024-01815-8] [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: 08/21/2023] [Accepted: 03/23/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Chitinase-like proteins (CLPs) play a key role in immunosuppression under inflammatory conditions such as cancer. CLPs are enzymatically inactive and become neutralized upon binding of their natural ligand chitin, potentially reducing CLP-driven immunosuppression. We investigated the efficacy of chitin treatment in the context of triple-negative breast cancer (TNBC) using complementary mouse models. We also evaluated the immunomodulatory influence of chitin on immune checkpoint blockade (ICB) and compared its efficacy as general CLP blocker with blockade of a single CLP, i.e. chitinase 3-like 1 (CHI3L1). METHODS Female BALB/c mice were intraductally injected with luciferase-expressing 4T1 or 66cl4 cells and systemically treated with chitin in combination with or without anti-programmed death (PD)-1 ICB. For single CLP blockade, tumor-bearing mice were treated with anti-CHI3L1 antibodies. Metastatic progression was monitored through bioluminescence imaging. Immune cell changes in primary tumors and lymphoid organs (i.e. axillary lymph nodes and spleen) were investigated through flow cytometry, immunohistochemistry, cytokine profiling and RNA-sequencing. CHI3L1-stimulated RAW264.7 macrophages were subjected to 2D lymphatic endothelial cell adhesion and 3D lymphatic integration in vitro assays for studying macrophage-mediated lymphatic remodeling. RESULTS Chitin significantly reduced primary tumor progression in the 4T1-based model by decreasing the high production of CLPs that originate from tumor-associated neutrophils (TANs) and Stat3 signaling, prominently affecting the CHI3L1 and CHI3L3 primary tumor levels. It reduced immunosuppressive cell types and increased anti-tumorigenic T-cells in primary tumors as well as axillary lymph nodes. Chitin also significantly reduced CHI3L3 primary tumor levels and immunosuppression in the 66cl4-based model. Compared to anti-CHI3L1, chitin enhanced primary tumor growth reduction and anti-tumorigenicity. Both treatments equally inhibited lymphatic adhesion and integration of macrophages, thereby hampering lymphatic tumor cell spreading. Upon ICB combination therapy, chitin alleviated anti-PD-1 resistance in both TNBC models, providing a significant add-on reduction in primary tumor and lung metastatic growth compared to chitin monotherapy. These add-on effects occurred through additional increase in CD8α+ T-cell infiltration and activation in primary tumor and lymphoid organs. CONCLUSIONS Chitin, as a general CLP blocker, reduces CLP production, enhances anti-tumor immunity as well as ICB responses, supporting its potential clinical relevance in immunosuppressed TNBC patients.
Collapse
Affiliation(s)
- Robbe Salembier
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Caro De Haes
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Julie Bellemans
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Kristel Demeyere
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Wim Van Den Broeck
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Niek N Sanders
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Steven Van Laere
- Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Traci R Lyons
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, CO, USA
| | - Evelyne Meyer
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Jonas Steenbrugge
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
- Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium.
| |
Collapse
|
8
|
Zhou Y, Liu Z, Liu Y. The potential roles and mechanisms of Chitinase-3-like-1 in the pathogenesis of type 2-biased airway diseases. Clin Immunol 2023; 257:109856. [PMID: 38036279 DOI: 10.1016/j.clim.2023.109856] [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: 09/13/2023] [Revised: 11/14/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
The immune modulation in the epithelium is a protective feature of the epithelial function in the mucosal airways. Dysfunction of the epithelium can lead to chronic allergic airway inflammatory diseases, such as chronic rhinosinusitis with nasal polyps (CRSwNP), allergic rhinitis (AR), and allergic asthma. Chitinase-3-like-1 (CHI3L1) is a key modulator in the epithelium against irritants, pathogens, and allergens and is involved in cancers, autoimmune diseases, neurological disorders, and other chronic diseases. Induction of epithelial cell-derived CHI3L1 is also confirmed to be implicated in the pathogenesis of Th2-related airway diseases like CRSwNP, AR, and allergic asthma, triggering a cascade of subsequent inflammatory reactions leading to the disease development. The techniques that block the biological function of CHI3L1 include small interfering RNA, neutralizing antibodies, and microRNAs and these methods proved to be successful in preclinical and clinical investigation in cancers, autoimmune diseases, asthma, and chronic obstructive pulmonary disease. Therefore, treatment with CHI3L1-blocking methods could open up therapeutic options for allergic airway diseases. This review article discusses the role of epithelial cell-derived CHI3L1 in the development of CRSwNP, AR, and allergic asthma and examines the use of CHI3L1 as a potential therapeutic agent for allergic airway diseases.
Collapse
Affiliation(s)
- Yian Zhou
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China; Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China; Hubei Clinical Research Center for Nasal Inflammatory Diseases, Wuhan, PR China
| | - Zheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China; Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China; Hubei Clinical Research Center for Nasal Inflammatory Diseases, Wuhan, PR China.
| | - Yang Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China; Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China; Hubei Clinical Research Center for Nasal Inflammatory Diseases, Wuhan, PR China.
| |
Collapse
|
9
|
Zhao H, Huang M, Jiang L. Potential Roles and Future Perspectives of Chitinase 3-like 1 in Macrophage Polarization and the Development of Diseases. Int J Mol Sci 2023; 24:16149. [PMID: 38003338 PMCID: PMC10671302 DOI: 10.3390/ijms242216149] [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: 10/16/2023] [Revised: 11/03/2023] [Accepted: 11/05/2023] [Indexed: 11/26/2023] Open
Abstract
Chitinase-3-like protein 1 (CHI3L1), a chitinase-like protein family member, is a secreted glycoprotein that mediates macrophage polarization, inflammation, apoptosis, angiogenesis, and carcinogenesis. Abnormal CHI3L1 expression has been associated with multiple metabolic and neurological disorders, including diabetes, atherosclerosis, and Alzheimer's disease. Aberrant CHI3L1 expression is also reportedly associated with tumor migration and metastasis, as well as contributions to immune escape, playing important roles in tumor progression. However, the physiological and pathophysiological roles of CHI3L1 in the development of metabolic and neurodegenerative diseases and cancer remain unclear. Understanding the polarization relationship between CHI3L1 and macrophages is crucial for disease progression. Recent research has uncovered the complex mechanisms of CHI3L1 in different diseases, highlighting its close association with macrophage functional polarization. In this article, we review recent findings regarding the various disease types and summarize the relationship between macrophages and CHI3L1. Furthermore, this article also provides a brief overview of the various mechanisms and inhibitors employed to inhibit CHI3L1 and disrupt its interaction with receptors. These endeavors highlight the pivotal roles of CHI3L1 and suggest therapeutic approaches targeting CHI3L1 in the development of metabolic diseases, neurodegenerative diseases, and cancers.
Collapse
Affiliation(s)
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou 350116, China;
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fuzhou 350116, China;
| |
Collapse
|
10
|
Toledano S, Neufeld G. Plexins as Regulators of Cancer Cell Proliferation, Migration, and Invasivity. Cancers (Basel) 2023; 15:4046. [PMID: 37627074 PMCID: PMC10452846 DOI: 10.3390/cancers15164046] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/24/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
Plexins are a family of nine single-pass transmembrane receptors with a conserved GTPase activating protein (GAP) domain. The plexin family is divided into four subfamilies: Type-A, type-B, type-C, and type-D plexins. Plexins function as receptors for axon guidance factors of the semaphorin family. The semaphorin gene family contains 22 genes that are divided into eight subclasses of which subclasses three to seven represent vertebrate semaphorins. The plexins and their semaphorin ligands have important roles as regulators of angiogenesis, cancer proliferation, and metastasis. Class 3 semaphorins, with the exception of sema3E, are the only semaphorins that do not bind directly to plexins. In order to transduce their signals, they bind instead to complexes consisting of receptors of the neuropilin family and various plexins. Some plexins also form complexes with tyrosine-kinase receptors such as the epidermal growth factor receptor ErbB2, the mesenchymal epithelial transition factor receptor (MET), and the Vascular endothelial growth factor receptor 2 (VEGFR2) and, as a result, can modulate cell proliferation and tumor progression. This review focuses on the roles of the different plexins in the control of cancer cell proliferation and invasiveness. Plexins also affect tumor progression and tumor metastasis by indirect mechanisms, such as modulation of angiogenesis and immune responses. However, these topics are not covered in the present review.
Collapse
Affiliation(s)
| | - Gera Neufeld
- The Cancer Research Center, The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109602, Israel;
| |
Collapse
|
11
|
Hong DE, Yu JE, Lee JW, Son DJ, Lee HP, Kim Y, Chang JY, Lee DW, Lee WK, Yun J, Han SB, Hwang BY, Hong JT. A Natural CHI3L1-Targeting Compound, Ebractenoid F, Inhibits Lung Cancer Cell Growth and Migration and Induces Apoptosis by Blocking CHI3L1/AKT Signals. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010329. [PMID: 36615523 PMCID: PMC9822003 DOI: 10.3390/molecules28010329] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/24/2022] [Accepted: 12/26/2022] [Indexed: 01/04/2023]
Abstract
Our previous big data analyses reported a strong association between CHI3L1 expression and lung tumor development. In this present study, we investigated whether a CHI3L1-inhibiting natural compound, ebractenoid F, inhibits lung cancer cell growth and migration and induces apoptosis. Ebractenoid F concentration-dependently (0, 17, 35, 70 µM) and significantly inhibited the proliferation and migration of A549 and H460 lung cancer cells and induced apoptosis. In the mechanism study, we found that ebractenoid F bound to CHI3L1 and suppressed CHI3L1-associated AKT signaling. Combined treatment with an AKT inhibitor, LY294002, and ebractenoid F synergistically decreased the expression of CHI3L1. Moreover, the combination treatment further inhibited the growth and migration of lung cancer cells and further induced apoptosis, as well as the expression levels of apoptosis-related proteins. Thus, our data demonstrate that ebractenoid F may serve as a potential anti-lung cancer compound targeting CHI3L1-associated AKT signaling.
Collapse
Affiliation(s)
- Da Eun Hong
- Medical Research Center, College of Pharmacy, Chungbuk National University, Osongsaengmyeong 1-ro, 194-21, Osong-eup, Heungduk-gu, Cheongju 28160, Republic of Korea
| | - Ji Eun Yu
- Medical Research Center, College of Pharmacy, Chungbuk National University, Osongsaengmyeong 1-ro, 194-21, Osong-eup, Heungduk-gu, Cheongju 28160, Republic of Korea
| | - Jin Woo Lee
- Medical Research Center, College of Pharmacy, Chungbuk National University, Osongsaengmyeong 1-ro, 194-21, Osong-eup, Heungduk-gu, Cheongju 28160, Republic of Korea
| | - Dong Ju Son
- Medical Research Center, College of Pharmacy, Chungbuk National University, Osongsaengmyeong 1-ro, 194-21, Osong-eup, Heungduk-gu, Cheongju 28160, Republic of Korea
| | - Hee Pom Lee
- Medical Research Center, College of Pharmacy, Chungbuk National University, Osongsaengmyeong 1-ro, 194-21, Osong-eup, Heungduk-gu, Cheongju 28160, Republic of Korea
| | - Yuri Kim
- Medical Research Center, College of Pharmacy, Chungbuk National University, Osongsaengmyeong 1-ro, 194-21, Osong-eup, Heungduk-gu, Cheongju 28160, Republic of Korea
| | - Ju Young Chang
- Medical Research Center, College of Pharmacy, Chungbuk National University, Osongsaengmyeong 1-ro, 194-21, Osong-eup, Heungduk-gu, Cheongju 28160, Republic of Korea
| | - Dong Won Lee
- Medical Research Center, College of Pharmacy, Chungbuk National University, Osongsaengmyeong 1-ro, 194-21, Osong-eup, Heungduk-gu, Cheongju 28160, Republic of Korea
| | - Won Kyu Lee
- Department of New Drug Development Center, Osong Medical Innovation Foundation (KBio Health), Cheongju 28644, Republic of Korea
| | - Jaesuk Yun
- Medical Research Center, College of Pharmacy, Chungbuk National University, Osongsaengmyeong 1-ro, 194-21, Osong-eup, Heungduk-gu, Cheongju 28160, Republic of Korea
| | - Sang Bae Han
- Medical Research Center, College of Pharmacy, Chungbuk National University, Osongsaengmyeong 1-ro, 194-21, Osong-eup, Heungduk-gu, Cheongju 28160, Republic of Korea
| | - Bang Yeon Hwang
- Medical Research Center, College of Pharmacy, Chungbuk National University, Osongsaengmyeong 1-ro, 194-21, Osong-eup, Heungduk-gu, Cheongju 28160, Republic of Korea
| | - Jin Tae Hong
- Medical Research Center, College of Pharmacy, Chungbuk National University, Osongsaengmyeong 1-ro, 194-21, Osong-eup, Heungduk-gu, Cheongju 28160, Republic of Korea
- Correspondence: ; Tel.: +82-43-261-2813; Fax: +82-43-268-2732
| |
Collapse
|
12
|
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: 21] [Impact Index Per Article: 7.0] [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
|
13
|
Crump LS, Kines KT, Richer JK, Lyons TR. Breast cancers co-opt normal mechanisms of tolerance to promote immune evasion and metastasis. Am J Physiol Cell Physiol 2022; 323:C1475-C1495. [PMID: 36189970 PMCID: PMC9662806 DOI: 10.1152/ajpcell.00189.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 11/22/2022]
Abstract
Normal developmental processes, such as those seen during embryonic development and postpartum mammary gland involution, can be reactivated by cancer cells to promote immune suppression, tumor growth, and metastatic spread. In mammalian embryos, paternal-derived antigens are at risk of being recognized as foreign by the maternal immune system. Suppression of the maternal immune response toward the fetus, which is mediated in part by the trophoblast, is critical to ensure embryonic survival and development. The postpartum mammary microenvironment also exhibits immunosuppressive mechanisms accompanying the massive cell death and tissue remodeling that occurs during mammary gland involution. These normal immunosuppressive mechanisms are paralleled during malignant transformation, where tumors can develop neoantigens that may be recognized as foreign by the immune system. To circumvent this, tumors can dedifferentiate and co-opt immune-suppressive mechanisms normally utilized during fetal tolerance and postpartum mammary involution. In this review, we discuss those similarities and how they can inform our understanding of cancer progression and metastasis.
Collapse
Affiliation(s)
- Lyndsey S Crump
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Kelsey T Kines
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jennifer K Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center, Aurora, Colorado
| | - Traci R Lyons
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center, Aurora, Colorado
| |
Collapse
|
14
|
Yang PS, Yu MH, Hou YC, Chang CP, Lin SC, Kuo IY, Su PC, Cheng HC, Su WC, Shan YS, Wang YC. Targeting protumor factor chitinase-3-like-1 secreted by Rab37 vesicles for cancer immunotherapy. Am J Cancer Res 2022; 12:340-361. [PMID: 34987649 PMCID: PMC8690922 DOI: 10.7150/thno.65522] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023] Open
Abstract
Background: Chitinase 3-like-1 (CHI3L1) is a secretion glycoprotein associated with the immunosuppressive tumor microenvironment (TME). The secretory mode of CHI3L1 makes it a promising target for cancer treatment. We have previously reported that Rab37 small GTPase mediates secretion of IL-6 in macrophages to promote cancer progression, whereas the roles of Rab37 in the intracellular trafficking and exocytosis of CHI3L1 are unclear. Methods: We examined the concentration of CHI3L1 in the culture medium of splenocytes and bone marrow derived macrophages (BMDMs) from wild-type or Rab37 knockout mice, and macrophage or T cell lines expressing wild type, active GTP-bound or inactive GDP-bound Rab37. Vesicle isolation, total internal reflection fluorescence microscopy, and real-time confocal microscopy were conducted. We developed polyclonal neutralizing-CHI3L1 antibodies (nCHI3L1 Abs) to validate the therapeutic efficacy in orthotopic lung, pancreas and colon cancer allograft models. Multiplex fluorescence immunohistochemistry was performed to detect the protein level of Rab37 and CHI3L1, and localization of the tumor-infiltrating immune cells in allografts from mice or tumor specimens from cancer patients. Results: We demonstrate a novel secretion mode of CHI3L1 mediated by the small GTPase Rab37 in T cells and macrophages. Rab37 mediated CHI3L1 intracellular vesicle trafficking and exocytosis in a GTP-dependent manner, which is abolished in the splenocytes and BMDMs from Rab37 knockout mice and attenuated in macrophage or T cell lines expressing the inactive Rab37. The secreted CHI3L1 activated AKT, ß-catenin and NF-κB signal pathways in cancer cells and macrophages to foster a protumor TME characterized by activating M2 macrophages and increasing the population of regulatory T cells. Our developed nCHI3L1 Abs showed the dual properties of reducing tumor growth/metastases and eliciting an immunostimulatory TME in syngeneic orthotopic lung, pancreas and colon tumor models. Clinically, high plasma level or intratumoral expression of CHI3L1 correlated with poor survival in 161 lung cancer, 155 pancreatic cancer and 180 colon cancer patients. Conclusions: These results provide the first evidence that Rab37 mediates CHI3L1 secretion in immune cells and highlight nCHI3L1 Abs that can simultaneously target both cancer cells and tumor microenvironment.
Collapse
|
15
|
Ma B, Kamle S, Akosman B, Khan H, Lee CM, Lee CG, Elias JA. CHI3L1 enhances melanoma lung metastasis via regulation of T cell co-stimulators and CTLA-4/B7 axis. Front Immunol 2022; 13:1056397. [PMID: 36618349 PMCID: PMC9812560 DOI: 10.3389/fimmu.2022.1056397] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
ICOS/ICOSL and CD28/B7-1/B7-2 are T cell co-stimulators and CTLA-4 is an immune checkpoint inhibitor that play critical roles in the pathogenesis of neoplasia. Chitinase 3-like-1 (CHI3L1) is induced in many cancers where it portends a poor prognosis and contributes to tumor metastasis. Here we demonstrate that CHI3L1 inhibits the expression of ICOS, ICOSL and CD28 while stimulating CTLA-4 and the B7 moieties in melanoma lung metastasis. We also demonstrate that RIG-like helicase innate immune activation augments T cell co-stimulation, inhibits CTLA-4 and suppresses pulmonary metastasis. At least additive antitumor responses were seen in melanoma lung metastasis treated with anti-CTLA-4 and anti-CHI3L1 antibodies in combination. Synergistic cytotoxic T cell-induced tumor cell death and the heightened induction of the tumor suppressor PTEN were seen in co-cultures of T and tumor cells treated with bispecific antibodies that target both CHI3L1 and CTLA-4. Thus, CHI3L1 contributes to pulmonary metastasis by inhibiting T cell co-stimulation and stimulating CTLA-4. The simultaneous targeting of CHI3L1 and the CTLA-4 axis with individual and, more powerfully with bispecific antibodies, represent promising therapeutic strategies for pulmonary metastasis.
Collapse
Affiliation(s)
- Bing Ma
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | - Suchitra Kamle
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | - Bedia Akosman
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | - Hina Khan
- Division of Hematology-Oncology, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Chang-Min Lee
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | - Chun Geun Lee
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | - Jack A. Elias
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
- Department of Medicine, Brown University, Providence, RI, United States
- *Correspondence: Jack A. Elias,
| |
Collapse
|
16
|
Zhang KS, Pelleg T, Campbell S, Rubio C, Loschner AL, Ie S. Pulmonary metastatic melanoma: current state of diagnostic imaging and treatments. Melanoma Manag 2021; 8:MMT58. [PMID: 34900220 PMCID: PMC8656320 DOI: 10.2217/mmt-2021-0001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/16/2021] [Indexed: 12/13/2022] Open
Abstract
Melanoma is the deadliest form of skin cancer with an estimated incidence of over 160,000 cases annually and about 41,000 melanoma-related deaths per year worldwide. Malignant melanoma (MM) primarily occurs in the skin but has been described in other organs. Although the respiratory system is generally afflicted by tumors such as lung cancer, it is also rarely affected by primary MM. The estimated incidence of pulmonary MM of the lung accounts for 0.01% of all primary lung tumors. The current understanding of pulmonary MM of the lung pathophysiology and its management are not well established. We aim to survey current clinical modalities with a focus on diagnostic imaging and therapeutic intervention to guide providers in the management of patients with a high index of suspicion.
Collapse
Affiliation(s)
- Kermit S Zhang
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - Tomer Pelleg
- Samaritan Health Services, Corvallis, OR 97330, USA
| | - Sabrina Campbell
- Department of Pulmonary & Sleep Medicine, Carilion Clinic, Roanoke, VA 24016, USA
| | - Catalina Rubio
- Department of Basic Science Education, Liberty University College of Osteopathic Medicine, Lynchburg, VA 24502, USA
| | - Anthony Lukas Loschner
- Samaritan Health Services, Corvallis, OR 97330, USA.,Department of Pulmonary & Sleep Medicine, Carilion Clinic, Roanoke, VA 24016, USA
| | - Susanti Ie
- Samaritan Health Services, Corvallis, OR 97330, USA.,Department of Pulmonary & Sleep Medicine, Carilion Clinic, Roanoke, VA 24016, USA
| |
Collapse
|
17
|
Yu JE, Yeo IJ, Son DJ, Yun J, Han SB, Hong JT. Anti-Chi3L1 antibody suppresses lung tumor growth and metastasis through inhibition of M2 polarization. Mol Oncol 2021; 16:2214-2234. [PMID: 34861103 PMCID: PMC9168758 DOI: 10.1002/1878-0261.13152] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/29/2021] [Accepted: 12/01/2021] [Indexed: 11/08/2022] Open
Abstract
Chitinase 3-like 1 (Chi3L1) is associated with various biological processes, such as inflammation, tissue repair, proliferation, cell survival, invasion, and extracellular matrix remodeling. Recent studies indicated that Chi3L1 is critical for cancer development and metastasis. In this study, we demonstrate that Chi3L1 serum and tissue levels were significantly increased in lung cancer patients compared with controls. We previously developed an anti-Chi3L1-humanized antibody, and here, we investigate its antitumor and antimetastatic effect. The anti-Chi3L1 antibody attenuated tumor growth and metastasis both in vitro and in vivo in a lung cancer mouse model. These inhibitory effects are associated with signal transducer and activator of transcription 6 (STAT6)-dependent M2 polarization inhibition. Proteomics analysis revealed that plasminogen (PLG) interacts with Chi3L1 and affects M2 polarization. Chi3L1 plays a critical role in lung cancer progression, and the anti-Chi3L1 antibody could be a new anticancer therapy.
Collapse
Affiliation(s)
- Ji Eun Yu
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju-si, Korea
| | - In Jun Yeo
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju-si, Korea
| | - Dong Ju Son
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju-si, Korea
| | - Jaesuk Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju-si, Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju-si, Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju-si, Korea
| |
Collapse
|
18
|
Lee YS, Yu JE, Kim KC, Lee DH, Son DJ, Lee HP, Jung JK, Kim ND, Ham YW, Yun J, Han SB, Hong JT. A small molecule targeting CHI3L1 inhibits lung metastasis by blocking IL-13Rα2-mediated JNK-AP-1 signals. Mol Oncol 2021; 16:508-526. [PMID: 34758182 PMCID: PMC8763653 DOI: 10.1002/1878-0261.13138] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 09/07/2021] [Accepted: 11/09/2021] [Indexed: 11/24/2022] Open
Abstract
Our previous big data analyses showed a high level of association between chitinase 3 like1 (CHI3L1) expression and lung tumor development. In the present study, we investigated whether a CHI3L1‐inhibiting chemical, 2‐({3‐[2‐(1‐cyclohexen‐1‐yl)ethyl]‐6,7‐dimethoxy‐4‐oxo‐3,4‐dihydro‐2‐quinazolinyl}sulfanyl)‐N‐(4‐ethylphenyl)butanamide (K284), could inhibit lung metastasis and studied its mechanism of action. We investigated the antitumor effect of K284 both in vitro and in vivo. K284 (0.5 mg·kg−1 body weight) significantly inhibited lung metastasis in in vivo models after injection of murine melanoma cells (B16F10) or adenocarcinomic human alveolar basal epithelial cells (A549). K284 significantly and concentration‐dependently also inhibited cancer cell proliferation and migration in the A549 and H460 lung cancer cell lines. We found that the binding of K284 to the chitin‐binding domain (CBD) of CHI3L1 prevented the binding of CHI3L1 to its receptor, interleukin‐13 receptor subunit alpha‐2 (IL‐13Rα2), thereby suppressing the CHI3L1 signal. This blocking of the CHI3L1‐IL‐13Rα2 signal caused the inhibition of c‐Jun N‐terminal kinase (JNK)‐activator protein 1 (AP‐1) signals, resulting in the prevention of lung metastasis and cancer cell growth. Our data demonstrate that K284 may serve as a potential candidate anticancer compound targeting CHI3L1.
Collapse
Affiliation(s)
- Yong Sun Lee
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Korea
| | - Ji Eun Yu
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Korea
| | - Ki Cheon Kim
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Korea
| | - Dong Hun Lee
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Korea
| | - Dong Ju Son
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Korea
| | - Hee Pom Lee
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Korea
| | - Jae-Kyung Jung
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Korea
| | | | - Young Wan Ham
- Department of Chemistry, Utah Valley University, Orem, UT, USA
| | - Jaesuk Yun
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Korea
| | - Sang-Bae Han
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Korea
| | - Jin Tae Hong
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Korea
| |
Collapse
|
19
|
Williams MM, Hafeez SA, Christenson JL, O’Neill KI, Hammond NG, Richer JK. Reversing an Oncogenic Epithelial-to-Mesenchymal Transition Program in Breast Cancer Reveals Actionable Immune Suppressive Pathways. Pharmaceuticals (Basel) 2021; 14:ph14111122. [PMID: 34832904 PMCID: PMC8622696 DOI: 10.3390/ph14111122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/16/2022] Open
Abstract
Approval of checkpoint inhibitors for treatment of metastatic triple negative breast cancer (mTNBC) has opened the door for the use of immunotherapies against this disease. However, not all patients with mTNBC respond to current immunotherapy approaches such as checkpoint inhibitors. Recent evidence demonstrates that TNBC metastases are more immune suppressed than primary tumors, suggesting that combination or additional immunotherapy strategies may be required to activate an anti-tumor immune attack at metastatic sites. To identify other immune suppressive mechanisms utilized by mTNBC, our group and others manipulated oncogenic epithelial-to-mesenchymal transition (EMT) programs in TNBC models to reveal differences between this breast cancer subtype and its more epithelial counterpart. This review will discuss how EMT modulation revealed several mechanisms, including tumor cell metabolism, cytokine milieu and secretion of additional immune modulators, by which mTNBC cells may suppress both the innate and adaptive anti-tumor immune responses. Many of these pathways/proteins are under preclinical or clinical investigation as therapeutic targets in mTNBC and other advanced cancers to enhance their response to chemotherapy and/or checkpoint inhibitors.
Collapse
|
20
|
Song X, Meng J, Yan G, Wang H, Li H, Lou D. Semaphorin 7A knockdown improves injury and prevents endothelial-to-mesenchymal transition in ox-LDL-induced HUVECs by regulating β1 integrin expression. Exp Ther Med 2021; 22:1441. [PMID: 34721683 PMCID: PMC8549106 DOI: 10.3892/etm.2021.10876] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 07/30/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is the most common cause of cardiovascular disease and is accompanied by high mortality rates and a poor prognosis. Semaphorin 7A (Sema7A) and its receptor β1 integrin have been reported to participate in the development of atherosclerosis. However, the role of Sema7A and β1 integrin in endothelial cell injury and endothelial-to-mesenchymal transition (EMT) in atherosclerosis remains undetermined, to the best of our knowledge. The mRNA and protein expression levels of Sema7A and β1 integrin in HUVECs were analyzed using reverse transcription-quantitative PCR (RT-qPCR) and western blot analyses, respectively. HUVECs were induced with 50 µg/ml oxidized low-density lipoprotein (ox-LDL) to establish an atherosclerosis cell model. Cell viability was measured using Cell Counting Kit-8 assay and the production of IL-1β, IL-6 and C-C motif chemokine ligand 2 was determined using ELISA. The expression levels of cell adhesion factors, intracellular adhesion molecule-1 and vascular cell adhesion molecule-1 were analyzed using RT-qPCR and western blot analyses. Cell apoptosis was detected using flow cytometry and western blotting. The levels of EMT-related markers were evaluated using RT-qPCR, western blotting and immunofluorescence staining. The results of the present study revealed that the expression levels of Sema7A and β1 integrin were significantly upregulated in ox-LDL-treated HUVECs. Treatment with ox-LDL significantly decreased cell viability, and increased the levels of inflammatory and adhesion factors, the cell apoptotic rate and the expression levels of EMT-related proteins. Knockdown of Sema7A reversed the ox-LDL-induced inflammatory responses and EMT, while the overexpression of β1 integrin reversed the Sema7A-mediated inhibitory effects on ox-LDL-treated HUVECs. In conclusion, the findings of the present study indicated that Sema7A and β1 integrin may play significant roles in atherosclerosis by mediating endothelial cell injury and EMT progression.
Collapse
Affiliation(s)
- Xiaoying Song
- Department of Geriatrics, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Jing Meng
- Department of Geriatrics, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Guoliang Yan
- Emergency Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Haihui Wang
- Emergency Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Haitao Li
- Emergency Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Danfei Lou
- Department of Geriatrics, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| |
Collapse
|
21
|
Ma B, Akosman B, Kamle S, Lee CM, He CH, Koo JS, Lee CG, Elias JA. CHI3L1 regulates PD-L1 and anti-CHI3L1-PD-1 antibody elicits synergistic antitumor responses. J Clin Invest 2021; 131:137750. [PMID: 34720089 DOI: 10.1172/jci137750] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 09/02/2021] [Indexed: 12/24/2022] Open
Abstract
Evasion of the immune response is a hallmark of cancer, and programmed cell death 1 (PD-1) and PD-1 ligand 1 (PD-L1) are major mediators of this immunosuppression. Chitinase 3-like 1 (CHI3L1) is induced in many cancers, where it portends a poor prognosis and contributes to tumor metastasis and spread. However, the mechanism(s) that CHI3L1 uses in metastasis have not been defined. Here we demonstrate that CHI3L1 regulates the expression of PD-L1, PD-L2, PD-1, LAG3, and TIM3 and plays a critical role in melanoma progression and lymphatic spread. CHI3L1 also contributed to IFN-γ-stimulated macrophage PD-L1 expression, and RIG-like helicase innate immunity suppressed CHI3L1, PD-L1, and melanoma progression. Individual antibodies against CHI3L1 or PD-1 had discrete antitumor effects and additive antitumor responses in metastasis models and T cell-tumor cell cocultures when administered simultaneously. Synergistic cytotoxic tumor cell death was seen in T cell-tumor cell cocultures, and significantly enhanced antitumor responses were seen in in vivo tumor models treated with bispecific antibodies that simultaneously target CHI3L1 and PD-1. CHI3L1 contributes to tumor progression by stimulating the PD-1/PD-L1 axis and other checkpoint molecules. The simultaneous targeting of CHI3L1 and the PD-1/PD-L1 axis with individual and, more powerfully, with bispecific antibodies represents a promising therapy for pulmonary metastasis and progression.
Collapse
Affiliation(s)
- Bing Ma
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Bedia Akosman
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Suchitra Kamle
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Chang-Min Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Chuan Hua He
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Ja Seok Koo
- Section of Medical Oncology, Department of Internal Medicine, Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Jack A Elias
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA.,Department of Medicine, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
22
|
Song Y, Wang L, Li J, Yang F, Gao Y, Song D, Sun J, Ye L, Zhang L, Huang D. The Expression of Semaphorin 7A in Human Periapical Lesions. J Endod 2021; 47:1631-1639. [PMID: 34126161 DOI: 10.1016/j.joen.2021.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 06/04/2021] [Accepted: 06/05/2021] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Semaphorin 7A (SEMA7A) is a membrane-bound or secretory protein exerting multiple functions in the regulation of inflammation, neural degradation, and cancer progression. Human periapical lesions are chronic and infectious diseases mainly caused by bacteria. However, the involvement of SEMA7A in human periapical lesions is still unclear. This study aimed to explore the expression of SEMA7A in human periapical lesions accompanied by the potential association of SEMA7A with matrix metalloproteinase (MMP)-1 and MMP-3 during the progression of apical periodontitis. METHODS Samples of periapical lesions and healthy controls were collected. Total RNA and protein were extracted respectively for quantitative real-time polymerase chain reaction and Western blot analysis. Additionally, 6 healthy samples and 27 periapical lesion samples were fixed, dehydrated, and embedded for further histologic and immunochemical analysis. The expression of SEMA7A was quantified by average integrated optical density. Immunofluorescence analysis was conducted to explore the colocalization of SEMA7A/MMP-1 and SEMA7A/MMP-3. RESULTS Compared with healthy controls, the messenger RNA and protein expression of SEMA7A was markedly up-regulated in periapical lesions. A stronger expression of MMP-1, MMP-3, and inflammatory cytokines was exhibited in periapical lesions than in healthy groups. An increasing expression of SEMA7A can be observed in both the periapical granuloma group and the radicular cyst group compared with the normal group (P < .01). Immunofluorescence results showed the colocalization of SEMA7A with both MMP-1 and MMP-3 in vascular vessels and extracellular matrix. CONCLUSIONS SEMA7A was up-regulated in periapical periodontitis and might be involved in the tissue destruction and infiltration of immune cells in periapical lesions.
Collapse
Affiliation(s)
- Yao Song
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liu Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiatong Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fan Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuxuan Gao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dongzhe Song
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jianxun Sun
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lan Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Dingming Huang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
23
|
Valentini E, Di Martile M, Del Bufalo D, D'Aguanno S. SEMAPHORINS and their receptors: focus on the crosstalk between melanoma and hypoxia. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:131. [PMID: 33858502 PMCID: PMC8050914 DOI: 10.1186/s13046-021-01929-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/25/2021] [Indexed: 12/11/2022]
Abstract
Hypoxia, a condition of oxygen deprivation, is considered a hallmark of tumor microenvironment regulating several pathways and promoting cancer progression and resistance to therapy. Semaphorins, a family of about 20 secreted, transmembrane and GPI-linked glycoproteins, and their cognate receptors (plexins and neuropilins) play a pivotal role in the crosstalk between cancer and stromal cells present in the tumor microenvironment. Many studies reported that some semaphorins are involved in the development of a permissive tumor niche, guiding cell-cell communication and, consequently, the development and progression, as well as the response to therapy, of different cancer histotypes, including melanoma. In this review we will summarize the state of art of semaphorins regulation by hypoxic condition in cancer with different origin. We will also describe evidence about the ability of semaphorins to affect the expression and activity of transcription factors activated by hypoxia, such as hypoxia-inducible factor-1. Finally, we will focus our attention on findings reporting the role of semaphorins in melanocytes transformation, melanoma progression and response to therapy. Further studies are necessary to understand the mechanisms through which semaphorins induce their effect and to shed light on the possibility to use semaphorins or their cognate receptors as prognostic markers and/or therapeutic targets in melanoma or other malignancies.
Collapse
Affiliation(s)
- Elisabetta Valentini
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Via Chianesi 53 (00144), Rome, Italy
| | - Marta Di Martile
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Via Chianesi 53 (00144), Rome, Italy
| | - Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Via Chianesi 53 (00144), Rome, Italy.
| | - Simona D'Aguanno
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Via Chianesi 53 (00144), Rome, Italy
| |
Collapse
|
24
|
Song Y, Wang L, Zhang L, Huang D. The involvement of semaphorin 7A in tumorigenic and immunoinflammatory regulation. J Cell Physiol 2021; 236:6235-6248. [PMID: 33611799 DOI: 10.1002/jcp.30340] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/12/2021] [Accepted: 02/05/2021] [Indexed: 02/04/2023]
Abstract
Semaphorins, a large group of highly conserved proteins, consist of eight subfamilies that are widely expressed in vertebrates, invertebrates, and viruses and exist in membrane-bound or secreted forms. First described as axon guidance cues during neurogenesis, semaphorins also perform physiological functions in other organ systems, such as bone homeostasis, immune response, and tumor progression. Semaphorin 7A (SEMA7A), also known as CDw108, is an immune semaphorin that modulates diverse immunoinflammatory processes, including immune cell interactions, inflammatory infiltration, and cytokine production. In addition, SEMA7A regulates the proliferation, migration, invasion, lymph formation, and angiogenesis of multiple types of tumor cells, and these effects are mediated by the interaction of SEMA7A with two specific receptors, PLXNC1 and integrins. Thus, SEMA7A is intimately related to the pathogenesis of multiple autoimmune and inflammation-related diseases and tumors. This review focuses on the role of SEMA7A in the pathogenesis of autoimmune disorders, inflammatory diseases, and tumors, as well as the underlying mechanisms. Furthermore, strategies targeting SEMA7A as a potential predictive, diagnostic, and therapeutic agent for these diseases are also addressed.
Collapse
Affiliation(s)
- Yao Song
- State Key Laboratory of Oral Diseases and National Clinical Research Center of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liu Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lan Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Dingming Huang
- State Key Laboratory of Oral Diseases and National Clinical Research Center of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
25
|
Gao R, Peng X, Perry C, Sun H, Ntokou A, Ryu C, Gomez JL, Reeves BC, Walia A, Kaminski N, Neumark N, Ishikawa G, Black KE, Hariri LP, Moore MW, Gulati M, Homer RJ, Greif DM, Eltzschig HK, Herzog EL. Macrophage-derived netrin-1 drives adrenergic nerve-associated lung fibrosis. J Clin Invest 2021; 131:136542. [PMID: 33393489 PMCID: PMC7773383 DOI: 10.1172/jci136542] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022] Open
Abstract
Fibrosis is a macrophage-driven process of uncontrolled extracellular matrix accumulation. Neuronal guidance proteins such as netrin-1 promote inflammatory scarring. We found that macrophage-derived netrin-1 stimulates fibrosis through its neuronal guidance functions. In mice, fibrosis due to inhaled bleomycin engendered netrin-1-expressing macrophages and fibroblasts, remodeled adrenergic nerves, and augmented noradrenaline. Cell-specific knockout mice showed that collagen accumulation, fibrotic histology, and nerve-associated endpoints required netrin-1 of macrophage but not fibroblast origin. Adrenergic denervation; haploinsufficiency of netrin-1's receptor, deleted in colorectal carcinoma; and therapeutic α1 adrenoreceptor antagonism improved collagen content and histology. An idiopathic pulmonary fibrosis (IPF) lung microarray data set showed increased netrin-1 expression. IPF lung tissues were enriched for netrin-1+ macrophages and noradrenaline. A longitudinal IPF cohort showed improved survival in patients prescribed α1 adrenoreceptor blockade. This work showed that macrophages stimulate lung fibrosis via netrin-1-driven adrenergic processes and introduced α1 blockers as a potentially new fibrotic therapy.
Collapse
Affiliation(s)
- Ruijuan Gao
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xueyan Peng
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Carrighan Perry
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Huanxing Sun
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Aglaia Ntokou
- Section of Cardiology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Changwan Ryu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jose L. Gomez
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Benjamin C. Reeves
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Anjali Walia
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Nir Neumark
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Genta Ishikawa
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Lida P. Hariri
- Division of Pulmonary and Critical Care Medicine, and
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Meagan W. Moore
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Mridu Gulati
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Robert J. Homer
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, and
| | - Daniel M. Greif
- Section of Cardiology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Holger K. Eltzschig
- Department of Anesthesiology, University of Texas at Houston Medical School, Houston, Texas, USA
| | - Erica L. Herzog
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, and
| |
Collapse
|
26
|
Williams MM, Spoelstra NS, Arnesen S, O'Neill KI, Christenson JL, Reese J, Torkko KC, Goodspeed A, Rosas E, Hanamura T, Sams SB, Li Z, Oesterreich S, Riggins RB, Jacobsen BM, Elias A, Gertz J, Richer JK. Steroid Hormone Receptor and Infiltrating Immune Cell Status Reveals Therapeutic Vulnerabilities of ESR1-Mutant Breast Cancer. Cancer Res 2020; 81:732-746. [PMID: 33184106 DOI: 10.1158/0008-5472.can-20-1200] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 09/17/2020] [Accepted: 11/09/2020] [Indexed: 11/16/2022]
Abstract
Mutations in ESR1 that confer constitutive estrogen receptor alpha (ER) activity in the absence of ligand are acquired by ≥40% of metastatic breast cancers (MBC) resistant to adjuvant aromatase inhibitor (AI) therapy. To identify targetable vulnerabilities in MBC, we examined steroid hormone receptors and tumor-infiltrating immune cells in metastatic lesions with or without ER mutations. ER and progesterone receptor (PR) were significantly lower in metastases with wild-type (WT) ER compared with those with mutant ER, suggesting that metastases that evade AI therapy by mechanism(s) other than acquiring ER mutations lose dependency on ER and PR. Metastases with mutant ER had significantly higher T regulatory and Th cells, total macrophages, and programmed death ligand-1 (PD-L1)-positive immune-suppressive macrophages than those with WT ER. Breast cancer cells with CRISPR-Cas9-edited ER (D538G, Y537S, or WT) and patient-derived xenografts harboring mutant or WT ER revealed genes and proteins elevated in mutant ER cells, including androgen receptor (AR), chitinase-3-like protein 1 (CHI3L1), and IFN-stimulated genes (ISG). Targeting these proteins blunted the selective advantage of ER-mutant tumor cells to survive estrogen deprivation, anchorage independence, and invasion. Thus, patients with mutant ER MBC might respond to standard-of-care fulvestrant or other selective ER degraders when combined with AR or CHI3L1 inhibition, perhaps with the addition of immunotherapy. SIGNIFICANCE: Targetable alterations in MBC, including AR, CHI3L1, and ISG, arise following estrogen-deprivation, and ER-mutant metastases may respond to immunotherapies due to elevated PD-L1+ macrophages.See related article by Arnesen et al., p. 539.
Collapse
Affiliation(s)
- Michelle M Williams
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Nicole S Spoelstra
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Spencer Arnesen
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Kathleen I O'Neill
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jessica L Christenson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jordan Reese
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Kathleen C Torkko
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Andrew Goodspeed
- Department of Pharmacology and University of Colorado Comprehensive Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Emmanuel Rosas
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Toru Hanamura
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sharon B Sams
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Zheqi Li
- Women's Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, Pittsburgh, Pennsylvania.,Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Steffi Oesterreich
- Women's Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, Pittsburgh, Pennsylvania.,Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rebecca B Riggins
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Britta M Jacobsen
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Anthony Elias
- School of Medicine, Division of Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jason Gertz
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Jennifer K Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
27
|
Glycoside hydrolase family 18 chitinases: The known and the unknown. Biotechnol Adv 2020; 43:107553. [DOI: 10.1016/j.biotechadv.2020.107553] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/09/2020] [Accepted: 04/20/2020] [Indexed: 12/13/2022]
|
28
|
Crump LS, Wyatt GL, Rutherford TR, Richer JK, Porter WW, Lyons TR. Hormonal Regulation of Semaphorin 7a in ER + Breast Cancer Drives Therapeutic Resistance. Cancer Res 2020; 81:187-198. [PMID: 33122307 DOI: 10.1158/0008-5472.can-20-1601] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/10/2020] [Accepted: 10/26/2020] [Indexed: 11/16/2022]
Abstract
Approximately 70% of all breast cancers are estrogen receptor-positive (ER+ breast cancer), and endocrine therapy has improved survival for patients with ER+ breast cancer. However, up to half of these tumors recur within 20 years. Recurrent ER+ breast cancers develop resistance to endocrine therapy; thus, novel targets are needed to treat recurrent ER+ breast cancer. Here we report that semaphorin 7A (SEMA7A) confers significantly decreased patient survival rates in ER+ breast cancer. SEMA7A was hormonally regulated in ER+ breast cancer, but its expression did not uniformly decrease with antiestrogen treatments. Additionally, overexpression of SEMA7A in ER+ cell lines drove increased in vitro growth in the presence of estrogen deprivation, tamoxifen, and fulvestrant. In vivo, SEMA7A conferred primary tumor resistance to fulvestrant and induced lung metastases. Prosurvival signaling was identified as a therapeutic vulnerability of ER+SEMA7A+ tumors. We therefore propose that targeting this pathway with inhibitors of survival signaling such as venetoclax may prove efficacious for treating SEMA7A+ tumors. SIGNIFICANCE: SEMA7A predicts for and likely contributes to poor response to standard-of-care therapies, suggesting that patients with SEMA7A+ER+ tumors may benefit from alternative therapeutic strategies. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/1/187/F1.large.jpg.
Collapse
Affiliation(s)
- Lyndsey S Crump
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Garhett L Wyatt
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | - Taylor R Rutherford
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jennifer K Richer
- Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Weston W Porter
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | - Traci R Lyons
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado. .,Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,University of Colorado Cancer Center, Aurora, Colorado
| |
Collapse
|
29
|
Xue YN, Xue YN, Wang ZC, Mo YZ, Wang PY, Tan WQ. A Novel Signature of 23 Immunity-Related Gene Pairs Is Prognostic of Cutaneous Melanoma. Front Immunol 2020; 11:576914. [PMID: 33193373 PMCID: PMC7604355 DOI: 10.3389/fimmu.2020.576914] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 09/29/2020] [Indexed: 01/11/2023] Open
Abstract
In this study, we aimed to identify an immune-related signature for predicting prognosis in cutaneous melanoma (CM). Sample data from The Cancer Genome Atlas (TCGA; n = 460) were used to develop a prognostic signature with 23 immune-related gene pairs (23 IRGPs) for CM. Patients were divided into high- and low-risk groups using the TCGA and validation datasets GSE65904 (n = 214), GSE59455 (n = 141), and GSE22153 (n = 79). The ability of the 23-IRGP signature to predict CM was precise, with the stratified high-risk groups showing a poor prognosis, and it had a significant predictive power when used for immune microenvironment and biological analyses. We subsequently established a novel promising prognostic model in CM to determine the association between the immune microenvironment and CM patient results. This approach may be used to discover signatures in other diseases while avoiding the technical biases associated with other platforms.
Collapse
Affiliation(s)
- Ya-Nan Xue
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi-Nan Xue
- Department of Biological Science, College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Zheng-Cai Wang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yong-Zhen Mo
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Pin-Yan Wang
- Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wei-Qiang Tan
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
30
|
He CH, Lee CG, Ma B, Kamle S, Choi AMK, Elias JA. N-Glycosylation Regulates Chitinase 3-like-1 and IL-13 Ligand Binding to IL-13 Receptor α2. Am J Respir Cell Mol Biol 2020; 63:386-395. [PMID: 32402213 DOI: 10.1165/rcmb.2019-0446oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Chitinase 3-like-1 (Chi3l1) and IL-13 are both ligands of IL-13 receptor α2 (IL-13Rα2). The binding of the former activates mitogen-activated protein kinase, AKT, and Wnt/β-catenin signaling, and plays important roles in innate and adaptive immunity, cellular apoptosis, oxidative injury, allergic inflammation, tumor metastasis and wound healing, fibrosis, and repair in the lung. In contrast, the latter binding is largely a decoy event that diminishes the effects of IL-13. Here, we demonstrate that IL-13Rα2 N-glycosylation is a critical determinant of which ligand binds. Structure-function evaluations demonstrated that Chi3l1-IL-13Rα2 binding was increased when sites of N-glycosylation are mutated, and studies with tunicamycin and Peptide:N-glycosidase F (PNGase F) demonstrated that Chi3l1-IL-13Rα2 binding and signaling were increased when N-glycosylation was diminished. In contrast, structure-function experiments demonstrated that IL-13 binding to IL-13Rα2 was dependent on each of the four sites of N-glycosylation in IL-13Rα2, and experiments with tunicamycin and PNGase F demonstrated that IL-13-IL-13Rα2 binding was decreased when IL-13Rα2 N-glycosylation was diminished. Studies with primary lung epithelial cells also demonstrated that Chi3l1 inhibited, whereas IL-13 stimulated, N-glycosylation as evidenced by the ability of Chi3l1 to inhibit and IL-13 to stimulate the subunits of the oligosaccharide complex A and B (STT3A and STT3B). These studies demonstrate that N-glycosylation is a critical determinant of Chi3l1 and IL-13 binding to IL-13Rα2, and highlight the ability of Chi3l1 and IL-13 to alter key elements of the N-glycosylation apparatus in a manner that would augment their respective binding.
Collapse
Affiliation(s)
- Chuan Hua He
- Department of Molecular Microbiology and Immunology and
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology and
| | - Bing Ma
- Department of Molecular Microbiology and Immunology and
| | | | - Augustine M K Choi
- Department of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York
| | - Jack A Elias
- Department of Molecular Microbiology and Immunology and.,Department of Medicine, Brown University, Providence, Rhode Island; and
| |
Collapse
|
31
|
Zhao T, Su Z, Li Y, Zhang X, You Q. Chitinase-3 like-protein-1 function and its role in diseases. Signal Transduct Target Ther 2020; 5:201. [PMID: 32929074 PMCID: PMC7490424 DOI: 10.1038/s41392-020-00303-7] [Citation(s) in RCA: 286] [Impact Index Per Article: 57.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/28/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022] Open
Abstract
Non-enzymatic chitinase-3 like-protein-1 (CHI3L1) belongs to glycoside hydrolase family 18. It binds to chitin, heparin, and hyaluronic acid, and is regulated by extracellular matrix changes, cytokines, growth factors, drugs, and stress. CHI3L1 is synthesized and secreted by a multitude of cells including macrophages, neutrophils, synoviocytes, chondrocytes, fibroblast-like cells, smooth muscle cells, and tumor cells. It plays a major role in tissue injury, inflammation, tissue repair, and remodeling responses. CHI3L1 has been strongly associated with diseases including asthma, arthritis, sepsis, diabetes, liver fibrosis, and coronary artery disease. Moreover, following its initial identification in the culture supernatant of the MG63 osteosarcoma cell line, CHI3L1 has been shown to be overexpressed in a wealth of both human cancers and animal tumor models. To date, interleukin-13 receptor subunit alpha-2, transmembrane protein 219, galectin-3, chemo-attractant receptor-homologous 2, and CD44 have been identified as CHI3L1 receptors. CHI3L1 signaling plays a critical role in cancer cell growth, proliferation, invasion, metastasis, angiogenesis, activation of tumor-associated macrophages, and Th2 polarization of CD4+ T cells. Interestingly, CHI3L1-based targeted therapy has been increasingly applied to the treatment of tumors including glioma and colon cancer as well as rheumatoid arthritis. This review summarizes the potential roles and mechanisms of CHI3L1 in oncogenesis and disease pathogenesis, then posits investigational strategies for targeted therapies.
Collapse
Affiliation(s)
- Ting Zhao
- Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, China
| | - Zhongping Su
- Department of Biotherapy, Department of Geriatrics, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yingchang Li
- Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes, Guangzhou Medical University, Guangzhou, China
| | - Xiaoren Zhang
- Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes, Guangzhou Medical University, Guangzhou, China
| | - Qiang You
- Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, China.
- Department of Biotherapy, Department of Geriatrics, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
- Key Laboratory of Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
32
|
Fard D, Tamagnone L. Semaphorins in health and disease. Cytokine Growth Factor Rev 2020; 57:55-63. [PMID: 32900601 DOI: 10.1016/j.cytogfr.2020.05.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 05/12/2020] [Indexed: 11/18/2022]
Abstract
Cell-cell communication is pivotal to guide embryo development, as well as to maintain adult tissues homeostasis and control immune response. Among extracellular factors responsible for this function, are the Semaphorins, a broad family of around 20 different molecular cues conserved in evolution and widely expressed in all tissues. The signaling cascades initiated by semaphorins depend on a family of conserved receptors, called Plexins, and on several additional molecules found in the receptor complexes. Moreover, multiple intracellular pathways have been described to act downstream of semaphorins, highlighting significant diversity in the signaling cascades controlled by this family. Notably, semaphorin expression is altered in many human diseases, such as immunopathologies, neurodegenerative diseases and cancer. This underscores the importance of semaphorins as regulatory factors in the tissue microenvironment and has prompted growing interest for assessing their potential relevance in medicine. This review article surveys the main contexts in which semaphorins have been found to regulate developing and healthy adult tissues, and the signaling cascades implicated in these functions. Vis a vis, we will highlight the main pathological processes in which semaphorins are thought to have a role thereof.
Collapse
Affiliation(s)
- Damon Fard
- University of Torino School of Medicine, Torino, Italy
| | - Luca Tamagnone
- Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy.
| |
Collapse
|
33
|
Tarullo SE, Hill RC, Hansen KC, Behbod F, Borges VF, Nelson AC, Lyons TR. Postpartum breast cancer progression is driven by semaphorin 7a-mediated invasion and survival. Oncogene 2020; 39:2772-2785. [PMID: 32020054 PMCID: PMC7103487 DOI: 10.1038/s41388-020-1192-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/13/2020] [Accepted: 01/24/2020] [Indexed: 11/09/2022]
Abstract
Young women diagnosed with breast cancer (BC) have poor prognosis due to increased rates of metastasis. In addition, women diagnosed within 10 years of most recent childbirth are approximately three times more likely to develop metastasis than age- and stage-matched nulliparous women. We define these cases as postpartum BC (PPBC) and propose that the unique biology of the postpartum mammary gland drives tumor progression. Our published results revealed roles for SEMA7A in breast tumor cell growth, motility, invasion, and tumor-associated lymphangiogenesis, all of which are also increased in preclinical models of PPBC. However, whether SEMA7A drives progression in PPBC remains largely unexplored. Our results presented herein show that silencing of SEMA7A decreases tumor growth in a model of PPBC, while overexpression is sufficient to increase growth in nulliparous hosts. Further, we show that SEMA7A promotes multiple known drivers of PPBC progression including tumor-associated COX-2 expression and fibroblast-mediated collagen deposition in the tumor microenvironment. In addition, we show for the first time that SEMA7A-expressing cells deposit fibronectin to promote tumor cell survival. Finally, we show that co-expression of SEMA7A/COX-2/FN predicts for poor prognosis in breast cancer patient cohorts. These studies suggest SEMA7A as a key mediator of BC progression, and that targeting SEMA7A may open avenues for novel therapeutic strategies.
Collapse
Affiliation(s)
- Sarah E Tarullo
- Division of Medical Oncology, Department of Medicine, CU Anschutz Medical Campus, Aurora, CO, 80045, USA
- Young Women's BC Translational Program, CU Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Ryan C Hill
- Department of Biochemistry and Molecular Genetics, CU Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, CU Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Fariba Behbod
- Division of Cancer and Developmental Biology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Virginia F Borges
- Division of Medical Oncology, Department of Medicine, CU Anschutz Medical Campus, Aurora, CO, 80045, USA
- Young Women's BC Translational Program, CU Anschutz Medical Campus, Aurora, CO, 80045, USA
- University of Colorado Cancer Center, Aurora, CO, 80045, USA
| | - Andrew C Nelson
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Traci R Lyons
- Division of Medical Oncology, Department of Medicine, CU Anschutz Medical Campus, Aurora, CO, 80045, USA.
- Young Women's BC Translational Program, CU Anschutz Medical Campus, Aurora, CO, 80045, USA.
- University of Colorado Cancer Center, Aurora, CO, 80045, USA.
| |
Collapse
|
34
|
Kzhyshkowska J, Larionova I, Liu T. YKL-39 as a Potential New Target for Anti-Angiogenic Therapy in Cancer. Front Immunol 2020; 10:2930. [PMID: 32038607 PMCID: PMC6988383 DOI: 10.3389/fimmu.2019.02930] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 11/28/2019] [Indexed: 12/20/2022] Open
Abstract
YKL-39 belongs to the evolutionarily conserved family of Glyco_18-containing proteins composed of chitinases and chitinase-like proteins. Chitinase-like proteins (CLPs) are secreted lectins that lack hydrolytic activity due to the amino acid substitutions in their catalytic domain and combine the functions of cytokines and growth factors. One of the major cellular sources that produce CLPs in various pathologies, including cancer, are macrophages. Monocytes recruited to the tumor site and programmed by tumor cells differentiate into tumor-associated macrophages (TAMs), which are the primary source of pro-angiogenic factors. Tumor angiogenesis is a crucial process for supplying rapidly growing tumors with essential nutrients and oxygen. We recently determined that YKL-39 is produced by tumor-associated macrophages in breast cancer. YKL-39 acts as a strong chemotactic factor for monocytes and stimulates angiogenesis. Chemotherapy is a common strategy to reduce tumor size and aggressiveness before surgical intervention, but chemoresistance, resulting in the relapse of tumors, is a common clinical problem that is critical for survival in cancer patients. Accumulating evidence indicates that TAMs are essential regulators of chemoresistance. We have recently found that elevated levels of YKL-39 expression are indicative of the efficiency of the metastatic process in patients who undergo neoadjuvant chemotherapy. We suggest YKL-39 as a new target for anti-angiogenic therapy that can be combined with neoadjuvant chemotherapy to reduce chemoresistance and inhibit metastasis in breast cancer patients.
Collapse
Affiliation(s)
- Julia Kzhyshkowska
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, University of Heidelberg, Mannheim, Germany
- German Red Cross Blood Service Baden-Württemberg—Hessen, Mannheim, Germany
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, Russia
| | - Irina Larionova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, Russia
- Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Tengfei Liu
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
35
|
Yeo IJ, Lee CK, Han SB, Yun J, Hong JT. Roles of chitinase 3-like 1 in the development of cancer, neurodegenerative diseases, and inflammatory diseases. Pharmacol Ther 2019; 203:107394. [PMID: 31356910 DOI: 10.1016/j.pharmthera.2019.107394] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2019] [Indexed: 02/07/2023]
Abstract
Chitinase 3-like 1 (CHI3L1) is a secreted glycoprotein that mediates inflammation, macrophage polarization, apoptosis, and carcinogenesis. The expression of CHI3L1 is strongly increased by various inflammatory and immunological conditions, including rheumatoid arthritis, multiple sclerosis, Alzheimer's disease, and several cancers. However, its physiological and pathophysiological roles in the development of cancer and neurodegenerative and inflammatory diseases remain unclear. Several studies have reported that CHI3L1 promotes cancer proliferation, inflammatory cytokine production, and microglial activation, and that multiple receptors, such as advanced glycation end product, syndecan-1/αVβ3, and IL-13Rα2, are involved. In addition, the pro-inflammatory action of CHI3L1 may be mediated via the protein kinase B and phosphoinositide-3 signaling pathways and responses to various pro-inflammatory cytokines, including tumor necrosis factor-α, interleukin-1β, interleukin-6, and interferon-γ. Therefore, CHI3L1 could contribute to a vast array of inflammatory diseases. In this article, we review recent findings regarding the roles of CHI3L1 and suggest therapeutic approaches targeting CHI3L1 in the development of cancers, neurodegenerative diseases, and inflammatory diseases.
Collapse
Affiliation(s)
- In Jun Yeo
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk 28160, Republic of Korea
| | - Chong-Kil Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk 28160, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk 28160, Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk 28160, Republic of Korea.
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk 28160, Republic of Korea.
| |
Collapse
|
36
|
Wallace TR, Tarullo SE, Crump LS, Lyons TR. Studies of postpartum mammary gland involution reveal novel pro-metastatic mechanisms. ACTA ACUST UNITED AC 2019; 5. [PMID: 30847405 PMCID: PMC6400586 DOI: 10.20517/2394-4722.2019.01] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Postpartum involution is the process by which the lactating mammary gland returns to the pre-pregnant state after weaning. Expression of tumor-promotional collagen, upregulation of matrix metalloproteinases, infiltration of M2 macrophages, and remodeling of blood and lymphatic vasculature are all characteristics shared by the involuting mammary gland and breast tumor microenvironment. The tumor promotional nature of the involuting mammary gland is perhaps best evidenced by cases of postpartum breast cancer (PPBC), or those cases diagnosed within 10 years of most recent childbirth. Women with PPBC experience more aggressive disease and higher risk of metastasis than nulliparous patients and those diagnosed outside the postpartum window. Semaphorin 7a (SEMA7A), cyclooxygenase-2 (COX-2), and collagen are all expressed in the involuting mammary gland and, together, predict for decreased metastasis free survival in breast cancer. Studies investigating the role of these proteins in involution have been important for understanding their contributions to PPBC. Postpartum involution thus represents a valuable model for the identification of novel molecular drivers of PPBC and classical cancer hallmarks. In this review, we will highlight the similarities between involution and cancer in the mammary gland, and further define the contribution of SEMA7A/COX-2/collagen interplay to postpartum involution and breast tumor progression and metastasis.
Collapse
Affiliation(s)
- Taylor R Wallace
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.,Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sarah E Tarullo
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.,Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Lyndsey S Crump
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.,Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Traci R Lyons
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.,Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.,University of Colorado Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
37
|
Kumar A, Zhang KYJ. Human Chitinases: Structure, Function, and Inhibitor Discovery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1142:221-251. [PMID: 31102249 DOI: 10.1007/978-981-13-7318-3_11] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chitinases are glycosyl hydrolases that hydrolyze the β-(1-4)-linkage of N-acetyl-D-glucosamine units present in chitin polymers. Chitinases are widely distributed enzymes and are present in a wide range of organisms including insects, plants, bacteria, fungi, and mammals. These enzymes play key roles in immunity, nutrition, pathogenicity, and arthropod molting. Humans express two chitinases, chitotriosidase 1 (CHIT1) and acid mammalian chitinase (AMCase) along with several chitinase-like proteins (CLPs). Human chitinases are reported to play a protective role against chitin-containing pathogens through their capability to degrade chitin present in the cell wall of pathogens. Now, human chitinases are gaining attention as the key players in innate immune response. Although the exact mechanism of their role in immune response is not known, studies in recent years begin to relate chitin recognition and degradation with the activation of signaling pathways involved in inflammation. The roles of both CHIT1 and AMCase in the development of various diseases have been revealed and several classes of inhibitors have been developed. However, a clear understanding could not be established due to complexities in the design of the right experiment for studying the role of human chitinase in various diseases. In this chapter, we will first outline the structural features of CHIT1 and AMcase. We will then review the progress in understanding the role of human chitinases in the development of various diseases. Finally, we will summarize the inhibitor discovery efforts targeting both CHIT1 and AMCase.
Collapse
Affiliation(s)
- Ashutosh Kumar
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan
| | - Kam Y J Zhang
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan.
| |
Collapse
|
38
|
Kinehara Y, Nagatomo I, Koyama S, Ito D, Nojima S, Kurebayashi R, Nakanishi Y, Suga Y, Nishijima-Futami Y, Osa A, Nakatani T, Kato Y, Nishide M, Hayama Y, Higashiguchi M, Morimura O, Miyake K, Kang S, Minami T, Hirata H, Iwahori K, Takimoto T, Takamatsu H, Takeda Y, Hosen N, Hoshino S, Shintani Y, Okumura M, Kumagai T, Nishino K, Imamura F, Nakatsuka SI, Kijima T, Kida H, Kumanogoh A. Semaphorin 7A promotes EGFR-TKI resistance in EGFR mutant lung adenocarcinoma cells. JCI Insight 2018; 3:123093. [PMID: 30568033 DOI: 10.1172/jci.insight.123093] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 11/06/2018] [Indexed: 12/17/2022] Open
Abstract
Although responses to EGFR tyrosine kinase inhibitors (EGFR-TKIs) are initially positive, 30%-40% of patients with EGFR-mutant tumors do not respond well to EGFR-TKIs, and most lung cancer patients harboring EGFR mutations experience relapse with resistance. Therefore, it is necessary to identify not only the mechanisms underlying EGFR-TKI resistance, but also potentially novel therapeutic targets and/or predictive biomarkers for EGFR-mutant lung adenocarcinoma. We found that the GPI-anchored protein semaphorin 7A (SEMA7A) is highly induced by the EGFR pathway, via mTOR signaling, and that expression levels of SEMA7A in human lung adenocarcinoma specimens were correlated with mTOR activation. Investigations using cell culture and animal models demonstrated that loss or overexpression of SEMA7A made cells less or more resistant to EGFR-TKIs, respectively. The resistance was due to the inhibition of apoptosis by aberrant activation of ERK. The ERK signal was suppressed by knockdown of integrin β1 (ITGB1). Furthermore, in patients with EGFR mutant tumors, higher SEMA7A expression in clinical samples predicted poorer response to EGFR-TKI treatment. Collectively, these data show that the SEMA7A-ITGB1 axis plays pivotal roles in EGFR-TKI resistance mediated by ERK activation and apoptosis inhibition. Moreover, our results reveal the potential utility of SEMA7A not only as a predictive biomarker, but also as a potentially novel therapeutic target in EGFR-mutant lung adenocarcinoma.
Collapse
Affiliation(s)
- Yuhei Kinehara
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Izumi Nagatomo
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shohei Koyama
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Daisuke Ito
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Satoshi Nojima
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan.,Department of Pathology, Osaka University Graduate school of Medicine, Suita, Osaka, Japan
| | - Ryota Kurebayashi
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshimitsu Nakanishi
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Yasuhiko Suga
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Yu Nishijima-Futami
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Akio Osa
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takeshi Nakatani
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Yasuhiro Kato
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Masayuki Nishide
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Yoshitomo Hayama
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Masayoshi Higashiguchi
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Osamu Morimura
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kotaro Miyake
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Sujin Kang
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan.,Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Toshiyuki Minami
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Division of Respiratory Medicine, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Haruhiko Hirata
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kota Iwahori
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takayuki Takimoto
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hyota Takamatsu
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Yoshito Takeda
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Naoki Hosen
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | | | - Yasushi Shintani
- Department of General Thoracic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Meinoshin Okumura
- Department of General Thoracic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | | | | | | | - Shin-Ichi Nakatsuka
- Department of Pathology, Osaka International Cancer Institute, Osaka, Osaka, Japan
| | - Takashi Kijima
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Division of Respiratory Medicine, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Hiroshi Kida
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| |
Collapse
|
39
|
Semaphorin 5A drives melanoma progression: role of Bcl-2, miR-204 and c-Myb. J Exp Clin Cancer Res 2018; 37:278. [PMID: 30454024 PMCID: PMC6245779 DOI: 10.1186/s13046-018-0933-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 10/18/2018] [Indexed: 11/17/2022] Open
Abstract
Background Melanoma, the most aggressive form of skin cancer, is characterized by high rates of metastasis, drug resistance and mortality. Here we investigated the role of Semaphorin 5A (Sema5A) on the properties associated with melanoma progression and the factors involved in Sema5A regulation. Methods Western blotting, qRT-PCR, Chromatin immunoprecipitation (ChIP) assay, immunohistochemistry of melanoma patient specimens and xenograft tissues, in vitro Transwell assay for cell migration and invasion evaluation, in vitro capillary-like structure formation analysis. Results A significant correlation of Sema5A mRNA expression and melanoma progression was observed by analyzing GEO profile dataset. Endogenous Sema5A protein was detected in 95% of human melanoma cell lines tested, in 70% of metastatic specimens from patients affected by melanoma, and 16% of in situ melanoma specimens showed a focal positivity. We demonstrated that Sema5A regulates in vitro cell migration and invasion and the formation of vasculogenic structures. We also found an increase of Sema5A at both mRNA and protein level after forced expression of Bcl-2. By use of transcriptional and proteasome inhibitors, we showed that Bcl-2 increases the stability of Sema5A mRNA and protein. Moreover, by ChIP we demonstrated that Sema5A expression is under the control of the transcription factor c-Myb and that c-Myb recruitment on Sema5A promoter is increased after Bcl-2 overexpression. Finally, a concomitant decrease in the expression of Sema5A, Bcl-2 and c-Myb proteins was observed in melanoma cells after miR-204 overexpression. Conclusion Overall our data provide evidences supporting the role of Sema5A in melanoma progression and the involvement of Bcl-2, miR-204 and c-Myb in regulating its expression. Electronic supplementary material The online version of this article (10.1186/s13046-018-0933-x) contains supplementary material, which is available to authorized users.
Collapse
|
40
|
Elder AM, Tamburini BAJ, Crump LS, Black SA, Wessells VM, Schedin PJ, Borges VF, Lyons TR. Semaphorin 7A Promotes Macrophage-Mediated Lymphatic Remodeling during Postpartum Mammary Gland Involution and in Breast Cancer. Cancer Res 2018; 78:6473-6485. [PMID: 30254150 PMCID: PMC6239927 DOI: 10.1158/0008-5472.can-18-1642] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 08/15/2018] [Accepted: 09/20/2018] [Indexed: 02/07/2023]
Abstract
Postpartum mammary gland involution is a tissue remodeling event that occurs in all mammals in the absence of nursing or after weaning to return the gland to the pre-pregnant state. The tissue microenvironment created by involution has proven to be tumor promotional. Here we report that the GPI-linked protein semaphorin 7A (SEMA7A) is expressed on mammary epithelial cells during involution and use preclinical models to demonstrate that tumors induced during involution express high levels of SEMA7A. Overexpression of SEMA7A promoted the presence of myeloid-derived podoplanin (PDPN)-expressing cells in the tumor microenvironment and during involution. SEMA7A drove the expression of PDPN in macrophages, which led to integrin- and PDPN-dependent motility and adherence to lymphatic endothelial cells to promote lymphangiogenesis. In support of this mechanism, mammary tissue from SEMA7A-knockout mice exhibited decreased myeloid-derived PDPN-expressing cells, PDPN-expressing endothelial cells, and lymphatic vessel density. Furthermore, coexpression of SEMA7A, PDPN, and macrophage marker CD68 predicted for decreased distant metastasis-free survival in a cohort of over 600 cases of breast cancer as well as in ovarian, lung, and gastric cancers. Together, our results indicate that SEMA7A may orchestrate macrophage-mediated lymphatic vessel remodeling, which in turn drives metastasis in breast cancer.Signficance: SEMA7A, which is expressed on mammary cells during glandular involution, alters macrophage biology and lymphangiogenesis to drive breast cancer metastasis. Cancer Res; 78(22); 6473-85. ©2018 AACR.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- Breast Neoplasms/pathology
- Cell Movement
- Crosses, Genetic
- Endothelial Cells/pathology
- Epithelial Cells/metabolism
- Female
- GPI-Linked Proteins/metabolism
- Humans
- Integrins/metabolism
- Lymphangiogenesis
- Lymphatic Vessels/pathology
- Macrophages/cytology
- Mammary Glands, Animal/metabolism
- Mammary Glands, Human/pathology
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasm Metastasis
- Postpartum Period
- Semaphorins/genetics
- Semaphorins/metabolism
- Tumor Microenvironment
Collapse
Affiliation(s)
- Alan M Elder
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, Colorado
| | - Beth A J Tamburini
- Division of Gastroenterology, University of Colorado School of Medicine, Aurora, Colorado
| | - Lyndsey S Crump
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, Colorado
| | - Sarah A Black
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, Colorado
| | - Veronica M Wessells
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, Colorado
| | - Pepper J Schedin
- University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, Colorado
- Department of Cell, Development and Cancer Biology, Oregon Health Sciences University, Oregon
| | - Virginia F Borges
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, Colorado
| | - Traci R Lyons
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
- University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, Colorado
| |
Collapse
|
41
|
Plexin C1 Marks Liver Cancer Cells with Epithelial Phenotype and Is Overexpressed in Hepatocellular Carcinoma. Can J Gastroenterol Hepatol 2018; 2018:4040787. [PMID: 30327758 PMCID: PMC6169229 DOI: 10.1155/2018/4040787] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 09/10/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND AIMS Hepatocellular carcinoma is an aggressive malignancy of the liver and is ranked as the sixth most common cancer worldwide. There is still room for novel markers to improve the diagnosis and monitoring of HCC. Our observations in cancer databases that PLXNC1 is upregulated in HCC led us to investigate the expression profile of Plexin C1 mRNA and protein in HCC cell lines and tissues. METHODS A recombinant protein encompassing part of the extracellular domain of Plexin C1 was used as an antigen for monoclonal antibody development. Transcript and protein levels of Plexin C1 in HCC cell lines were determined by RT-qPCR and Western blotting, respectively. In vivo evaluation of Plexin C1 expression in HCC tissues was accomplished by immunohistochemistry studies in tissue microarrays. RESULTS A monoclonal antibody, clone PE4, specific to Plexin C1, was generated. In silico and in vitro analyses revealed a Plexin C1-based clustering of well-differentiated HCC cell lines. Staining of HCC and nontumoral liver tissues with PE4 showed a membrane-localized overexpression of Plexin C1 in tumors (p=0.0118). In addition, this expression was correlated with the histological grades of HCC cases. CONCLUSIONS Plexin C1 distinguishes HCC cells of epithelial characteristics from those with the mesenchymal phenotype. Compared to the nontumoral liver, HCC tissues significantly overexpress Plexin C1. The newly generated PE4 antibody can be evaluated in larger HCC cohorts and might be exploited for the examination of Plexin C1 expression pattern in other epithelial malignancies.
Collapse
|
42
|
Geng B, Pan J, Zhao T, Ji J, Zhang C, Che Y, Yang J, Shi H, Li J, Zhou H, Mu X, Xu C, Wang C, Xu Y, Liu Z, Wen H, You Q. Chitinase 3-like 1-CD44 interaction promotes metastasis and epithelial-to-mesenchymal transition through β-catenin/Erk/Akt signaling in gastric cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:208. [PMID: 30165890 PMCID: PMC6117920 DOI: 10.1186/s13046-018-0876-2] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 08/13/2018] [Indexed: 01/14/2023]
Abstract
Background Enzymatically inactive chitinase-like protein CHI3L1 drives inflammatory response and promotes tumor progression. However, its role in gastric cancer (GC) tumorigenesis and metastasis has not yet been fully elucidated. We determined the significance of CHI3L1 expression in patients with GC. We also explored an as-yet unknown receptor of CHI3L1 and investigated the involved signaling in GC metastasis. Methods CHI3L1 expression was evaluated by immunoblotting, tissue microarray-based immunohistochemistry analysis (n = 100), and enzyme linked immunosorbent assay (ELISA) (n = 150). The interactions between CD44 and CHI3L1 or Interleukin-13 receptor alpha 2 (IL-13Rα2) were analyzed by co-immunoprecipitation, immunofluorescence co-localization assay, ELISA, and bio-layer interferometry. The roles of CHI3L1/CD44 axis in GC metastasis were investigated in GC cell lines and experimental animal model by gain and loss of function. Results CHI3L1 upregulation occurred during GC development, and positively correlated with GC invasion depth, lymph node status, and tumor staging. Mechanically, CHI3L1 binding to CD44 activated Erk and Akt, along with β-catenin signaling by phosphorylating β-catenin at Ser552 and Ser675. CD44 also interacted with IL-13Rα2 to form a complex. Notably, CD44v3 peptide and protein, but not CD44v6 peptide or CD44s protein, bound to both CHI3L1 and IL-13Rα2. Our in vivo and in vitro data further demonstrated that CHI3L1 promoted GC cell proliferation, migration, and metastasis. Conclusions CHI3L1 binding to CD44v3 activates Erk, Akt, and β-catenin signaling, therefore enhances GC metastasis. CHI3L1 expression is a novel biomarker for the prognosis of GC, and these findings have thus identified CHI3L1/CD44 axis as a vital pathway and potential therapeutic target in GC. Electronic supplementary material The online version of this article (10.1186/s13046-018-0876-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Biao Geng
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, 210011, Jiangsu, China.,Department of Respiratory Medicine, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui, China
| | - Jinshun Pan
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, 210011, Jiangsu, China
| | - Ting Zhao
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, 210011, Jiangsu, China
| | - Jie Ji
- First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chen Zhang
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, 210011, Jiangsu, China
| | - Ying Che
- Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jing Yang
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, 210011, Jiangsu, China
| | - Hui Shi
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Juan Li
- Cancer Medical Center, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hong Zhou
- Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xianmin Mu
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, 210011, Jiangsu, China
| | - Che Xu
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, 210011, Jiangsu, China
| | - Chao Wang
- Department of Surgery, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yue Xu
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, 210011, Jiangsu, China
| | - Zheng Liu
- Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao Wen
- Department of Surgery, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qiang You
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, 210011, Jiangsu, China. .,Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China. .,Cancer Medical Center, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China. .,Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu, China. .,Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
43
|
Kim KC, Yun J, Son DJ, Kim JY, Jung JK, Choi JS, Kim YR, Song JK, Kim SY, Kang SK, Shin DH, Roh YS, Han SB, Hong JT. Suppression of metastasis through inhibition of chitinase 3-like 1 expression by miR-125a-3p-mediated up-regulation of USF1. Am J Cancer Res 2018; 8:4409-4428. [PMID: 30214629 PMCID: PMC6134921 DOI: 10.7150/thno.26467] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 07/15/2018] [Indexed: 12/30/2022] Open
Abstract
Rationale: Chitinase 3-like 1 (Chi3L1) protein is up-regulated in various diseases including solid cancers. According to Genome-Wide Association Study (GWAS)/Online Mendelian Inheritance in Man (OMIM)/Differentially Expressed Gene (DEG) analyses, Chi3L1 is associated with 38 cancers, and more highly associated with cancer compared to other oncogenes such as EGFR, TNFα, etc. However, the mechanisms and pathways by which Chi3L1 is associated with cancer are not clear. In current study, we investigated the role of Chi3L1 in lung metastasis. Methods: We performed the differentially expressed gene analysis to explore the genes which are associated with Chi3L1 using the web-based platform from Biomart. We investigated the metastases in lung tissues of C57BL/6 mice injected with B16F10 melanoma following treatment with Ad-shChi3L1. We also investigated the expression of USF1 and Chi3L1 in Chi3L1 KD mice lung tissues by Western blotting and IHC. We also analyzed lung cancer cells metastases induced by Chi3L1 using migration and cell proliferation assay in human lung cancer cell lines. The involvement of miR-125a-3p in Chi3L1 regulation was determined by miRNA qPCR and luciferase reporter assay. Results: We showed that melanoma metastasis in lung tissues was significantly reduced in Chi3L1 knock-down mice, accompanied by down-regulation of MMP-9, MMP-13, VEGF, and PCNA in Chi3L1 knock-down mice lung tissue, as well as in human lung cancer cell lines. We also found that USF1 was conversely expressed against Chi3L1. USF1 was increased by knock-down of Chi3L1 in mice lung tissues, as well as in human lung cancer cell lines. In addition, knock-down of USF1 increased Chi3L1 levels in addition to augmenting metastasis cell migration and proliferation in mice model, as well as in human cancer cell lines. Moreover, in human lung tumor tissues, the expression of Chi3L1 was increased but USF1 was decreased in a stage-dependent manner. Finally, Chi3L1 expression was strongly regulated by the indirect translational suppressing activity of USF1 through induction of miR-125a-3p, a target of Chi3L1. Conclusion: Metastases in mice lung tissues and human lung cancer cell lines were decreased by KD of Chi3L1. USF1 bound to the Chi3L1 promoter, however, Chi3L1 expression was decreased by USF1, despite USF1 enhancing the transcriptional activity of Chi3L1. We found that USF1 induced miR-125a-3p levels which suppressed Chi3L1 expression. Ultimately, our results suggest that lung metastasis is suppressed by knock-down of Chi3L1 through miR-125a-3p-mediated up-regulation of USF1.
Collapse
|
44
|
Wang Z, Wang X, Zhou H, Dan X, Jiang L, Wu Y. Long non-coding RNA CASC2 inhibits tumorigenesis via the miR-181a/PLXNC1 axis in melanoma. Acta Biochim Biophys Sin (Shanghai) 2018. [PMID: 29514220 DOI: 10.1093/abbs/gmx148] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Melanoma is the most malignant and aggressive form of skin carcinoma originating in the pigment-producing melanocytes. In this study, to further investigate the molecular mechanisms of the development and progression of melanoma, we explored the impacts of long non-coding RNA (lncRNA) CASC2 on melanoma cell functions. Microarray analysis was carried out to identify the expression of lncRNA CASC2 in melanoma cells. MiR-181a was predicted as a sponging target of CASC2 by miRcode, while the 3'-UTR of Plexin C1 (PLXNC1) was a potential target of miR-181a according to the TargetScan database. The correlation among CASC2, miR-181a, and PLXNC1 was verified by dual luciferase reporter assay and qRT-PCR. After manipulation of CASC2, miR-181a and PLXNC1 expression with transfection in A375 and M14 cells, cell viability, apoptosis, and invasive ability were evaluated using CCK-8, flow cytometry and Transwell assays, respectively. A low expression of CASC2 was detected in melanoma tissues and cells. Dual luciferase reporting assay confirmed that miR-181a targeted the 3'-UTR of PLXNC1. Furthermore, CASC2 could efficiently sponge miR-181a, thereby facilitating the expression of PLXNC1. Up-regulation of CASC2 suppressed the cell proliferation and invasion, but induced the apoptosis of melanoma cells. Our results demonstrated that lncRNA CASC2 can promote PLXNC1 expression by sponging miR-181a, thereby inhibiting the proliferation and invasion of melanoma cells, indicating that lncRNA CASC2 functions via the miR-181a/PLXNC1 axis in melanoma.
Collapse
Affiliation(s)
- Zhiqiong Wang
- Department of Dermatology, The First People's Hospital of Yunnan Province, Kunming 650032, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - Xiaochuan Wang
- Department of Dermatology, The First People's Hospital of Yunnan Province, Kunming 650032, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - Hongying Zhou
- Department of Dermatology, The First People's Hospital of Yunnan Province, Kunming 650032, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - Xiao Dan
- Department of Dermatology, The First People's Hospital of Yunnan Province, Kunming 650032, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - Lixiang Jiang
- Department of Dermatology, The First People's Hospital of Yunnan Province, Kunming 650032, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - Yifei Wu
- Department of Dermatology, The First People's Hospital of Yunnan Province, Kunming 650032, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| |
Collapse
|
45
|
Pouyafar A, Heydarabad MZ, Mahboob S, Mokhtarzadeh A, Rahbarghazi R. Angiogenic potential of YKL-40 in the dynamics of tumor niche. Biomed Pharmacother 2018; 100:478-485. [PMID: 29477911 DOI: 10.1016/j.biopha.2018.02.050] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/09/2018] [Accepted: 02/13/2018] [Indexed: 12/20/2022] Open
Abstract
A multitude of clinical studies showed the elevation of YKL-40 in subjects with different kinds of tumors. It is predicted that an inherent correlation exists between survivals of cancer patients with total YKL-40 serum levels, making this factor as a potential novel biomarker. However, the crucial role of YKL-40 in the dynamics of cancers, especially angiogenesis, has not yet been completely addressed. In this review, we highlighted the various facets of YKL-40 and its importance in cancer biology as a bio-shuttle in gene therapy.
Collapse
Affiliation(s)
- Ayda Pouyafar
- Department of Biochemistry, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Milad Zadi Heydarabad
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soltanali Mahboob
- Department of Biochemistry, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Department of Biochemistry, Higher Education Institute of Rab-Rashid, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
46
|
Kim DH, Park HJ, Lim S, Koo JH, Lee HG, Choi JO, Oh JH, Ha SJ, Kang MJ, Lee CM, Lee CG, Elias JA, Choi JM. Regulation of chitinase-3-like-1 in T cell elicits Th1 and cytotoxic responses to inhibit lung metastasis. Nat Commun 2018; 9:503. [PMID: 29403003 PMCID: PMC5799380 DOI: 10.1038/s41467-017-02731-6] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 12/20/2017] [Indexed: 01/30/2023] Open
Abstract
Chitinase-3-like-1 (Chi3l1) is known to play a significant role in the pathogenesis of Type 2 inflammation and cancer. However, the function of Chi3l1 in T cell and its clinical implications are largely unknown. Here we show that Chi3l1 expression was increased in activated T cells, especially in Th2 cells. In addition, Chi3l1-deficient T cells are hyper-responsive to TcR stimulation and are prone to differentiating into Th1 cells. Chi3l1-deficient Th1 cells show increased expression of anti-tumor immunity genes and decreased Th1 negative regulators. Deletion of Chi3l1 in T cells in mice show reduced melanoma lung metastasis with increased IFNγ and TNFα-producing T cells in the lung. Furthermore, silencing of Chi3l1 expression in the lung using peptide-siRNA complex (dNP2-siChi3l1) efficiently inhibit lung metastasis with enhanced Th1 and CTL responses. Collectively, this study demonstrates Chi3l1 is a regulator of Th1 and CTL which could be a therapeutic target to enhance anti-tumor immunity.
Collapse
Affiliation(s)
- Do-Hyun Kim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea
| | - Hong-Jai Park
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Sangho Lim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea
| | - Ja-Hyun Koo
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea
| | - Hong-Gyun Lee
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea
| | - Jin Ouk Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea
| | - Ji Hoon Oh
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Min-Jong Kang
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Chang-Min Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, USA
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, USA
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, 04763, Korea
| | - Jack A Elias
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, USA
- Division of Medical and Biological Sciences, Warren Alpert Medical School, Brown University, Providence, RI, 02903, USA
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea.
- Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea.
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon, 16419, Korea.
| |
Collapse
|
47
|
Hu S, Liu Y, You T, Heath J, Xu L, Zheng X, Wang A, Wang Y, Li F, Yang F, Cao Y, Zhang H, van Gils JM, van Zonneveld AJ, Jo H, Wu Q, Zhang Y, Tang C, Zhu L. Vascular Semaphorin 7A Upregulation by Disturbed Flow Promotes Atherosclerosis Through Endothelial β1 Integrin. Arterioscler Thromb Vasc Biol 2017; 38:335-343. [PMID: 29269512 DOI: 10.1161/atvbaha.117.310491] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 12/06/2017] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Accumulating evidence suggests a role of semaphorins in vascular homeostasis. Here, we investigate the role of Sema7A (semaphorin 7A) in atherosclerosis and its underlying mechanism. APPROACH AND RESULTS Using genetically engineered Sema7A-/-ApoE-/- mice, we showed that deletion of Sema7A attenuates atherosclerotic plaque formation primarily in the aorta of ApoE-/- mice on a high-fat diet. A higher level of Sema7A in the atheroprone lesser curvature suggests a correlation of Sema7A with disturbed flow. This notion is supported by elevated Sema7A expression in human umbilical venous endothelial cells either subjected to oscillatory shear stress or treated with the PKA (protein kinase A)/CREB (cAMP response element-binding protein) inhibitor H89 (N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinolinesulfonamide·2HCl hydrate). Further studies using the partial carotid artery ligation model showed that disturbed flow in the left carotid artery of Sema7A+/+ApoE-/- mice promoted the expression of endothelial Sema7A and cell adhesion molecules, leukocyte adhesion, and plaque formation, whereas such changes were attenuated in Sema7A-/-ApoE-/- mice. Further studies showed that blockage of β1 integrin, a known Sema7A receptor, or inhibition of FAK (focal adhesion kinase), MEK1/2 (mitogen-activated protein kinase kinase 1/2), or NF-κB (nuclear factor-κB) significantly reduced the expression of cell adhesion molecules and THP-1 (human acute monocytic leukemia cell line) monocyte adhesion in Sema7A-overexpressing human umbilical venous endothelial cells. Studies using chimeric mice suggest that vascular, most likely endothelial, Sema7A plays a major role in atherogenesis. CONCLUSIONS Our findings indicate a significant role of Sema7A in atherosclerosis by mediating endothelial dysfunction in a β1 integrin-dependent manner.
Collapse
Affiliation(s)
- Shuhong Hu
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Yifei Liu
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Tao You
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Jack Heath
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Linru Xu
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Xiaowei Zheng
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Aili Wang
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Yinyan Wang
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Fengchan Li
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Fei Yang
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Yiren Cao
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Huayu Zhang
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Janine M van Gils
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Anton Jan van Zonneveld
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Hanjoong Jo
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Qingyu Wu
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Yonghong Zhang
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.)
| | - Chaojun Tang
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.).
| | - Li Zhu
- From the Cyrus Tang Hematology Center (S.H., Y.L., T.Y., L.X., Y.W., F.L., F.Y., Y.C., Q.W., C.T., L.Z.), Department of Epidemiology, School of Public Health (X.Z., A.W., Y.Z.), Collaborative Innovation Center of Hematology of Jiangsu Province (S.H., Y.L., T.Y., Q.W., C.T., L.Z.), and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases (X.Z., A.W., Q.W., Y.Z.), Soochow University, Suzhou, China; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (J.H., H.J.); Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, the Netherlands (H.Z., J.M.v.G., A.J.v.Z.); and Department of Molecular Cardiology, Cleveland Clinic, OH (Q.W.).
| |
Collapse
|
48
|
Garcia-Areas R, Libreros S, Simoes M, Castro-Silva C, Gazaniga N, Amat S, Jaczewska J, Keating P, Schilling K, Brito M, Wojcikiewicz EP, Iragavarpu-Charyulu V. Suppression of tumor-derived Semaphorin 7A and genetic ablation of host-derived Semaphorin 7A impairs tumor progression in a murine model of advanced breast carcinoma. Int J Oncol 2017; 51:1395-1404. [PMID: 29048670 PMCID: PMC5642386 DOI: 10.3892/ijo.2017.4144] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 07/31/2017] [Indexed: 12/12/2022] Open
Abstract
Solid tumors can generate a plethora of neurogenesis-related molecules that enhance their growth and metastasis. Among them, we have identified axonal guidance molecule Semaphorin 7A (SEMA7A) in breast cancer. The goal of this study was to determine the therapeutic effect of suppressing SEMA7A levels in the 4T1 murine model of advanced breast carcinoma. We used anti-SEMA7A short hairpin RNA (shRNA) to gene silence SEMA7A in 4T1 mammary tumor cells. When implanted into the mammary fat pads of syngeneic mice, SEMA7A shRNA-expressing 4T1 tumors exhibited decreased growth rates, deferred metastasis and reduced mortality. In vitro, SEMA7A shRNA-expressing 4T1 cells had weakened proliferative, migratory and invasive abilities, and decreased levels of mesenchymal factors. Atomic force microscopy studies showed that SEMA7A shRNA-expressing 4T1 cells had an increase in cell stiffness that corresponded with their decreased malignant potential. Genetic ablation of host-derived SEMA7A further enhanced the antitumor effects of SEMA7A shRNA gene silencing in 4T1 cells. Our preclinical findings demonstrate a critical role for SEMA7A in mediating mammary tumor progression.
Collapse
Affiliation(s)
- R Garcia-Areas
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - S Libreros
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - M Simoes
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - C Castro-Silva
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - N Gazaniga
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - S Amat
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - J Jaczewska
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - P Keating
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - K Schilling
- Lynn Women's Health & Wellness Institute, Boca Raton Regional Hospital, Boca Raton, FL 33431, USA
| | - M Brito
- Department of Pathology, Boca Raton Regional Hospital, Boca Raton, FL 33431, USA
| | - E P Wojcikiewicz
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - V Iragavarpu-Charyulu
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| |
Collapse
|
49
|
Hamilton G, Rath B. Circulating tumor cell interactions with macrophages: implications for biology and treatment. Transl Lung Cancer Res 2017; 6:418-430. [PMID: 28904886 DOI: 10.21037/tlcr.2017.07.04] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer and metastasis are closely associated with inflammation. Macrophages are important effector cells in enhancing tumor proliferation, invasion and providing protection against the immune system. Despite advanced knowledge of tumor-macrophage interactions, the role of macrophages in emergence and invasion of circulating tumor cells (CTCs) is not known. A series of six CTC cell lines have been derived from blood of patients with extensive disease small cell lung cancer (ED-SCLC) in our lab, most likely representing a homogenous cell population of the actual metastasis-initiating cells (MIC) of CTCs. SCLC has an unfavorable prognosis due to rapid dissemination and early chemoresistant relapses. SCLC CTCs recruit macrophages and elicit secretion of various cytokines and the six CTC lines express chitinase-3-like-1 (CHI3L1), vascular endothelial growth factor (VEGF) and matrix metalloproteinase-9 (MMP9) in abundance. CHI3L1 is cytokine/growth factor expressed in inflammation and cancer and found to be correlated to metastasis and a dismal prognosis. In conclusion, SCLC CTCs have acquired the essential means for aggressiveness and invasion in a tumor microenvironment specifically shaped by macrophages and inflammation.
Collapse
Affiliation(s)
- Gerhard Hamilton
- Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Barbara Rath
- Department of Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
50
|
Erturk K, Tas F, Serilmez M, Bilgin E, Yasasever V. Clinical Signifıcance of Serum Ykl-40 (Chitinase-3-Like-1 Protein) as a Biomarker in Melanoma: an Analysis of 112 Turkish Patients. Asian Pac J Cancer Prev 2017; 18:1383-1387. [PMID: 28612591 PMCID: PMC5555551 DOI: 10.22034/apjcp.2017.18.5.1383] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Background: Angiogenesis plays an essential role in tumor growth and serum levels of YKL-40 , a strong angiogenic
factor that promotes tumor vessel development, has been found to be elevated in various cancers. We here investigated
correlation between melanoma parameters and serum YKL-40 levels, to assess potential diagnostic, prognostic and
predictive values. Material and Methods: Data for 112 pathologically confirmed cutaneous melanomas of any stage
were examined retrospectively. ELISA assays were used to measure serum YKL-40 in plasma samples. Results: The
baseline serum YKL-40 levels were significantly higher in patients than healthy controls (174.88 vs 120.10 ng/mL,
p<0.001). However, values did not correlate with clinicopathological parameters, (p>0.05), and furthermore there was
no apparent prognostic influence on melanoma survival (HR: 1.568; 95% CI, 0.580-3.051; p=0.838). Conclusion:
Serum YKL-40 can be useful for diagnosis of melanoma, but reliability in assessing prognosis is questionable. We
believe that efforts should be made to understand the interaction between YKL-40 and the tumor environment, and
establish whether it might be the target for treatment of malignancies.
Collapse
Affiliation(s)
- Kayhan Erturk
- Medical Oncology,Istanbul University, Istanbul, Turkey.
| | | | | | | | | |
Collapse
|