1
|
Kobayashi H, Asano T, Suzuki H, Tanaka T, Yoshikawa T, Kaneko MK, Kato Y. Establishment of a Sensitive Monoclonal Antibody Against Mouse CCR9 (C 9Mab-24) for Flow Cytometry. Monoclon Antib Immunodiagn Immunother 2023; 42:15-21. [PMID: 36516144 DOI: 10.1089/mab.2022.0032] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The CC chemokine receptor 9 (CCR9), also known as CD199, is one of chemokine receptors. The CC chemokine ligand 25 (CCL25) is known to be the only ligand for CCR9. The CCR9-CCL25 interaction plays important roles in chemotaxis of lymphocytes and tumor cell migration. Therefore, CCR9-CCL25 axis is a promising target for tumor therapy and diagnosis. In this study, we established a sensitive and specific monoclonal antibody (mAb) against mouse CCR9 (mCCR9) using N-terminal peptide immunization method. The established anti-mCCR9 mAb, C9Mab-24 (rat immunoglobulin [IgG]2a, kappa), reacted with mCCR9-overexpressed Chinese hamster ovary-K1 (CHO/mCCR9) and mCCR9-endogenously expressed cell line, RL2, through flow cytometry. Kinetic analyses using flow cytometry showed that the dissociation constants (KD) of C9Mab-24 for CHO/mCCR9 and RL2 cell lines were 6.0 × 10-9 M and 4.7 × 10-10 M, respectively. Results indicated that C9Mab-24 is useful for detecting mCCR9 through flow cytometry, thereby providing a possibility for targeting mCCR9-expressing cells in vivo experiments.
Collapse
Affiliation(s)
- Hiyori Kobayashi
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Teizo Asano
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroyuki Suzuki
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takeo Yoshikawa
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mika K Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yukinari Kato
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
2
|
Ganguly K, Shah A, Atri P, Rauth S, Ponnusamy MP, Kumar S, Batra SK. Chemokine-mucinome interplay in shaping the heterogeneous tumor microenvironment of pancreatic cancer. Semin Cancer Biol 2022; 86:511-520. [PMID: 35346803 PMCID: PMC9793394 DOI: 10.1016/j.semcancer.2022.03.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/15/2022] [Accepted: 03/23/2022] [Indexed: 02/06/2023]
Abstract
Pancreatic cancer (PC) is exemplified by a complex immune-suppressive, fibrotic tumor microenvironment (TME), and aberrant expression of mucins. The constant crosstalk between cancer cells, cancer-associated fibroblasts (CAFs), and the immune cells mediated by the soluble factors and inflammatory mediators including cytokines, chemokines, reactive oxygen species (ROS) promote the dynamic temporal switch towards an immune-escape phenotype in the neoplastic cells and its microenvironment that bolsters disease progression. Chemokines have been studied in PC pathogenesis, albeit poorly in the context of mucins, tumor glycocalyx, and TME heterogeneity (CAFs and immune cells). With correlative analysis from PC patients' transcriptome data, support from available literature, and scientific arguments-based speculative extrapolations in terms of disease pathogenesis, we have summarized in this review a comprehensive understanding of chemokine-mucinome interplay during stromal modulation and immune-suppression in PC. Future studies should focus on deciphering the complexities of chemokine-mediated control of glycocalyx maturation, immune infiltration, and CAF-associated immune suppression. Knowledge extracted from such studies will be beneficial to mechanistically correlate the mucin-chemokine abundance in serum versus pancreatic tumors of patients, which may aid in prognostication and stratification of PC patients for immunotherapy.
Collapse
Affiliation(s)
- Koelina Ganguly
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ashu Shah
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Pranita Atri
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sanchita Rauth
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
3
|
Santamaria S, Delgado M, Botas M, Castellano E, Corraliza-Gorjon I, Lafuente P, Muñoz-Calleja C, Toribio M, Kremer L, Garcia-Sanz JA. Therapeutic potential of an anti-CCR9 mAb evidenced in xenografts of human CCR9+ tumors. Front Immunol 2022; 13:825635. [PMID: 35967322 PMCID: PMC9363564 DOI: 10.3389/fimmu.2022.825635] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
Relapsed or refractory T acute lymphoblastic leukemia (T-ALL) still carries poor prognosis. Aiming to improve outcomes, the therapeutic potential of an anti-CCR9 monoclonal antibody (mAb 92R), targeting the human chemokine-receptor CCR9 is analyzed on orthotopic xenotransplants. 92R mAb treatment of mice carrying human CCR9+ T-ALL cell lines or primary T cell leukemias inhibits tumor growth and increases survival. The therapeutic effects of 92R are specific and synergize with chemotherapeutic agents increasing survival. Furthermore, 92R decreases size of non-hematopoietic tumors with a forced CCR9 expression and of solid tumors generated by the pancreatic adenocarcinoma cell line AsPC-1. In addition, a humanized version of 92R mAb (Srb1) is also able to inhibit growth of CCR9+ T-ALL tumor cells in vivo, increasing survival 2.66-fold. Finally, 92R mAb prevents liver accumulation of infiltrates and reduces tumor cell numbers in already formed infiltrates. Thus, the humanized version of 92R mAb (Srb1), displays therapeutic potential for CCR9+ tumor treatment and might represent one of the first therapeutic antibodies for precision medicine on T-ALL patients.
Collapse
Affiliation(s)
- Silvia Santamaria
- Centro de Investigaciones Biologicas Margarita Salas (CIB-CSIC), Department of Molecular Medicine, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Marisa Delgado
- Centro de Investigaciones Biologicas Margarita Salas (CIB-CSIC), Department of Molecular Medicine, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Marta Botas
- Centro de Investigaciones Biologicas Margarita Salas (CIB-CSIC), Department of Molecular Medicine, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Eva Castellano
- Centro de Investigaciones Biologicas Margarita Salas (CIB-CSIC), Department of Molecular Medicine, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Isabel Corraliza-Gorjon
- Centro Nacional de Biotecnología (CNB-CSIC), Department of Immunology and Oncology, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Paloma Lafuente
- Centro de Investigaciones Biologicas Margarita Salas (CIB-CSIC), Department of Molecular Medicine, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Cecilia Muñoz-Calleja
- Servicio de Inmunología, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Maria L. Toribio
- Centro de Biología Molecular Severo Ochoa (CBMSO-CSIC-UAM), Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Leonor Kremer
- Centro Nacional de Biotecnología (CNB-CSIC), Department of Immunology and Oncology, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- *Correspondence: Jose A. Garcia-Sanz, ; Leonor Kremer,
| | - Jose A. Garcia-Sanz
- Centro de Investigaciones Biologicas Margarita Salas (CIB-CSIC), Department of Molecular Medicine, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- *Correspondence: Jose A. Garcia-Sanz, ; Leonor Kremer,
| |
Collapse
|
4
|
Murray ER, Menezes S, Henry JC, Williams JL, Alba-Castellón L, Baskaran P, Quétier I, Desai A, Marshall JJT, Rosewell I, Tatari M, Rajeeve V, Khan F, Wang J, Kotantaki P, Tyler EJ, Singh N, Reader CS, Carter EP, Hodivala-Dilke K, Grose RP, Kocher HM, Gavara N, Pearce O, Cutillas P, Marshall JF, Cameron AJM. Disruption of pancreatic stellate cell myofibroblast phenotype promotes pancreatic tumor invasion. Cell Rep 2022; 38:110227. [PMID: 35081338 PMCID: PMC8810397 DOI: 10.1016/j.celrep.2021.110227] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 10/18/2021] [Accepted: 12/15/2021] [Indexed: 12/13/2022] Open
Abstract
In pancreatic ductal adenocarcinoma (PDAC), differentiation of pancreatic stellate cells (PSCs) into myofibroblast-like cancer-associated fibroblasts (CAFs) can both promote and suppress tumor progression. Here, we show that the Rho effector protein kinase N2 (PKN2) is critical for PSC myofibroblast differentiation. Loss of PKN2 is associated with reduced PSC proliferation, contractility, and alpha-smooth muscle actin (α-SMA) stress fibers. In spheroid co-cultures with PDAC cells, loss of PKN2 prevents PSC invasion but, counter-intuitively, promotes invasive cancer cell outgrowth. PKN2 deletion induces a myofibroblast to inflammatory CAF switch in the PSC matrisome signature both in vitro and in vivo. Further, deletion of PKN2 in the pancreatic stroma induces more locally invasive, orthotopic pancreatic tumors. Finally, we demonstrate that a PKN2KO matrisome signature predicts poor outcome in pancreatic and other solid human cancers. Our data indicate that suppressing PSC myofibroblast function can limit important stromal tumor-suppressive mechanisms, while promoting a switch to a cancer-supporting CAF phenotype.
Collapse
Affiliation(s)
- Elizabeth R Murray
- Kinase Biology Laboratory, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Shinelle Menezes
- Kinase Biology Laboratory, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Jack C Henry
- Kinase Biology Laboratory, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Josie L Williams
- Kinase Biology Laboratory, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Lorena Alba-Castellón
- Kinase Biology Laboratory, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Priththivika Baskaran
- Kinase Biology Laboratory, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Ivan Quétier
- Kinase Biology Laboratory, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Ami Desai
- Kinase Biology Laboratory, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Jacqueline J T Marshall
- Protein Phosphorylation Laboratory, Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Ian Rosewell
- Transgenic Services, Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Marianthi Tatari
- Barts Cancer Institute, Queen Mary, University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Vinothini Rajeeve
- Barts Cancer Institute, Queen Mary, University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Faraz Khan
- Barts Cancer Institute, Queen Mary, University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Jun Wang
- Barts Cancer Institute, Queen Mary, University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Panoraia Kotantaki
- Barts Cancer Institute, Queen Mary, University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Eleanor J Tyler
- Barts Cancer Institute, Queen Mary, University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Namrata Singh
- Kinase Biology Laboratory, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Claire S Reader
- Barts Cancer Institute, Queen Mary, University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Edward P Carter
- Barts Cancer Institute, Queen Mary, University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Kairbaan Hodivala-Dilke
- Barts Cancer Institute, Queen Mary, University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Richard P Grose
- Barts Cancer Institute, Queen Mary, University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Hemant M Kocher
- Barts Cancer Institute, Queen Mary, University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK; Barts and the London HPB Centre, The Royal London Hospital, Barts Health NHS Trust, Whitechapel, London E1 1BB, UK
| | - Nuria Gavara
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Oliver Pearce
- Barts Cancer Institute, Queen Mary, University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Pedro Cutillas
- Barts Cancer Institute, Queen Mary, University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - John F Marshall
- Barts Cancer Institute, Queen Mary, University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Angus J M Cameron
- Kinase Biology Laboratory, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
5
|
Nanamiya R, Takei J, Asano T, Tanaka T, Sano M, Nakamura T, Yanaka M, Hosono H, Kaneko MK, Kato Y. Development of Anti-Human CC Chemokine Receptor 9 Monoclonal Antibodies for Flow Cytometry. Monoclon Antib Immunodiagn Immunother 2021; 40:101-106. [PMID: 34161159 DOI: 10.1089/mab.2021.0007] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
CC chemokine receptor 9 (CCR9) belongs to the beta chemokine receptor family and is mainly distributed on the surface of immature T lymphocytes and enterocytes. This receptor is highly expressed in rheumatoid arthritis, colitis, type 2 diabetes, and various tumors. Therefore, more sensitive monoclonal antibodies (mAbs) need to be developed to predict the prognosis of many high CCR9 expression diseases. Because CCR9 is a structurally unstable G protein-coupled receptor, it has been difficult to develop anti-CCR9 mAbs using the traditional method. This study developed anti-human CCR9 (hCCR9) mAbs for flow cytometry using a Cell-Based Immunization and Screening (CBIS) method. Two mice were immunized with hCCR9-overexpressed Chinese hamster ovary (CHO)-K1 cells (CHO/hCCR9), and hybridomas showing strong signals from CHO/hCCR9 and no signals from CHO-K1 cells were selected by flow cytometry. We established an anti-hCCR9 mAb, C9Mab-1 (IgG1, kappa), which detected hCCR9 in MOLT-4 leukemia T lymphoblast cells and CHO/hCCR9 cells by flow cytometry. Our study showed that an anti-hCCR9 mAb was developed more rapidly by the CBIS method than the previous method.
Collapse
Affiliation(s)
- Ren Nanamiya
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Junko Takei
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Teizo Asano
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masato Sano
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takuro Nakamura
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Miyuki Yanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hideki Hosono
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mika K Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan.,New Industry Creation Hatchery Center, Tohoku University, Sendai, Japan
| |
Collapse
|
6
|
Umar S, Palasiewicz K, Van Raemdonck K, Volin MV, Romay B, Ahmad I, Tetali C, Sweiss N, Amin MA, Zomorrodi RK, Shahrara S. CCL25 and CCR9 is a unique pathway that potentiates pannus formation by remodeling RA macrophages into mature osteoclasts. Eur J Immunol 2021; 51:903-914. [PMID: 33347617 PMCID: PMC10041658 DOI: 10.1002/eji.202048681] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 10/26/2020] [Accepted: 12/18/2020] [Indexed: 12/18/2022]
Abstract
This study elucidates the mechanism of CCL25 and CCR9 in rheumatoid arthritis (RA). RA synovial fluid (SF) expresses elevated levels of CCL25 compared to OA SF and plasma from RA and normal. CCL25 was released into RA SF by fibroblasts (FLS) and macrophages (MΦs) stimulated with IL-1β and IL-6. CCR9 is also presented on IL-1β and IL-6 activated RA FLS and differentiated MΦs. Conversely, in RA PBMCs neither CCL25 nor CCR9 are impacted by 3-month longitudinal TNF inhibitor therapy. CCL25 amplifies RA FLS and monocyte infiltration via p38 and ERK phosphorylation. CCL25-stimulated RA FLS secrete potentiated levels of IL-8 which is disrupted by p38 and ERK inhibitors. CCL25 polarizes RA monocytes into nontraditional M1 MΦs that produce IL-8 and CCL2. Activation of p38 and ERK cascades are also responsible for the CCL25-induced M1 MΦ development. Unexpectedly, CCL25 was unable to polarize RA PBMCs into effector Th1/Th17 cells. Consistently, lymphokine like RANKL was uninvolved in CCL25-induced osteoclastogenesis; however, this manifestation was regulated by osteoclastic factors such as RANK, cathepsin K (CTSK), and TNF-α. In short, we reveal that CCL25/CCR9 manipulates RA FLS and MΦ migration and inflammatory phenotype in addition to osteoclast formation via p38 and ERK activation.
Collapse
Affiliation(s)
- Sadiq Umar
- Jesse Brown VA Medical Center, Chicago, IL
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL
| | - Karol Palasiewicz
- Jesse Brown VA Medical Center, Chicago, IL
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL
| | - Katrien Van Raemdonck
- Jesse Brown VA Medical Center, Chicago, IL
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL
| | - Michael V. Volin
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL
| | - Bianca Romay
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL
| | - Imran Ahmad
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL
| | - Chandana Tetali
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL
| | - Nadera Sweiss
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL
| | - M Asif Amin
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, MI 481096
| | - Ryan K Zomorrodi
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL
| | - Shiva Shahrara
- Jesse Brown VA Medical Center, Chicago, IL
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL
| |
Collapse
|
7
|
CCL25 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1302:99-111. [PMID: 34286444 DOI: 10.1007/978-3-030-62658-7_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Multiple checkpoint mechanisms are overridden by cancer cells in order to develop into a tumor. Neoplastic cells, while constantly changing during the course of cancer progression, also craft their surroundings to meet their growing needs. This crafting involves changing cell surface receptors, affecting response to extracellular signals and secretion of signals that affect the nearby cells and extracellular matrix architecture. This chapter briefly comprehends the non-cancer cells facilitating the cancer growth and elaborates on the notable role of the CCR9-CCL25 chemokine axis in shaping the tumor microenvironment (TME), directly and via immune cells. Association of increased CCR9 and CCL25 levels in various tumors has demonstrated the significance of this axis as a tool commonly used by cancer to flourish. It is involved in attracting immune cells in the tumor and determining their fate via various direct and indirect mechanisms and, leaning the TME toward immunosuppressive state. Besides, elevated CCR9-CCL25 signaling allows survival and rapid proliferation of cancer cells in an otherwise repressive environment. It modulates the intra- and extracellular protein matrix to instigate tumor dissemination and creates a supportive metastatic niche at the secondary sites. Lastly, this chapter abridges the latest research efforts and challenges in using the CCR9-CCL25 axis as a cancer-specific target.
Collapse
|
8
|
Korbecki J, Grochans S, Gutowska I, Barczak K, Baranowska-Bosiacka I. CC Chemokines in a Tumor: A Review of Pro-Cancer and Anti-Cancer Properties of Receptors CCR5, CCR6, CCR7, CCR8, CCR9, and CCR10 Ligands. Int J Mol Sci 2020; 21:ijms21207619. [PMID: 33076281 PMCID: PMC7590012 DOI: 10.3390/ijms21207619] [Citation(s) in RCA: 195] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/05/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
CC chemokines (or β-chemokines) are 28 chemotactic cytokines with an N-terminal CC domain that play an important role in immune system cells, such as CD4+ and CD8+ lymphocytes, dendritic cells, eosinophils, macrophages, monocytes, and NK cells, as well in neoplasia. In this review, we discuss human CC motif chemokine ligands: CCL1, CCL3, CCL4, CCL5, CCL18, CCL19, CCL20, CCL21, CCL25, CCL27, and CCL28 (CC motif chemokine receptor CCR5, CCR6, CCR7, CCR8, CCR9, and CCR10 ligands). We present their functioning in human physiology and in neoplasia, including their role in the proliferation, apoptosis resistance, drug resistance, migration, and invasion of cancer cells. We discuss the significance of chemokine receptors in organ-specific metastasis, as well as the influence of each chemokine on the recruitment of various cells to the tumor niche, such as cancer-associated fibroblasts (CAF), Kupffer cells, myeloid-derived suppressor cells (MDSC), osteoclasts, tumor-associated macrophages (TAM), tumor-infiltrating lymphocytes (TIL), and regulatory T cells (Treg). Finally, we show how the effect of the chemokines on vascular endothelial cells and lymphatic endothelial cells leads to angiogenesis and lymphangiogenesis.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (S.G.)
| | - Szymon Grochans
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (S.G.)
| | - Izabela Gutowska
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72 Av., 70-111 Szczecin, Poland;
| | - Katarzyna Barczak
- Department of Conservative Dentistry and Endodontics, Pomeranian Medical University, Powstańców Wlkp. 72 Av., 70-111 Szczecin, Poland;
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (S.G.)
- Correspondence: ; Tel.: +48-914661515
| |
Collapse
|
9
|
Xu B, Deng C, Wu X, Ji T, Zhao L, Han Y, Yang W, Qi Y, Wang Z, Yang Z, Yang Y. CCR9 and CCL25: A review of their roles in tumor promotion. J Cell Physiol 2020; 235:9121-9132. [PMID: 32401349 DOI: 10.1002/jcp.29782] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/25/2020] [Accepted: 05/02/2020] [Indexed: 12/16/2022]
Abstract
Chemokines constitute a superfamily of small chemotactic cytokines with functions that are based on interactions with their corresponding receptors. It has been found that, among other functions, chemokines regulate the migratory and invasive abilities of cancer cells. Multiple studies have confirmed that chemokine receptor 9 (CCR9) and its exclusive ligand, chemokine 25 (CCL25), are overexpressed in a variety of malignant tumors and are closely associated with tumor proliferation, apoptosis, invasion, migration and drug resistance. This review evaluates recent advances in understanding the role of CCR9/CCL25 in cancer development. First, we outline the general background of chemokines in cancer and the structure and function of CCR9 and CCL25. Next, we describe the basic function of CCR9/CCL25 in the cancer process. Then, we introduce the role of CCR9/CCL25 and related signaling pathways in various cancers. Finally, future research directions are proposed. In general, this paper is intended to serve as a comprehensive repository of information on this topic and is expected to contribute to the design of other research projects and future efforts to develop treatment strategies for ameliorating the effects of CCR9/CCL25 in cancer.
Collapse
Affiliation(s)
- Baoping Xu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Chao Deng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xue Wu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Ting Ji
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Lin Zhao
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yuehu Han
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wenwen Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Yating Qi
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Zheng Wang
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, Wuhan, China
| | - Zhi Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an, China
| |
Collapse
|
10
|
Kamata K, Watanabe T, Minaga K, Hara A, Yoshikawa T, Okamoto A, Yamao K, Takenaka M, Park AM, Kudo M. Intestinal dysbiosis mediates experimental autoimmune pancreatitis via activation of plasmacytoid dendritic cells. Int Immunol 2020; 31:795-809. [PMID: 31287532 DOI: 10.1093/intimm/dxz050] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 06/27/2019] [Indexed: 12/13/2022] Open
Abstract
Autoimmune pancreatitis (AIP) is a pancreatic manifestation of a newly proposed disease entity, IgG4-related disease (IgG4-RD), characterized by enhanced IgG4 antibody responses and involvement of multiple organs. We have previously reported that innate immune activation contributes to the development of AIP and IgG4-RD, as these diseases are characterized by the production of IFN-α and IL-33 by plasmacytoid dendritic cells (pDCs) that mediate chronic fibroinflammatory responses. In this study, we investigated the roles played by innate immunity against intestinal microflora in experimental AIP induced in MRL/MpJ mice by repeated administrations of 100 µg of polyinosinic-polycytidylic acid [poly (I:C)]. Bowel sterilization with a broad spectrum of antibiotics inhibited pancreatic accumulation of pDCs producing IFN-α and IL-33, and thereby suppressed the development of AIP. Mice treated with 10 µg of poly (I:C) developed severe AIP equivalent to that induced by 100 µg of poly (I:C) upon co-housing with mice treated with 100 µg of poly (I:C). Fecal microbiota transplantation (FMT) from donor mice treated with 100 µg of poly (I:C) led to the development of severe AIP in the recipient mice upon injection with 10 µg of poly (I:C). Induction of severe AIP in mice with 10 µg of poly (I:C) was associated with pancreatic accumulation of pDCs producing IFN-α and IL-33 in the co-housing and FMT experiments. These data collectively suggest that innate immune responses against intestinal microflora are involved in the development of experimental AIP, and that intestinal dysbiosis increases sensitivity to experimental AIP via activation of pDCs.
Collapse
Affiliation(s)
- Ken Kamata
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Tomohiro Watanabe
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Kosuke Minaga
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Akane Hara
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Tomoe Yoshikawa
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Ayana Okamoto
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Kentaro Yamao
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Mamoru Takenaka
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Ah-Mee Park
- Department of Microbiology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| |
Collapse
|
11
|
Chen L, Zhang S, Shen Y, Qi L, Zhang Z, Tian H, Zou Z. Thymus‑expressed chemokine secreted by breast cancer cells promotes metastasis and inhibits apoptosis. Oncol Rep 2020; 43:1875-1884. [PMID: 32323823 PMCID: PMC7160534 DOI: 10.3892/or.2020.7575] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 03/10/2020] [Indexed: 12/12/2022] Open
Abstract
The aim of the present study was to investigate the underlying mechanisms of thymus-expressed chemokine (TECK) autocrine signaling, and its effect on carcinogenesis and the development of breast cancer. The present study also assessed epithelial-mensenchymal transition (EMT) and cell migration, invasion, proliferation and apoptosis. Breast cancer cell lines MCF-7 and MDA-MB-231 were used in the present study, and TECK basic expression in cancer cells was investigated using western blotting (WB). EMT markers, Akt pathway molecules and apoptosis indicators were detected by reverse transcription-quantitative PCR or WB. In order to assess migration and invasion, wound healing and Matrigel invasion assays were performed. Moreover, flow cytometry was used to assess the rate of proliferation and apoptosis. In vivo experiments were conducted in nude mice to assess cancer growth. It was revealed that breast cancer cells could secrete TECK in an autocrine manner. Furthermore, TECK could increase cell migration and invasion by promoting EMT and inhibit apoptosis via the Akt signaling pathway.
Collapse
Affiliation(s)
- Lu Chen
- Department of Anatomy, Histology and Embryology, School of Basic Medical Science, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Shuming Zhang
- Department of Radiotherapy, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, Shandong 250014, P.R. China
| | - Yaqian Shen
- Department of Anatomy, Histology and Embryology, School of Basic Medical Science, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Linzeng Qi
- Department of Anatomy, Histology and Embryology, School of Basic Medical Science, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Zhaolin Zhang
- Department of Special Examination, Penglai People's Hospital, Penglai, Shandong 265600, P.R. China
| | - Hua Tian
- Department of Anatomy, Histology and Embryology, School of Basic Medical Science, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Zhigeng Zou
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
12
|
Lu L, Du H, Huang H, Wang C, Wang P, Zha Z, Wu Y, Liu X, Weng C, Fang X, Li B, Mao H, Wang L, Guan M, Liu G. CCR9 Promotes Migration and Invasion of Lung Adenocarcinoma Cancer Stem Cells. Int J Med Sci 2020; 17:912-920. [PMID: 32308544 PMCID: PMC7163367 DOI: 10.7150/ijms.40864] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 02/22/2020] [Indexed: 12/24/2022] Open
Abstract
Aim: CC chemokine receptor 9 (CCR9) interacts with its exclusive ligand CCL25, resulting in promoting tumor progression and metastasis. However, the effect and mechanisms of CCR9 on lung adenocarcinoma distant metastasis remain largely unknown. To preliminary clarify the underlying mechanisms, we investigate the correlation between CCR9 and ALDH1A1+cancer stem cells (CSCs), as well as the effect of CCR9 on the migration and invasion of CSCs. Methods: Immunohistochemistry was performed to detect the expression of CCR9 in lung adenocarcinoma tissues. The correlations of CCR9 with distant metastasis and overall survival were investigated. Serial paraffin-embedded tissue blocks were used to detect ALDH1A1+CSCs expression. The correlations between CCR9 expression and ALDH1A1+CSCs were evaluated. We further studied the effect of CCR9/CCL25 on the migration and invasion of CSCs using transwell assays. Results: There were positive correlations between CCR9 expression and distant metastasis, as well as poor overall survival. Patients with high CCR9 expression were more likely to develop distant metastasis and demonstrated poorer overall survival than patients with low CCR9 expression. In addition, there was positive correlation between the expression of CCR9 and ALDH1A1 in the same tumor microenvironment. ALDHhigh CSCs demonstrated enhanced expression of CCR9 than ALDHlow cells. Further transwell assays demonstrated that the numbers of CSCs migrated or invaded in response to CCL25 were more than that without CCL25 stimulation. Additional application of anti-CCR9 antibody reversed the CCL25-induced migration and invasion of CSCs. Conclusions: In summary, our study demonstrated that CCR9/CCL25 promoted the migration and invasion of CSCs, which might contribute to distant metastasis and poor overall survival. Our findings provided evidence that CCR9/CCL25 could be used as novel therapeutic targets for lung adenocarcinoma.
Collapse
Affiliation(s)
- Lin Lu
- Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China, 510180.,Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Huan Du
- Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China, 510180
| | - Haowei Huang
- Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China, 510180
| | - Chenxi Wang
- Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China, 510180
| | - Peipei Wang
- Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China, 510180
| | - Zhiqiang Zha
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Yong Wu
- Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China, 510180.,Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Xia Liu
- Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China, 510180.,Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Chengyin Weng
- Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China, 510180.,Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Xisheng Fang
- Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China, 510180.,Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Baoxiu Li
- Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China, 510180.,Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Haibo Mao
- Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China, 510180.,Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Lina Wang
- Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China, 510180.,Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Mingmei Guan
- Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China, 510180.,Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Guolong Liu
- Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China, 510180.,Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
13
|
Wang C, Liu Z, Xu Z, Wu X, Zhang D, Zhang Z, Wei J. The role of chemokine receptor 9/chemokine ligand 25 signaling: From immune cells to cancer cells. Oncol Lett 2018; 16:2071-2077. [PMID: 30008902 PMCID: PMC6036326 DOI: 10.3892/ol.2018.8896] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 05/15/2018] [Indexed: 02/05/2023] Open
Abstract
Chemokine ligand 25 (CCL25) and chemokine receptor 9 (CCR9) are important regulators of migration, proliferation and apoptosis in leukocytes and cancer cells. Blocking of the CCR9/CCL25 signal has been demonstrated to be a potential novel cancer therapy. Research into CCR9 and CCL25 has revealed their associated upstream and downstream signaling pathways; CCR9 is regulated by several immunological factors, including NOTCH, interleukin 2, interleukin 4 and retinoic acid. NOTCH in particular, has been revealed to be a crucial upstream regulator of CCR9. Furthermore, proteins including matrix metalloproteinases, P-glycoprotein, Ezrin/Radixin/Moesin and Livin are regulated via phosphatidylinositol-3 kinase/protein kinase B, which are in turn stimulated by CCR9/CCL25. This is a review of the current literature on the functions and signaling pathways of CCR9/CCL25.
Collapse
Affiliation(s)
- Cong Wang
- Department of Hepatopancreatobiliary Surgery, Affiliated Hospital of Qinghai University, Xining, Qinghai 810001, P.R. China
| | - Zhenghuan Liu
- Department of Urology, West China School of Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zhihui Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, P.R. China
| | - Xian Wu
- Department of Ultrasound, West China School of Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Dongyang Zhang
- Department of Ultrasound, West China School of Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ziqi Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, P.R. China
| | - Jianqin Wei
- The University of Miami Leonard M. Miller School of Medicine, University of Miami, Coral Gables, FL 33136, USA
| |
Collapse
|
14
|
Somovilla-Crespo B, Martín Monzón MT, Vela M, Corraliza-Gorjón I, Santamaria S, Garcia-Sanz JA, Kremer L. 92R Monoclonal Antibody Inhibits Human CCR9 + Leukemia Cells Growth in NSG Mice Xenografts. Front Immunol 2018; 9:77. [PMID: 29434597 PMCID: PMC5797297 DOI: 10.3389/fimmu.2018.00077] [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] [Received: 09/11/2017] [Accepted: 01/11/2018] [Indexed: 12/19/2022] Open
Abstract
CCR9 is as an interesting target for the treatment of human CCR9+-T cell acute lymphoblastic leukemia, since its expression is limited to immature cells in the thymus, infiltrating leukocytes in the small intestine and a small fraction of mature circulating T lymphocytes. 92R, a new mouse mAb (IgG2a isotype), was raised using the A-isoform of hCCR9 as immunogen. Its initial characterization demonstrates that binds with high affinity to the CCR9 N-terminal domain, competing with the previously described 91R mAb for receptor binding. 92R inhibits human CCR9+ tumor growth in T and B-cell deficient Rag2−/− mice. In vitro assays suggested complement-dependent cytotoxicity and antibody-dependent cell-mediated cytotoxicity as possible in vivo mechanisms of action. Unexpectedly, 92R strongly inhibited tumor growth also in a model with compromised NK and complement activities, suggesting that other mechanisms, including phagocytosis or apoptosis, might also be playing a role on 92R-mediated tumor elimination. Taken together, these data contribute to strengthen the hypothesis of the immune system’s opportunistic nature.
Collapse
Affiliation(s)
- Beatriz Somovilla-Crespo
- Department of Immunology and Oncology, Centro Nacional de Biotecnologia (CNB-CSIC), Madrid, Spain
| | | | - Maria Vela
- Department of Immunology and Oncology, Centro Nacional de Biotecnologia (CNB-CSIC), Madrid, Spain
| | - Isabel Corraliza-Gorjón
- Department of Immunology and Oncology, Centro Nacional de Biotecnologia (CNB-CSIC), Madrid, Spain
| | - Silvia Santamaria
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biologicas (CIB-CSIC), Madrid, Spain
| | - Jose A Garcia-Sanz
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biologicas (CIB-CSIC), Madrid, Spain
| | - Leonor Kremer
- Department of Immunology and Oncology, Centro Nacional de Biotecnologia (CNB-CSIC), Madrid, Spain.,Protein Tools Unit, Centro Nacional de Biotecnologia (CNB-CSIC), Madrid, Spain
| |
Collapse
|
15
|
Hu Y, Ma A, Lin S, Yang Y, Hong G. Novel peptide screened from a phage display library antagonizes the activity of CC chemokine receptor 9. Oncol Lett 2017; 14:6471-6476. [PMID: 29163684 PMCID: PMC5686441 DOI: 10.3892/ol.2017.7065] [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: 09/29/2016] [Accepted: 08/08/2017] [Indexed: 11/29/2022] Open
Abstract
CC chemokine receptor 9 (CCR9) serves a role in the drug resistance and metastasis of tumors. In the present study, a peptide specifically bound to CCR9 was obtained and the effect on tumor cells was observed. A Ph.D.-12 phage display peptide library was used to screen for peptides binding specifically to the second extracellular loop of CCR9. The ratios of the input and output of phage clones increased gradually following three rounds of biopanning. A total of 8 positive phage clones were identified from DNA analysis. A phage clone, C-4, was identified which exhibited higher affinity and specificity for the second extracellular loop of CCR9 in vitro compared with other clones. A peptide (P1; VHWDFRQWWQPS) was identified which may inhibit the corresponding phage, C-4, binding to the second extracellular loop of CCR9. Furthermore, P1 was able to bind specifically with MOLT4 cells which exhibit marked expression of CCR9. In addition, P1 promoted the apoptosis of MOLT4 cells induced by doxorubicin, and inhibited the migration of MOLT4 cells in the presence of chemokine (C-C motif) ligand 25. It was suggested that decreased activity in the phosphorylation of protein kinase B in MOLT4 cells may be responsible for the inhibition. In conclusion, the peptide P1 derived from a screened phage is able to specifically bind to CCR9 and inhibit the activity of CCR9. It has potential use as an antagonist in the treatment of CCR9-overexpressed carcinoma.
Collapse
Affiliation(s)
- Yi Hu
- Department of Laboratory Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Aiping Ma
- Department of Respiratory Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Shan Lin
- Department of Orthopedics, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Yang Yang
- College of Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, P.R. China
| | - Guolin Hong
- Department of Laboratory Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| |
Collapse
|
16
|
Zhang Z, Sun T, Chen Y, Gong S, Sun X, Zou F, Peng R. CCL25/CCR9 Signal Promotes Migration and Invasion in Hepatocellular and Breast Cancer Cell Lines. DNA Cell Biol 2016; 35:348-57. [PMID: 27008282 DOI: 10.1089/dna.2015.3104] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Cancer is one of the most lethal diseases worldwide, and metastasis is the most common cause of patients' deaths. Identification and inhibition of markers involved in metastasis process in cancer cells are promising works to block metastasis and improve prognoses of patients. Chemokines are a superfamily of small, chemotactic cytokines, whose functions are based on interaction with corresponding receptors. It has been found that one of the functions of chemokines is to regulate migration and invasion abilities of lymphocytes, as well as cancer cells. Chemokine receptor 9 (CCR9) regulates trafficking of lymphocytes and cancer cell lines when interacting with its exclusive ligand chemokine 25 (CCL25). However, the mechanisms of CCL25/CCR9 signal that regulates metastasis of cancer cells are not completely known yet. In this study, we stimulated or inhibited CCL25/CCR9 signal in breast cancer cell line (MDA-MB-231) and hepatocellular cancer cell lines (HepG2 and HUH7), and found that CCL25/CCR9 signal resulted in different promotion of migration and invasion in different cell lines. These phenomena could be explained by selective regulation of several markers of epithelial-mesenchymal transition (EMT). Our findings suggested that CCL25/CCR9 signal may provide cancer cells with chemotactic abilities through influencing several EMT markers.
Collapse
Affiliation(s)
- Ziqi Zhang
- 1 West China School of Medicine, Sichuan University , Chengdu, People's Republic of China .,2 Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University , Chengdu, People's Republic of China
| | - Tong Sun
- 1 West China School of Medicine, Sichuan University , Chengdu, People's Republic of China
| | - Yuxi Chen
- 1 West China School of Medicine, Sichuan University , Chengdu, People's Republic of China
| | - Shu Gong
- 2 Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University , Chengdu, People's Republic of China
| | - Xiye Sun
- 3 Chengdu Shude High School Guanghua Campus , Chengdu, People's Republic of China
| | - Fangdong Zou
- 2 Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University , Chengdu, People's Republic of China
| | - Rui Peng
- 2 Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University , Chengdu, People's Republic of China
| |
Collapse
|
17
|
Tu Z, Xiao R, Xiong J, Tembo KM, Deng X, Xiong M, Liu P, Wang M, Zhang Q. CCR9 in cancer: oncogenic role and therapeutic targeting. J Hematol Oncol 2016; 9:10. [PMID: 26879872 PMCID: PMC4754913 DOI: 10.1186/s13045-016-0236-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 01/21/2016] [Indexed: 11/10/2022] Open
Abstract
Cancer is currently one of the leading causes of death worldwide and is one of the most challenging major public health problems. The main challenges faced by clinicians in the management and treatment of cancer mainly arise from difficulties in early diagnosis and the emergence of tumor chemoresistance and metastasis. The structures of chemokine receptor 9 (CCR9) and its specific ligand chemokine ligand 25 (CCL25) have been elucidated, and, interestingly, a number of studies have demonstrated that CCR9 is a potential tumor biomarker in diagnosis and therapy, as it has been found to be highly expressed in a wide range of cancers. This expression pattern suggests that CCR9 may participate in many important biological activities involved in cancer progression. Researchers have shown that CCR9 that has been activated by its specific ligand CCL25 can interact with many signaling pathways, especially those involved in tumor chemoresistance and metastasis. This review, therefore, focuses on CCR9 induction activity and summarizes what is currently known regarding its role in cancers and its potential application in tumor-targeted therapy.
Collapse
Affiliation(s)
- Zhenbo Tu
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, 430071, China.
| | - Ruijing Xiao
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, 430071, China.
| | - Jie Xiong
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, 430071, China.
| | - Kingsley M Tembo
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, 430071, China.
| | - Xinzhou Deng
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, 430071, China.
| | - Meng Xiong
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, 430071, China.
| | - Pan Liu
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, 430071, China.
| | - Meng Wang
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, 430071, China.
| | - Qiuping Zhang
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
18
|
Intracellular expression profile and clinical significance of the CCR9-CCL25 chemokine receptor complex in nasopharyngeal carcinoma. The Journal of Laryngology & Otology 2015; 129:1013-9. [PMID: 26279399 DOI: 10.1017/s0022215115002108] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVES This study aimed to investigate the association of C-C chemokine receptor type 9 (CCR9) and C-C motif chemokine 25 (CCL25) expression levels with clinical and tumour-node-metastasis stage in nasopharyngeal carcinoma. METHODS A total of 42 nasopharyngeal carcinoma patients (nasopharyngeal carcinoma group) and 18 patients with a normal nasopharynx (control group) were included in this study. Tissues were collected during surgery and medical examinations. The CCR9 and CCL25 messenger RNA and protein levels were measured using quantitative reverse transcription polymerase chain reaction, Western blotting and immunohistochemical analysis. RESULTS CCR9 and CCL25 messenger RNA and protein levels were significantly increased in the nasopharyngeal carcinoma group compared with the control group (p < 0.05). Both CCR9 and CCL25 messenger RNA and protein levels were significantly higher in advanced-stage nasopharyngeal carcinoma (stages III and IV) patients compared with early-stage nasopharyngeal carcinoma (stages I and II) patients (p < 0.05). CONCLUSION The extent of CCR9 and CCL25 upregulation in nasopharyngeal carcinoma correlates with the tumour-node-metastasis stage.
Collapse
|
19
|
McLennan R, Schumacher LJ, Morrison JA, Teddy JM, Ridenour DA, Box AC, Semerad CL, Li H, McDowell W, Kay D, Maini PK, Baker RE, Kulesa PM. VEGF signals induce trailblazer cell identity that drives neural crest migration. Dev Biol 2015; 407:12-25. [PMID: 26278036 DOI: 10.1016/j.ydbio.2015.08.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 08/10/2015] [Accepted: 08/12/2015] [Indexed: 10/23/2022]
Abstract
Embryonic neural crest cells travel in discrete streams to precise locations throughout the head and body. We previously showed that cranial neural crest cells respond chemotactically to vascular endothelial growth factor (VEGF) and that cells within the migratory front have distinct behaviors and gene expression. We proposed a cell-induced gradient model in which lead neural crest cells read out directional information from a chemoattractant profile and instruct trailers to follow. In this study, we show that migrating chick neural crest cells do not display distinct lead and trailer gene expression profiles in culture. However, exposure to VEGF in vitro results in the upregulation of a small subset of genes associated with an in vivo lead cell signature. Timed addition and removal of VEGF in culture reveals the changes in neural crest cell gene expression are rapid. A computational model incorporating an integrate-and-switch mechanism between cellular phenotypes predicts migration efficiency is influenced by the timescale of cell behavior switching. To test the model hypothesis that neural crest cellular phenotypes respond to changes in the VEGF chemoattractant profile, we presented ectopic sources of VEGF to the trailer neural crest cell subpopulation and show diverted cell trajectories and stream alterations consistent with model predictions. Gene profiling of trailer cells that diverted and encountered VEGF revealed upregulation of a subset of 'lead' genes. Injection of neuropilin1 (Np1)-Fc into the trailer subpopulation or electroporation of VEGF morpholino to reduce VEGF signaling failed to alter trailer neural crest cell trajectories, suggesting trailers do not require VEGF to maintain coordinated migration. These results indicate that VEGF is one of the signals that establishes lead cell identity and its chemoattractant profile is critical to neural crest cell migration.
Collapse
Affiliation(s)
- Rebecca McLennan
- Stowers Institute for Medical Research, 1000 E. 50th St., Kansas City, MO 64110, USA
| | - Linus J Schumacher
- University of Oxford, Wolfson Centre for Mathematical Biology, Mathematical Institute, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford OX2 6GG, UK; Department of Computer Science, University of Oxford, Oxford OX1 3QD, UK
| | - Jason A Morrison
- Stowers Institute for Medical Research, 1000 E. 50th St., Kansas City, MO 64110, USA
| | - Jessica M Teddy
- Stowers Institute for Medical Research, 1000 E. 50th St., Kansas City, MO 64110, USA
| | - Dennis A Ridenour
- Stowers Institute for Medical Research, 1000 E. 50th St., Kansas City, MO 64110, USA
| | - Andrew C Box
- Stowers Institute for Medical Research, 1000 E. 50th St., Kansas City, MO 64110, USA
| | - Craig L Semerad
- Stowers Institute for Medical Research, 1000 E. 50th St., Kansas City, MO 64110, USA
| | - Hua Li
- Stowers Institute for Medical Research, 1000 E. 50th St., Kansas City, MO 64110, USA
| | - William McDowell
- Stowers Institute for Medical Research, 1000 E. 50th St., Kansas City, MO 64110, USA
| | - David Kay
- Department of Computer Science, University of Oxford, Oxford OX1 3QD, UK
| | - Philip K Maini
- University of Oxford, Wolfson Centre for Mathematical Biology, Mathematical Institute, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford OX2 6GG, UK
| | - Ruth E Baker
- University of Oxford, Wolfson Centre for Mathematical Biology, Mathematical Institute, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford OX2 6GG, UK
| | - Paul M Kulesa
- Stowers Institute for Medical Research, 1000 E. 50th St., Kansas City, MO 64110, USA; Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, KS 66160, USA.
| |
Collapse
|
20
|
Lee S, Heinrich EL, Li L, Lu J, Choi AH, Levy RA, Wagner JE, Yip MLR, Vaidehi N, Kim J. CCR9-mediated signaling through β-catenin and identification of a novel CCR9 antagonist. Mol Oncol 2015; 9:1599-611. [PMID: 26003048 DOI: 10.1016/j.molonc.2015.04.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 04/27/2015] [Accepted: 04/28/2015] [Indexed: 12/17/2022] Open
Abstract
Elevated levels of chemokine receptor CCR9 expression in solid tumors may contribute to poor patient prognosis. In this study, we characterized a novel CCR9-mediated pathway that promotes pancreatic cancer cell invasion and drug resistance, indicating that CCR9 may play a critical role in cancer progression through activation of β-catenin. We noted that the CCL25/CCR9 axis in pancreatic cancer cells induced the activation of β-catenin, which enhanced cell proliferation, invasion, and drug resistance. CCR9-mediated activation of β-catenin and the resulting downstream effects were effectively inhibited by blockade of the PI3K/AKT pathway, but not by antagonism of Wnt. Importantly, we discovered that CCR9/CCL25 increased the lethal dose of gemcitabine, suggesting decreased efficacy of anti-cancer drugs with CCR9 signaling. Through in silico computational modeling, we identified candidate CCR9 antagonists and tested their effects on CCR9/β-catenin regulation of cell signaling and drug sensitivity. When combined with gemcitabine, it resulted in synergistic cytotoxicity. Our results show that CCR9/β-catenin signaling enhances pancreatic cancer invasiveness and chemoresistance, and may be a highly novel therapeutic target.
Collapse
Affiliation(s)
- Sangjun Lee
- Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Eileen L Heinrich
- Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Lily Li
- Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Jianming Lu
- Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Audrey H Choi
- Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Rachel A Levy
- Department of Immunology, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Jeffrey E Wagner
- Department of Immunology, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - M L Richard Yip
- HTS Lab, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Nagarajan Vaidehi
- Department of Immunology, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Joseph Kim
- Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, California, USA.
| |
Collapse
|
21
|
Li B, Wang Z, Zhong Y, Lan J, Li X, Lin H. CCR9-CCL25 interaction suppresses apoptosis of lung cancer cells by activating the PI3K/Akt pathway. Med Oncol 2015; 32:66. [PMID: 25691296 DOI: 10.1007/s12032-015-0531-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 02/13/2015] [Indexed: 12/16/2022]
Abstract
CC chemokine receptor-9 (CCR9) is highly expressed in non-small cell lung cancer (NSCLC) tissues and cell lines. However, the biological functions and the signals elicited by the interaction between CCR9 and its natural ligand CCL25 in NSCLC are unknown. Here, we selectively depleted CCR9 and inhibited CCR9-CCL25 interaction in NSCLC cells using small recombinant lentivirus-mediated miRNA, and investigated the tumorigenic effects in vitro and in vivo. Compromised CCR9-CCL25 interaction promoted apoptosis in NSCLC cells by activating phosphoinositide 3-kinase (PI3K)/Akt in vitro. In addition, we showed that CCR9-CCL25 interaction mediated the activation of the PI3K/Akt pathway in NSCLC cells, resulting in the up-regulation of anti-apoptotic proteins, as well as the down-regulation of apoptotic proteins in a PI3K-/Akt-dependent manner. These CCR9-CCL25-mediated effects were abrogated in the presence of a PI3K inhibitor (wortmannin 10 nM) or by inhibiting the CCR9-CCL25 interaction through CCR9 silencing, which also suggested that the biological function of CCR9-CCL25 was mainly regulated by PI3K. In vivo studies also demonstrated a significantly lower tumor burden in mice receiving CCR9-silence cells than those in mice receiving control cells. Together, these data suggested that CCR9-CCL25 interaction induced tumorigenesis of NSCLC cells and that this induction might be accomplished through the activation of the PI3K/Akt pathway. These findings may lead to a better understanding of the biological effects of CCR9-CCL25 interaction and provide clues for identifying novel therapeutic and preventive molecular markers for NSCLC.
Collapse
Affiliation(s)
- Baijun Li
- The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | | | | | | | | | | |
Collapse
|
22
|
Haqq J, Howells LM, Garcea G, Metcalfe MS, Steward WP, Dennison AR. Pancreatic stellate cells and pancreas cancer: current perspectives and future strategies. Eur J Cancer 2014; 50:2570-82. [PMID: 25091797 DOI: 10.1016/j.ejca.2014.06.021] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 06/25/2014] [Accepted: 06/30/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant disease with a very poor prognosis. To date patient outcomes have not improved principally due to the limited number of patients suitable for surgical resections and the radiation and chemotherapy resistance of these tumours. In the last decade, a failure of conventional therapies has forced researchers to re-examine the environment of PDAC. The tumour environment has been demonstrated to consist of an abundance of stroma containing many cells but predominantly pancreatic stellate cells (PSCs). Recent research has focused on understanding the interaction between PSCs and PDAC cells in vitro and in vivo. It is believed that the interaction between these cells is responsible for supporting tumour growth, invasion and metastasis and creating the barrier to delivery of chemotherapeutics. Novel approaches which focus on the interactions between PDAC and PSCs which sustain the tumour microenvironment may achieve significant patient benefits. This manuscript reviews the current evidence regarding PSCs, their interaction with PDAC cells and the potential implication this may have for future therapies. METHODS A PubMed search was carried out for the terms 'pancreas cancer' OR 'pancreatic cancer', AND 'pancreatic stellate cells', NOT 'hepatic stellate cells'. All studies were screened and assessed for their eligibility and manuscripts exploring the relationship between PSCs and PDAC were included. The studies were subdivided into in vitro and in vivo groups. RESULTS One hundred and sixty-six manuscripts were identified and reduced to seventy-three in vitro and in vivo studies for review. The manuscripts showed that PDAC cells and PSCs interact with each other to enhance proliferation, reduce apoptosis and increase migration and invasion of cancer cells. The pathways through which they facilitate these actions provide potential targets for future novel therapies. CONCLUSION There is accumulating evidence supporting the multiple roles of PSCs in establishing the tumour microenvironment and supporting the survival of PDAC. To further validate these findings there is a need for greater use of physiologically relevant models of pancreatic cancer in vitro such as three dimensional co-cultures and the use of orthotopic and genetically engineered murine (GEM) models in vivo.
Collapse
Affiliation(s)
- Jonathan Haqq
- Department of Hepatobiliary and Pancreatic Surgery & Cancer Studies and Molecular Medicine Group, University Hospitals of Leicester & University of Leicester, Leicester LE5 4PW, United Kingdom.
| | - Lynne M Howells
- Department of Hepatobiliary and Pancreatic Surgery & Cancer Studies and Molecular Medicine Group, University Hospitals of Leicester & University of Leicester, Leicester LE5 4PW, United Kingdom
| | - Giuseppe Garcea
- Department of Hepatobiliary and Pancreatic Surgery & Cancer Studies and Molecular Medicine Group, University Hospitals of Leicester & University of Leicester, Leicester LE5 4PW, United Kingdom
| | - Matthew S Metcalfe
- Department of Hepatobiliary and Pancreatic Surgery & Cancer Studies and Molecular Medicine Group, University Hospitals of Leicester & University of Leicester, Leicester LE5 4PW, United Kingdom
| | - Will P Steward
- Department of Hepatobiliary and Pancreatic Surgery & Cancer Studies and Molecular Medicine Group, University Hospitals of Leicester & University of Leicester, Leicester LE5 4PW, United Kingdom
| | - Ashley R Dennison
- Department of Hepatobiliary and Pancreatic Surgery & Cancer Studies and Molecular Medicine Group, University Hospitals of Leicester & University of Leicester, Leicester LE5 4PW, United Kingdom
| |
Collapse
|
23
|
Zhou J, Xiang Y, Yoshimura T, Chen K, Gong W, Huang J, Zhou Y, Yao X, Bian X, Wang JM. The role of chemoattractant receptors in shaping the tumor microenvironment. BIOMED RESEARCH INTERNATIONAL 2014; 2014:751392. [PMID: 25110692 PMCID: PMC4119707 DOI: 10.1155/2014/751392] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/17/2014] [Indexed: 12/13/2022]
Abstract
Chemoattractant receptors are a family of seven transmembrane G protein coupled receptors (GPCRs) initially found to mediate the chemotaxis and activation of immune cells. During the past decades, the functions of these GPCRs have been discovered to not only regulate leukocyte trafficking and promote immune responses, but also play important roles in homeostasis, development, angiogenesis, and tumor progression. Accumulating evidence indicates that chemoattractant GPCRs and their ligands promote the progression of malignant tumors based on their capacity to orchestrate the infiltration of the tumor microenvironment by immune cells, endothelial cells, fibroblasts, and mesenchymal cells. This facilitates the interaction of tumor cells with host cells, tumor cells with tumor cells, and host cells with host cells to provide a basis for the expansion of established tumors and development of distant metastasis. In addition, many malignant tumors of the nonhematopoietic origin express multiple chemoattractant GPCRs that increase the invasiveness and metastasis of tumor cells. Therefore, GPCRs and their ligands constitute targets for the development of novel antitumor therapeutics.
Collapse
Affiliation(s)
- Jiamin Zhou
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
- Endoscopic Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yi Xiang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
- Department of Pulmonary Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Teizo Yoshimura
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Keqiang Chen
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Wanghua Gong
- Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Jian Huang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ye Zhou
- Department of Gastric Cancer and Soft Tissue Surgery, Fudan University Cancer Center, Shanghai 200032, China
| | - Xiaohong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ji Ming Wang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|