1
|
Gandhi AK, Huang YH, Sun ZYJ, Kim WM, Kondo Y, Hanley T, Beauchemin N, Blumberg RS. Structural aspects of CEACAM1 interactions. Eur J Clin Invest 2024; 54 Suppl 2:e14357. [PMID: 39555955 DOI: 10.1111/eci.14357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/03/2024] [Indexed: 11/19/2024]
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is a membrane protein that plays an important role in a variety of immune and non-immune functions. Such functions are regulated by its activity as a homophilic ligand but also through its ability to interact as a heterophilic ligand with various host proteins. These include CEACAM5, T cell immunoglobulin-mucin like protein-3 (TIM-3) and, potentially, protein death protein 1 (PD-1). Furthermore, CEACAM1 is targeted by various pathogens to allow them to invade a host and bypass an effective immune response. Clinically, CEACAM1 plays an important role in infectious diseases, autoimmunity and cancer. In this review, we describe the structural basis for CEACAM1 interactions as a homophilic and heterophilic ligand. We discuss the regulation of its monomeric, dimeric and oligomeric states in cis and trans binding as well as the consequences for eliciting downstream signalling activities. Furthermore, we explore the potential role of avidity in determining CEACAM1's activities.
Collapse
Affiliation(s)
- Amit K Gandhi
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yu-Hwa Huang
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Zhen-Yu J Sun
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Walter M Kim
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yasuyuki Kondo
- Division of Gastroenterology, Department of Internal Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Thomas Hanley
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nicole Beauchemin
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Canada
| | - Richard S Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Activation of CEACAM1 with an agonistic monoclonal antibody results in inhibition of melanoma cells. Cancer Gene Ther 2022; 29:1676-1685. [PMID: 35681020 DOI: 10.1038/s41417-022-00486-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/08/2022] [Accepted: 05/23/2022] [Indexed: 02/04/2023]
Abstract
Inhibitory receptors (IRs), such as the carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), are cell surface molecules expressed on both normal epithelial, endothelial, and hematopoietic cells and on neoplastic cells. IRs are usually used by cancer cells to inhibit immune cell functions. Thus, CEACAM1 positive tumor cells can interact homophilically with CEACAM1 expressed on T and NK cells to inhibit their antibody-dependent cell-mediated cytotoxicity (ADCC). In this study, we investigated the effect of agonistic/activating anti-CEACAM1 monoclonal antibody (mAb) on melanoma cell lines in vitro and in vivo, following our hypothesis that activation of CEACAM1 on melanoma cells by distinct mAbs may induce inhibition of cancer cell proliferation and/or their death. To address this, we established an activating anti-CEACAM1 mAb (CCM5.01) and characterized its binding to the CEACAM1 receptor. Using this mAb, we assessed the expression of CEACAM1 on four different human melanoma cell lines by western blot and flow cytometry and determined its effect on cell viability in vitro by MTT assay. Furthermore, we evaluated the mAb mechanism of action and found that binding of CEACAM1 with CCM5.01 induced SHP1 phosphorylation and p53 activation resulting in melanoma cell apoptosis. For in vivo studies, a xenograft model of melanoma was performed by injection of Mel-14 cells subcutaneously (s.c.) in SCID/Beige mice followed by intraperitoneal (i.p.) injection of CCM5.01 or of IgG1 isotype control every other day. CCM5.01 treated mice showed a slight but not significant decrease in tumor weight in comparison to the control group. Based on the obtained data, we suggest that activating CEACAM1 on melanoma cells might be a promising novel approach to fight cancers expressing this IR.
Collapse
|
3
|
Zhang X, Han X, Zuo P, Zhang X, Xu H. CEACAM5 stimulates the progression of non-small-cell lung cancer by promoting cell proliferation and migration. J Int Med Res 2020; 48:300060520959478. [PMID: 32993395 PMCID: PMC7536504 DOI: 10.1177/0300060520959478] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Objective To detect the expression of CEA-related cell adhesion molecule 5 (CEACAM5) in
non-small-cell lung cancer (NSCLC) and explore its function in the
progression and development of NSCLC. Methods qRT-PCR and immunohistochemistry were performed to detect CEACAM5 expression
in human NSCLC tissues and cell lines. The correlation between CEACAM5
expression and the clinicopathological features of patients with NSCLC was
also investigated. MTT, colony formation, wound healing, and immunoblot
assays were performed to detect the functions of CEACAM5 in NSCLC cells
in vitro, and immunoblotting was used to detect the
effects of CEACAM5 on p38–Smad2/3 signaling. Results CEACAM5 expression was elevated in human NSCLC tissues and cells. We further
found that CEACAM expression was correlated with clinicopathological
features including T division, lymph invasion, and histological grade in
patients with NSCLC. The in vitro assays confirmed that
CEACAM5 depletion inhibited the proliferation and migration of NSCLC cells
by activating p38–Smad2/3 signaling. We verified the involvement of CEACAM5
in the suppression of NSCLC tumor growth in mice. Conclusion CEACAM5 stimulated the progression of NSCLC by promoting cell proliferation
and migration in vitro and in vivo.
CEACAM5 may serve as a potential therapeutic target for the treatment of
NSCLC.
Collapse
Affiliation(s)
- Xinwen Zhang
- Department of General Practice, Linyi Central Hospital, Linyi, China
| | - Xingbao Han
- Department of Urology, Linyi Central Hospital, Linyi, China
| | - Pengli Zuo
- Central Laboratory, Linyi Central Hospital, Linyi, China
| | - Xiuying Zhang
- Department of Clinical Lab, Linyi Central Hospital, Linyi, China
| | - Hongbang Xu
- Department of Respiratory Medicine, Linyi Central Hospital, Linyi, China
| |
Collapse
|
4
|
Hayashi S, Osada Y, Miura K, Simizu S. Cell-dependent regulation of vasculogenic mimicry by carcinoembryonic antigen cell adhesion molecule 1 (CEACAM1). Biochem Biophys Rep 2020; 21:100734. [PMID: 32025578 PMCID: PMC6997815 DOI: 10.1016/j.bbrep.2020.100734] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 01/18/2020] [Accepted: 01/20/2020] [Indexed: 12/13/2022] Open
Abstract
Vasculogenic mimicry (VM) promotes tumor migration, metastasis, and invasion in various types of cancer, but the relationship between VM and these phenotypes remains undefined. In this study, we examined carcinoembryonic antigen cell adhesion molecule 1 (CEACAM1) as a novel target of VM. We found that ectopic expression of CEACAM1 in HT1080 human fibrosarcoma cells suppressed the formation of a VM-like network. Further, cell migration and proliferation were abated by the introduction of CEACAM1 into HT1080 cells. Conversely, knockout (KO) of the CEACAM1 gene in SK-MEL-28 melanoma cells, which normally express high levels of CEACAM1, inhibited formation of a VM-like network, which was covered on reintroduction of CEACAM1. These results suggest that CEACAM1 differentially regulates formation of the VM-like network between cancer cell types and implicate CEACAM1 as a novel therapeutic target in malignant cancer. CEACAM1 is not expressed in HT1080 cells, and overexpression of CEACAM1 in HT1080 cells suppresses vasculogenic mimicry. CEACAM1 is highly expressed in SK-MEL-28 cells, and deletion of CEACAM1 in SK-MEL-28 cells abolishes vasculogenic mimicry. CEACAM1 regulates vasculogenic mimicry in a cell-dependent manner.
Collapse
Affiliation(s)
- Soichiro Hayashi
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan
| | - Yoshiyuki Osada
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan
| | - Kazuki Miura
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan
| | - Siro Simizu
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan
| |
Collapse
|
5
|
Anti-carcinoembryonic antigen-related cell adhesion molecule antibody for fluorescence visualization of primary colon cancer and metastases in patient-derived orthotopic xenograft mouse models. Oncotarget 2020; 11:429-439. [PMID: 32064046 PMCID: PMC6996915 DOI: 10.18632/oncotarget.27446] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/04/2020] [Indexed: 01/30/2023] Open
Abstract
Background: Monoclonal antibody (mAb) 6G5j is a novel anti-CEACAM monoclonal antibody. Our aim was to investigate mAb 6G5j binding characteristics and to validate fluorescence targeting of colorectal tumors and metastases in patient derived orthotopic xenograft (PDOX) models with fluorescently labeled 6G5j. Materials/Methods: The MAb 6G5j binding profile was analyzed with ELISA, Western blot and immunohistochemistry. MAb 6G5j was conjugated to near-infrared dye IR800CW (LI-COR). Western blotting was performed with various colon cancer cell lysates to determine CEACAM expression. Nude mice received orthotopic implantation of patient-derived primary colon cancer and patient-derived colon cancer metastases. Mice were administered varying doses of 6G5j-IR800CW via tail vein injection and imaged 24 and 48 hours later. Results: MAb 6G5j bound to human CEACAM1, 3, 5, 6 and 8. Western blotting demonstrated varied expression of CEACAMs in 15 of 16 colon cancer lysates. Dose and time-response imaging demonstrated optimal imaging 48 hours after administration of 50 μg 6G5j-IR800CW (Tumor-to-liver ratio (TLR) 3.17, SEM ± 0.45). Primary cancers and multiple metastases were fluorescently visualized. Conclusions: Anti-CEACAM antibody 6G5j binds multiple CEACAMs which may lead to improved detection of tumor margins for tumors and metastases that have variable expression of CEA and other CEACAMs. 6G5j mAb may be useful for colon cancer detection for pre-surgical diagnosis and fluorescence-guided surgery.
Collapse
|
6
|
Size Matters: The Functional Role of the CEACAM1 Isoform Signature and Its Impact for NK Cell-Mediated Killing in Melanoma. Cancers (Basel) 2019; 11:cancers11030356. [PMID: 30871206 PMCID: PMC6468645 DOI: 10.3390/cancers11030356] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/21/2019] [Accepted: 03/09/2019] [Indexed: 12/22/2022] Open
Abstract
Malignant melanoma is the most aggressive and treatment resistant type of skin cancer. It is characterized by continuously rising incidence and high mortality rate due to its high metastatic potential. Various types of cell adhesion molecules have been implicated in tumor progression in melanoma. One of these, the carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), is a multi-functional receptor protein potentially expressed in epithelia, endothelia, and leukocytes. CEACAM1 often appears in four isoforms differing in the length of their extracellular and intracellular domains. Both the CEACAM1 expression in general, and the ratio of the expressed CEACAM1 splice variants appear very dynamic. They depend on both the cell activation stage and the cell growth phase. Interestingly, normal melanocytes are negative for CEACAM1, while melanomas often show high expression. As a cell–cell communication molecule, CEACAM1 mediates the direct interaction between tumor and immune cells. In the tumor cell this interaction leads to functional inhibitions, and indirectly to decreased cancer cell immunogenicity by down-regulation of ligands of the NKG2D receptor. On natural killer (NK) cells it inhibits NKG2D-mediated cytolysis and signaling. This review focuses on novel mechanistic insights into CEACAM1 isoforms for NK cell-mediated immune escape mechanisms in melanoma, and their clinical relevance in patients suffering from malignant melanoma.
Collapse
|
7
|
Lucarini G, Zizzi A, Re M, Sayeed MA, Di Primio R, Rubini C. Prognostic implication of CEACAM1 expression in squamous cell carcinoma of the larynx: Pilot study. Head Neck 2018; 41:1615-1621. [PMID: 30582236 DOI: 10.1002/hed.25589] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 10/01/2018] [Accepted: 12/05/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND CEACAM1, a valuable biomarker for several cancers, have remained unexplored up to the present in laryngeal squamous cell carcinoma (LSCC). We aimed to examine CEACAM1 expression and evaluate its combinational clinical significance for the diagnosis or prognosis and treatment decision making in LSCC. METHODS CEACAM1 expression was assessed by immunohistochemistry in 54 LSCCs and evaluate its correlation with clinical and histopathological features. RESULTS CEACAM subtype 1 (CEACAM1) expression was positive in 50% of the cases. No significant difference was observed in relation to age, gender, tumor size, and tumor stage. CEACAM1 expression correlated with tumor grade, development of local recurrence, node and distant metastasis. Kaplan-Meier survival curves showed that CEACAM1 staining was inversely correlated with both overall and disease-specific 5-year survival. CONCLUSIONS Our study is the first to demonstrate that CEACAM1 expression is associated with an adverse prognosis in LSCC. CEACAM1 is a valuable biomarker and a promising therapeutic target in LSCC.
Collapse
Affiliation(s)
- Guendalina Lucarini
- Department of Clinic and Molecular Sciences, Histology, Polytechnic University of Marche, Ancona, Italy
| | - Antonio Zizzi
- Department of Biomedical Sciences and Public Health, Section of Pathologic Anatomy and Histopathology, Polytechnic University of Marche, Ancona, Italy
| | - Massimo Re
- Department of Otorhinolaryngology, Umberto I University General Hospital, Polytechnic University of Marche, Ancona, Italy
| | - Md Abu Sayeed
- Section of Occupational Medicine, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Roberto Di Primio
- Department of Clinic and Molecular Sciences, Histology, Polytechnic University of Marche, Ancona, Italy
| | - Corrado Rubini
- Department of Biomedical Sciences and Public Health, Section of Pathologic Anatomy and Histopathology, Polytechnic University of Marche, Ancona, Italy
| |
Collapse
|
8
|
The Induction of Selected Wnt Target Genes by Tcf1 Mediates Generation of Tumorigenic Colon Stem Cells. Cell Rep 2017; 19:981-994. [DOI: 10.1016/j.celrep.2017.04.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 02/28/2017] [Accepted: 04/05/2017] [Indexed: 12/26/2022] Open
|
9
|
Helicobacter pylori adhesin HopQ engages in a virulence-enhancing interaction with human CEACAMs. Nat Microbiol 2016; 2:16189. [PMID: 27748768 DOI: 10.1038/nmicrobiol.2016.189] [Citation(s) in RCA: 179] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/31/2016] [Indexed: 02/08/2023]
Abstract
Helicobacter pylori specifically colonizes the human gastric epithelium and is the major causative agent for ulcer disease and gastric cancer development. Here, we identify members of the carcinoembryonic antigen-related cell adhesion molecule (CEACAM) family as receptors of H. pylori and show that HopQ is the surface-exposed adhesin that specifically binds human CEACAM1, CEACAM3, CEACAM5 and CEACAM6. HopQ-CEACAM binding is glycan-independent and targeted to the N-domain. H. pylori binding induces CEACAM1-mediated signalling, and the HopQ-CEACAM1 interaction enables translocation of the virulence factor CagA into host cells and enhances the release of pro-inflammatory mediators such as interleukin-8. Based on the crystal structure of HopQ, we found that a β-hairpin insertion (HopQ-ID) in HopQ's extracellular 3+4 helix bundle domain is important for CEACAM binding. A peptide derived from this domain competitively inhibits HopQ-mediated activation of the Cag virulence pathway, as genetic or antibody-mediated abrogation of the HopQ function shows. Together, our data suggest the HopQ-CEACAM1 interaction to be a potentially promising novel therapeutic target to combat H. pylori-associated diseases.
Collapse
|
10
|
Papadodima O, Moulos P, Koryllou A, Piroti G, Kolisis F, Chatziioannou A, Pletsa V. Modulation of Pathways Underlying Distinct Cell Death Mechanisms in Two Human Lung Cancer Cell Lines in Response to SN1 Methylating Agents Treatment. PLoS One 2016; 11:e0160248. [PMID: 27467507 PMCID: PMC4965087 DOI: 10.1371/journal.pone.0160248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 07/15/2016] [Indexed: 12/13/2022] Open
Affiliation(s)
- Olga Papadodima
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Panagiotis Moulos
- Institute of Molecular Biology and Genetics, Biomedical Sciences Research Centre ‘Alexander Fleming’, 16672 Vari, Greece
| | - Aggeliki Koryllou
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Georgia Piroti
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Fragiskos Kolisis
- Laboratory of Biotechnology, School of Chemical Engineering, National Technical University of Athens, 15780 Athens, Greece
| | - Aristotelis Chatziioannou
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
- Enios Applications Private Company, 25 Al.Pantou str., 17671 Athens, Greece
- * E-mail: (AC); (VP)
| | - Vasiliki Pletsa
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
- * E-mail: (AC); (VP)
| |
Collapse
|
11
|
Zhuo Y, Yang JY, Moremen KW, Prestegard JH. Glycosylation Alters Dimerization Properties of a Cell-surface Signaling Protein, Carcinoembryonic Antigen-related Cell Adhesion Molecule 1 (CEACAM1). J Biol Chem 2016; 291:20085-95. [PMID: 27471271 DOI: 10.1074/jbc.m116.740050] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Indexed: 12/12/2022] Open
Abstract
Human carcinoembryonic antigen-related cell adhesion molecule 1 (C?/Au: EACAM1) is a cell-surface signaling molecule involved in cell adhesion, proliferation, and immune response. It is also implicated in cancer angiogenesis, progression, and metastasis. This diverse set of effects likely arises as a result of the numerous homophilic and heterophilic interactions that CEACAM1 can have with itself and other molecules. Its N-terminal Ig variable (IgV) domain has been suggested to be a principal player in these interactions. Previous crystal structures of the β-sandwich-like IgV domain have been produced using Escherichia coli-expressed material, which lacks native glycosylation. These have led to distinctly different proposals for dimer interfaces, one involving interactions of ABED β-strands and the other involving GFCC'C″ β-strands, with the former burying one prominent glycosylation site. These structures raise questions as to which form may exist in solution and what the effect of glycosylation may have on this form. Here, we use NMR cross-correlation measurements to examine the effect of glycosylation on CEACAM1-IgV dimerization and use residual dipolar coupling (RDC) measurements to characterize the solution structure of the non-glycosylated form. Our findings demonstrate that even addition of a single N-linked GlcNAc at potential glycosylation sites inhibits dimer formation. Surprisingly, RDC data collected on E. coli expressed material in solution indicate that a dimer using the non-glycosylated GFCC'C″ interface is preferred even in the absence of glycosylation. The results open new questions about what other factors may facilitate dimerization of CEACAM1 in vivo, and what roles glycosylation may play in heterophylic interactions.
Collapse
Affiliation(s)
- You Zhuo
- From the Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602
| | - Jeong-Yeh Yang
- From the Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602
| | - Kelley W Moremen
- From the Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602
| | - James H Prestegard
- From the Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602
| |
Collapse
|
12
|
Wang Y, Chen Y, Yan Y, Li X, Chen G, He N, Shen S, Chen G, Zhang C, Liao W, Liao Y, Bin J. Loss of CEACAM1, a Tumor-Associated Factor, Attenuates Post-infarction Cardiac Remodeling by Inhibiting Apoptosis. Sci Rep 2016; 6:21972. [PMID: 26911181 PMCID: PMC4766464 DOI: 10.1038/srep21972] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 01/29/2016] [Indexed: 12/30/2022] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule1 (CEACAM1) is a tumor-associated factor that is known to be involved in apoptosis, but the role of CEACAM1 in cardiovascular disease is unclear. We aims to investigate whether CEACAM1 influences cardiac remodeling in mice with myocardial infarction (MI) and hypoxia-induced cardiomyocyte injury. Both serum in patients and myocardial CEACAM1 levels in mice were significantly increased in response to MI, while levels were elevated in neonatal rat cardiomyocytes (NRCs) exposed to hypoxia. Eight weeks after MI, a lower mortality rate, improved cardiac function, and less cardiac remodeling in CEACAM1 knock-out (KO) mice than in their wild-type (WT) littermates were observed. Moreover, myocardial expression of mitochondrial Bax, cytosolic cytochrome C, and cleaved caspase-3 was significantly lower in CEACAM1 KO mice than in WT mice. In cultured NRCs exposed to hypoxia, recombinant human CEACAM1 (rhCEACAM1) reduced mitochondrial membrane potential, upregulated mitochondrial Bax, increased cytosolic cytochrome C and cleaved caspase-3, and consequently increased apoptosis. RhCEACAM1 also increased the levels of GRP78 and CHOP in NRCs with hypoxia. All of these effects were abolished by silencing CEACAM1. Our study indicates that CEACAM1 exacerbates hypoxic cardiomyocyte injury and post-infarction cardiac remodeling by enhancing cardiomyocyte mitochondrial dysfunction and endoplasmic reticulum stress-induced apoptosis.
Collapse
Affiliation(s)
- Yan Wang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yanmei Chen
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yi Yan
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xinzhong Li
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Guojun Chen
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Nvqin He
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shuxin Shen
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Department of Cardiology, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, 450003, China
| | - Gangbin Chen
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chuanxi Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yulin Liao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jianping Bin
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
13
|
Kaposi's Sarcoma-Associated Herpesvirus Interleukin-6 Modulates Endothelial Cell Movement by Upregulating Cellular Genes Involved in Migration. mBio 2015; 6:e01499-15. [PMID: 26646010 PMCID: PMC4676281 DOI: 10.1128/mbio.01499-15] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Kaposi’s sarcoma-associated herpesvirus (KSHV) is the causative agent of human Kaposi’s sarcoma, a tumor that arises from endothelial cells, as well as two B cell lymphoproliferative diseases, primary effusion lymphoma and multicentric Castleman’s disease. KSHV utilizes a variety of mechanisms to evade host immune responses and promote cellular transformation and growth in order to persist for the life of the host. A viral homolog of human interleukin-6 (hIL-6) named viral interleukin-6 (vIL-6) is encoded by KSHV and expressed in KSHV-associated cancers. Similar to hIL-6, vIL-6 is secreted, but the majority of vIL-6 is retained within the endoplasmic reticulum, where it can initiate functional signaling through part of the interleukin-6 receptor complex. We sought to determine how intracellular vIL-6 modulates the host endothelial cell environment by analyzing vIL-6’s impact on the endothelial cell transcriptome. vIL-6 significantly altered the expression of many cellular genes associated with cell migration. In particular, vIL-6 upregulated the host factor carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) at the protein and message levels. CEACAM1 has been implicated in tumor invasion and metastasis and promotes migration and vascular remodeling in endothelial cells. We report that vIL-6 upregulates CEACAM1 by a STAT3-dependent mechanism and that CEACAM1 promotes vIL-6-mediated migration. Furthermore, latent and de novo KSHV infections of endothelial cells also induce CEACAM1 expression. Collectively, our data suggest that vIL-6 modulates endothelial cell migration by upregulating the expression of cellular factors, including CEACAM1. Kaposi’s sarcoma-associated herpesvirus (KSHV) is linked with the development of three human malignancies, Kaposi’s sarcoma, multicentric Castleman’s disease, and primary effusion lymphoma. KSHV expresses many factors that enable the virus to manipulate the host environment in order to persist and induce disease. The viral interleukin-6 (vIL-6) produced by KSHV is structurally and functionally homologous to the human cytokine interleukin-6, except that vIL-6 is secreted slowly and functions primarily from inside the host cell. To investigate the unique intracellular role of vIL-6, we analyzed the impact of vIL-6 on endothelial cell gene expression. We report that vIL-6 significantly alters the expression of genes associated with cell movement, including that for CEACAM1. The gene for CEACAM1 was upregulated by vIL-6 and by latent and primary KSHV infection and promotes vIL-6-mediated endothelial cell migration. This work advances the field’s understanding of vIL-6 function and its contribution to KSHV pathogenesis.
Collapse
|
14
|
Bonsor DA, Günther S, Beadenkopf R, Beckett D, Sundberg EJ. Diverse oligomeric states of CEACAM IgV domains. Proc Natl Acad Sci U S A 2015; 112:13561-6. [PMID: 26483485 PMCID: PMC4640789 DOI: 10.1073/pnas.1509511112] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) comprise a large family of cell surface adhesion molecules that bind to themselves and other family members to carry out numerous cellular functions, including proliferation, signaling, differentiation, tumor suppression, and survival. They also play diverse and significant roles in immunity and infection. The formation of CEACAM oligomers is caused predominantly by interactions between their N-terminal IgV domains. Although X-ray crystal structures of CEACAM IgV domain homodimers have been described, how CEACAMs form heterodimers or remain monomers is poorly understood. To address this key aspect of CEACAM function, we determined the crystal structures of IgV domains that form a homodimeric CEACAM6 complex, monomeric CEACAM8, and a heterodimeric CEACAM6-CEACAM8 complex. To confirm and quantify these interactions in solution, we used analytical ultracentrifugation to measure the dimerization constants of CEACAM homodimers and isothermal titration calorimetry to determine the thermodynamic parameters and binding affinities of CEACAM heterodimers. We found the CEACAM6-CEACAM8 heterodimeric state to be substantially favored energetically relative to the CEACAM6 homodimer. Our data provide a molecular basis for the adoption of the diverse oligomeric states known to exist for CEACAMs and suggest ways in which CEACAM6 and CEACAM8 regulate the biological functions of one another, as well as of additional CEACAMs with which they interact, both in cis and in trans.
Collapse
Affiliation(s)
- Daniel A Bonsor
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Sebastian Günther
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Robert Beadenkopf
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Dorothy Beckett
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742
| | - Eric J Sundberg
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201; Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201; Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
15
|
Löffek S, Ullrich N, Görgens A, Murke F, Eilebrecht M, Menne C, Giebel B, Schadendorf D, Singer BB, Helfrich I. CEACAM1-4L Promotes Anchorage-Independent Growth in Melanoma. Front Oncol 2015; 5:234. [PMID: 26539411 PMCID: PMC4609850 DOI: 10.3389/fonc.2015.00234] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 10/05/2015] [Indexed: 11/13/2022] Open
Abstract
Widespread metastasis is the leading course of death in many types of cancer, including malignant melanoma. The process of metastasis can be divided into a number of complex cell biological events, collectively termed the “invasion-metastasis cascade.” Previous reports have characterized the capability of anchorage-independent growth of cancer cells in vitro as a key characteristic of highly aggressive tumor cells, particularly with respect to metastatic potential. Biological heterogeneity as well as drastic alterations in cell adhesion of disseminated cancer cells support escape mechanisms for metastases to overcome conventional therapies. Here, we show that exclusively the carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) splice variant CEACAM1-4L supports an anchorage-independent signature in malignant melanoma. These results highlight important variant-specific modulatory functions of CEACAM1 for metastatic spread in patients suffering malignant melanoma.
Collapse
Affiliation(s)
- Stefanie Löffek
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University of Duisburg-Essen , Essen , Germany ; German Cancer Consortium (DKTK), Medical Faculty, University of Duisburg-Essen , Essen , Germany
| | - Nico Ullrich
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University of Duisburg-Essen , Essen , Germany ; German Cancer Consortium (DKTK), Medical Faculty, University of Duisburg-Essen , Essen , Germany
| | - André Görgens
- Institute for Transfusion Medicine, Medical Faculty, University of Duisburg-Essen , Essen , Germany
| | - Florian Murke
- Institute for Transfusion Medicine, Medical Faculty, University of Duisburg-Essen , Essen , Germany
| | - Mara Eilebrecht
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University of Duisburg-Essen , Essen , Germany ; German Cancer Consortium (DKTK), Medical Faculty, University of Duisburg-Essen , Essen , Germany
| | - Christopher Menne
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University of Duisburg-Essen , Essen , Germany ; German Cancer Consortium (DKTK), Medical Faculty, University of Duisburg-Essen , Essen , Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, Medical Faculty, University of Duisburg-Essen , Essen , Germany
| | - Dirk Schadendorf
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University of Duisburg-Essen , Essen , Germany ; German Cancer Consortium (DKTK), Medical Faculty, University of Duisburg-Essen , Essen , Germany
| | - Bernhard B Singer
- Institute of Anatomy, Medical Faculty, University of Duisburg-Essen , Essen , Germany
| | - Iris Helfrich
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University of Duisburg-Essen , Essen , Germany ; German Cancer Consortium (DKTK), Medical Faculty, University of Duisburg-Essen , Essen , Germany
| |
Collapse
|
16
|
Bonsor DA, Beckett D, Sundberg EJ. Structure of the N-terminal dimerization domain of CEACAM7. Acta Crystallogr F Struct Biol Commun 2015; 71:1169-75. [PMID: 26323304 PMCID: PMC4555925 DOI: 10.1107/s2053230x15013576] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 07/15/2015] [Indexed: 11/22/2022] Open
Abstract
CEACAM7 is a human cellular adhesion protein that is expressed on the surface of colon and rectum epithelial cells and is downregulated in colorectal cancers. It achieves cell adhesion through dimerization of the N-terminal IgV domain. The crystal structure of the N-terminal dimerization domain of CEACAM has been determined at 1.47 Å resolution. The overall fold of CEACAM7 is similar to those of CEACAM1 and CEACAM5; however, there are differences, the most notable of which is an insertion that causes the C'' strand to buckle, leading to the creation of a hydrogen bond in the dimerization interface. The Kdimerization for CEACAM7 determined by sedimentation equilibrium is tenfold tighter than that measured for CEACAM5. These findings suggest that the dimerization affinities of CEACAMs are modulated via sequence variation in the dimerization surface.
Collapse
Affiliation(s)
- Daniel A. Bonsor
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dorothy Beckett
- Department of Chemistry and Biochemistry, University of Maryland College Park, Baltimore, MD 20742, USA
| | - Eric J. Sundberg
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
17
|
Ling Y, Wang J, Wang L, Hou J, Qian P, Xiang-dong W. Roles of CEACAM1 in cell communication and signaling of lung cancer and other diseases. Cancer Metastasis Rev 2015; 34:347-57. [DOI: 10.1007/s10555-015-9569-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
18
|
Ullrich N, Heinemann A, Nilewski E, Scheffrahn I, Klode J, Scherag A, Schadendorf D, Singer BB, Helfrich I. CEACAM1-3S Drives Melanoma Cells into NK Cell-Mediated Cytolysis and Enhances Patient Survival. Cancer Res 2015; 75:1897-907. [PMID: 25744717 DOI: 10.1158/0008-5472.can-14-1752] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 02/17/2015] [Indexed: 11/16/2022]
Abstract
CEACAM1 is a widely expressed multifunctional cell-cell adhesion protein reported to serve as a poor prognosis marker in melanoma patients. In this study, we examine the functional and clinical contributions of the four splice isoforms of CEACAM1. Specifically, we present in vitro and in vivo evidence that they affect melanoma progression and immune surveillance in a negative or positive manner that is isoform specific in action. In contrast with isoforms CEACAM1-4S and CEACAM1-4L, expression of isoforms CEACAM1-3S and CEACAM1-3L is induced during disease progression shown to correlate with clinical stage. Unexpectedly, overall survival was prolonged in patients with advanced melanomas expressing CEACAM1-3S. The favorable effects of CEACAM1-3S related to enhanced immunogenicity, which was mediated by cell surface upregulation of NKG2D receptor ligands, thereby sensitizing melanoma cells to lysis by natural killer cells. Conversely, CEACAM1-4L downregulated cell surface levels of the NKG2D ligands MICA and ULBP2 by enhanced shedding, thereby promoting malignant character. Overall, our results define the splice isoform-specific immunomodulatory and cell biologic functions of CEACAM1 in melanoma pathogenesis.
Collapse
Affiliation(s)
- Nico Ullrich
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Anja Heinemann
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Elena Nilewski
- Institute of Anatomy, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Inka Scheffrahn
- Institute for Gastroenterology and Hepatology, Medical Faculty, University Duisburg-Essen, Essen, Germany
| | - Joachim Klode
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany
| | - André Scherag
- Clinical Epidemiology, Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - Dirk Schadendorf
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Bernhard B Singer
- Institute of Anatomy, University Hospital, University Duisburg-Essen, Essen, Germany.
| | - Iris Helfrich
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
19
|
Ortenberg R, Galore-Haskel G, Greenberg I, Zamlin B, Sapoznik S, Greenberg E, Barshack I, Avivi C, Feiler Y, Zan-Bar I, Besser MJ, Azizi E, Eitan F, Schachter J, Markel G. CEACAM1 promotes melanoma cell growth through Sox-2. Neoplasia 2014; 16:451-60. [PMID: 24931667 PMCID: PMC4198694 DOI: 10.1016/j.neo.2014.05.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The prognostic value of the carcinoembryonic antigen cell adhesion molecule 1 (CEACAM1) in melanoma was demonstrated more than a decade ago as superior to Breslow score. We have previously shown that intercellular homophilic CEACAM1 interactions protect melanoma cells from lymphocyte-mediated elimination. Here, we study the direct effects of CEACAM1 on melanoma cell biology. By employing tissue microarrays and low-passage primary cultures of metastatic melanoma, we show that CEACAM1 expression gradually increases from nevi to metastatic specimens, with a strong dominance of the CEACAM1-Long tail splice variant. Using experimental systems of CEACAM1 knockdown and overexpression of selective variants or truncation mutants, we prove that only the full-length long tail variant enhances melanoma cell proliferation in vitro and in vivo. This effect is not reversed with a CEACAM1-blocking antibody, suggesting that it is not mediated by intercellular homophilic interactions. Downstream, CEACAM1-Long increases the expression of Sox-2, which we show to be responsible for the CEACAM1-mediated enhanced proliferation. Furthermore, analysis of the CEACAM1 promoter reveals two single-nucleotide polymorphisms (SNPs) that significantly enhance the promoter's activity compared with the consensus nucleotides. Importantly, case-control genetic SNP analysis of 134 patients with melanoma and matched healthy donors show that patients with melanoma do not exhibit the Hardy-Weinberg balance and that homozygous SNP genotype enhances the hazard ratio to develop melanoma by 35%. These observations shed new mechanistic light on the role of CEACAM1 in melanoma, forming the basis for development of novel therapeutic and diagnostic technologies.
Collapse
Affiliation(s)
- Rona Ortenberg
- Ella Institute of Melanoma, Sheba Medical Center, Ramat-Gan, Israel; Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gilli Galore-Haskel
- Ella Institute of Melanoma, Sheba Medical Center, Ramat-Gan, Israel; Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ilanit Greenberg
- Ella Institute of Melanoma, Sheba Medical Center, Ramat-Gan, Israel
| | - Bella Zamlin
- Ella Institute of Melanoma, Sheba Medical Center, Ramat-Gan, Israel; Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sivan Sapoznik
- Ella Institute of Melanoma, Sheba Medical Center, Ramat-Gan, Israel
| | - Eyal Greenberg
- Ella Institute of Melanoma, Sheba Medical Center, Ramat-Gan, Israel; Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Iris Barshack
- Institute of Pathology, Sheba Medical Center, Ramat-Gan 526260, Israel
| | - Camila Avivi
- Institute of Pathology, Sheba Medical Center, Ramat-Gan 526260, Israel
| | - Yulia Feiler
- Cancer Research Center, Sheba Medical Center, Ramat-Gan, Israel
| | - Israel Zan-Bar
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michal J Besser
- Ella Institute of Melanoma, Sheba Medical Center, Ramat-Gan, Israel; Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ester Azizi
- Department of Dermatology, Sheba Medical Center, Ramat-Gan, Israel
| | - Friedman Eitan
- The Susanne-Levy Gertner Oncogenetics Unit, Danek Gertner Institute of Human Genetics, Ramat-Gan, Israel
| | - Jacob Schachter
- Ella Institute of Melanoma, Sheba Medical Center, Ramat-Gan, Israel
| | - Gal Markel
- Ella Institute of Melanoma, Sheba Medical Center, Ramat-Gan, Israel; Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Talpiot Medical Leadership Program, Sheba Medical Center, Ramat-Gan, Israel.
| |
Collapse
|
20
|
Zhao W, Zhang Y, Liu D, Zhong L, He Q, Zhao Y. Abnormal expression of CD66a promotes proliferation and inhibits apoptosis of human leukemic B cellsin vitro. Leuk Lymphoma 2014; 56:202-10. [DOI: 10.3109/10428194.2014.913287] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
21
|
Muturi HT, Dreesen JD, Nilewski E, Jastrow H, Giebel B, Ergun S, Singer BB. Tumor and endothelial cell-derived microvesicles carry distinct CEACAMs and influence T-cell behavior. PLoS One 2013; 8:e74654. [PMID: 24040308 PMCID: PMC3770572 DOI: 10.1371/journal.pone.0074654] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 08/08/2013] [Indexed: 01/15/2023] Open
Abstract
Normal and malignant cells release a variety of different vesicles into their extracellular environment. The most prominent vesicles are the microvesicles (MVs, 100-1 000 nm in diameter), which are shed of the plasma membrane, and the exosomes (70-120 nm in diameter), derivates of the endosomal system. MVs have been associated with intercellular communication processes and transport numerous proteins, lipids and RNAs. As essential component of immune-escape mechanisms tumor-derived MVs suppress immune responses. Additionally, tumor-derived MVs have been found to promote metastasis, tumor-stroma interactions and angiogenesis. Since members of the carcinoembryonic antigen related cell adhesion molecule (CEACAM)-family have been associated with similar processes, we studied the distribution and function of CEACAMs in MV fractions of different human epithelial tumor cells and of human and murine endothelial cells. Here we demonstrate that in association to their cell surface phenotype, MVs released from different human epithelial tumor cells contain CEACAM1, CEACAM5 and CEACAM6, while human and murine endothelial cells were positive for CEACAM1 only. Furthermore, MVs derived from CEACAM1 transfected CHO cells carried CEACAM1. In terms of their secretion kinetics, we show that MVs are permanently released in low doses, which are extensively increased upon cellular starvation stress. Although CEACAM1 did not transmit signals into MVs it served as ligand for CEACAM expressing cell types. We gained evidence that CEACAM1-positive MVs significantly increase the CD3 and CD3/CD28-induced T-cell proliferation. All together, our data demonstrate that MV-bound forms of CEACAMs play important roles in intercellular communication processes, which can modulate immune response, tumor progression, metastasis and angiogenesis.
Collapse
Affiliation(s)
| | | | - Elena Nilewski
- Institute of Anatomy, University Hospital Essen, Essen, Germany
| | - Holger Jastrow
- Institute of Anatomy, University Hospital Essen, Essen, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| | - Suleyman Ergun
- Institute of Anatomy, University Hospital Essen, Essen, Germany
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Bernhard B. Singer
- Institute of Anatomy, University Hospital Essen, Essen, Germany
- * E-mail:
| |
Collapse
|
22
|
Rao US, Hoerster NS, Thirumala S, Rao PS. The influence of metastatic site on the expression of CEA and cellular localization of β-catenin in colorectal cancer. J Gastroenterol Hepatol 2013; 28:505-12. [PMID: 23216017 DOI: 10.1111/jgh.12083] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2012] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIM The usefulness of carcinoembryonic antigen (CEA) in the diagnosis and prognosis of colorectal cancer (CRC) is unclear. The aim was to analyze changes in the expression of CEA during CRC progression and metastasis, so as to determine the influence of tumor metastatic organ on the CEA expression by CRC cells. METHODS The human biopsies of adenocarcinomas in colon and CRC liver and lung metastases were analyzed by immunohistochemistry for the expression of CEA. Expression of E-cadherin and β-catenin was also analyzed to localize the CRC neoplastic glands in metastatic tissues. RESULTS The CRC neoplastic glands in colon and liver expressed significantly higher amount of CEA compared with crypts in normal colon. In contrast, CRC neoplastic glands formed in lung expressed low CEA level. However, CEA expression was high in areas of tumor necrosis in lung. E-cadherin and β-catenin were cell membrane-bound in normal crypts and CRC neoplastic glands in colon and liver. Although these two proteins were also cell membrane-bound in a majority of CRC neoplastic glands in lungs, a significant proportion of these expressed β-catenin in the nucleus, which lacked either E-cadherin or β-catenin at the cell membrane. CONCLUSION Our findings indicate that lung microenvironment is unique in that it suppresses the expression of CEA by CRC cells forming neoplastic glands. In addition, lung microenvironment promotes nuclear localization of β-catenin, suggesting that the Wnt signaling pathway is relatively active highly in CRC metastasized to lung, when compared with liver or colon.
Collapse
Affiliation(s)
- U Subrahmanyeswara Rao
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Amarillo, USA
| | | | | | | |
Collapse
|
23
|
Slobodskaya O, Snijder EJ, Spaan WJM. Organ tropism of murine coronavirus does not correlate with the expression levels of the membrane-anchored or secreted isoforms of the carcinoembryonic antigen-related cell adhesion molecule 1 receptor. J Gen Virol 2012; 93:1918-1923. [PMID: 22673933 DOI: 10.1099/vir.0.043190-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is the sole known receptor of murine hepatitis virus (MHV) A59, but the available, often qualitative, data about CEACAM1 expression does not explain MHV organ tropism. Ceacam1 transcripts undergo alternative splicing resulting in multiple isoforms, including secreted CEACAM1 isoforms that can neutralize the virus. We determined the quantities of Ceacam1 transcripts encoding membrane-bound and secreted isoforms in mouse organs and a set of cell lines. In vivo, the lowest receptor mRNA levels were found in brain and muscle and these were similar to those in easily infectable cultured cells. While the quantities of the receptor transcripts varied between mouse organs, their abundance did not correlate with susceptibility to MHV infection. The proportion of transcripts encoding secreted isoforms also could not explain the selection of sites for virus replication, as it was constant in all organs. Our data suggest that neither of the two CEACAM1 isoforms defines MHV organ tropism.
Collapse
Affiliation(s)
- Olga Slobodskaya
- Molecular Virology Laboratory, Department of Medical Microbiology, Center of Infectious Diseases, Leiden University Medical Center, The Netherlands
| | - Eric J Snijder
- Molecular Virology Laboratory, Department of Medical Microbiology, Center of Infectious Diseases, Leiden University Medical Center, The Netherlands
| | - Willy J M Spaan
- Molecular Virology Laboratory, Department of Medical Microbiology, Center of Infectious Diseases, Leiden University Medical Center, The Netherlands
| |
Collapse
|
24
|
Arabzadeh A, Chan C, Nouvion AL, Breton V, Benlolo S, DeMarte L, Turbide C, Brodt P, Ferri L, Beauchemin N. Host-related carcinoembryonic antigen cell adhesion molecule 1 promotes metastasis of colorectal cancer. Oncogene 2012; 32:849-60. [PMID: 22469976 DOI: 10.1038/onc.2012.112] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Liver metastasis is the predominant cause of colorectal cancer (CRC)-related mortality in developed countries. Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is a cell adhesion molecule with reduced expression in early phases of CRC development and thus functions as a tumor growth inhibitor. However, CEACAM1 is upregulated in metastatic colon cancer, suggesting a bimodal role in CRC progression. To investigate the role of this protein in the host metastatic environment, Ceacam1(-/-) mice were injected intrasplenically with metastatic MC38 mouse CRC cells. A significant reduction in metastatic burden was observed in Ceacam1(-/-) compared with wild-type (WT) livers. Intravital microscopy showed decreased early survival of MC38 cells in Ceacam1(-/-) endothelial environment. Metastatic cell proliferation within the Ceacam1(-/-) livers was also diminished. Bone marrow-derived cell recruitment, attenuation of immune infiltrates and diminished CCL2, CCL3 and CCL5 chemokine production participated in the reduced Ceacam1(-/-) metastatic phenotype. Transplantations of WT bone marrow (BM) into Ceacam1(-/-) mice fully rescued metastatic development, whereas Ceacam1(-/-) BM transfer into WT mice showed reduced metastatic burden. Chimeric immune cell profiling revealed diminished recruitment of CD11b(+)Gr1(+) myeloid-derived suppressor cells (MDSCs) to Ceacam1(-/-) metastatic livers and adoptive transfer of MDSCs confirmed the involvement of these immune cells in reduction of liver metastasis. CEACAM1 may represent a novel metastatic CRC target for treatment.
Collapse
Affiliation(s)
- A Arabzadeh
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Chen Z, Chen L, Baker K, Olszak T, Zeissig S, Huang YH, Kuo TT, Mandelboim O, Beauchemin N, Lanier LL, Blumberg RS. CEACAM1 dampens antitumor immunity by down-regulating NKG2D ligand expression on tumor cells. ACTA ACUST UNITED AC 2011; 208:2633-40. [PMID: 22143889 PMCID: PMC3244030 DOI: 10.1084/jem.20102575] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
By retaining NKG2D ligands within tumor cells, carcinoembryonic antigen–related cell adhesion molecule 1 (CEACAM1) facilitates tumor cell escape from NK cell–mediated cytolysis in vitro and in vivo. Although carcinoembryonic antigen (CEA)–related cell adhesion molecule 1 (CEACAM1) has been viewed as a tumor suppressor, increasing clinical evidence shows that high levels of CEACAM1 expression on tumors correlates with poor prognosis and high risk of metastasis. Here, we examined the consequences of CEACAM1 expression on tumor cells. We show that tumor cell–associated CEACAM1 causes intracellular retention of various NKG2D ligands in mouse and human tumor cells. CEACAM1-silenced tumor cells expressed more cell surface NKG2D ligands and exhibited greater sensitivity to natural killer cell–mediated cytolysis in vitro and rejection in vivo. Our studies reveal a novel mechanism through which CEACAM1-bearing tumor cells may escape immune-surveillance.
Collapse
Affiliation(s)
- Zhangguo Chen
- Gastroenterology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Haruyama N, Hatakeyama J, Moriyama K, Kulkarni AB. Amelogenins: Multi-Functional Enamel Matrix Proteins and Their Binding Partners. J Oral Biosci 2011. [DOI: 10.1016/s1349-0079(11)80009-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
27
|
Piekielko-Witkowska A, Wiszomirska H, Wojcicka A, Poplawski P, Boguslawska J, Tanski Z, Nauman A. Disturbed expression of splicing factors in renal cancer affects alternative splicing of apoptosis regulators, oncogenes, and tumor suppressors. PLoS One 2010; 5:e13690. [PMID: 21082031 PMCID: PMC2972751 DOI: 10.1371/journal.pone.0013690] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Accepted: 10/07/2010] [Indexed: 12/02/2022] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) is the most common type of renal cancer. One of the processes disturbed in this cancer type is alternative splicing, although phenomena underlying these disturbances remain unknown. Alternative splicing consists of selective removal of introns and joining of residual exons of the primary transcript, to produce mRNA molecules of different sequence. Splicing aberrations may lead to tumoral transformation due to synthesis of impaired splice variants with oncogenic potential. In this paper we hypothesized that disturbed alternative splicing in ccRCC may result from improper expression of splicing factors, mediators of splicing reactions. Methodology/Principal Findings Using real-time PCR and Western-blot analysis we analyzed expression of seven splicing factors belonging to SR proteins family (SF2/ASF, SC35, SRp20, SRp75, SRp40, SRp55 and 9G8), and one non-SR factor, hnRNP A1 (heterogeneous nuclear ribonucleoprotein A1) in 38 pairs of tumor-control ccRCC samples. Moreover, we analyzed splicing patterns of five genes involved in carcinogenesis and partially regulated by analyzed splicing factors: RON, CEACAM1, Rac1, Caspase-9, and GLI1. Conclusions/Significance We found that the mRNA expression of splicing factors was disturbed in tumors when compared to paired controls, similarly as levels of SF2/ASF and hnRNP A1 proteins. The correlation coefficients between expression levels of specific splicing factors were increased in tumor samples. Moreover, alternative splicing of five analyzed genes was also disturbed in ccRCC samples and splicing pattern of two of them, Caspase-9 and CEACAM1 correlated with expression of SF2/ASF in tumors. We conclude that disturbed expression of splicing factors in ccRCC may possibly lead to impaired alternative splicing of genes regulating tumor growth and this way contribute to the process of carcinogenesis.
Collapse
Affiliation(s)
| | - Hanna Wiszomirska
- Department of Biochemistry and Molecular Biology, The Medical Centre of Postgraduate Education, Warsaw, Poland
| | - Anna Wojcicka
- Department of Biochemistry and Molecular Biology, The Medical Centre of Postgraduate Education, Warsaw, Poland
| | - Piotr Poplawski
- Department of Biochemistry and Molecular Biology, The Medical Centre of Postgraduate Education, Warsaw, Poland
| | - Joanna Boguslawska
- Department of Biochemistry and Molecular Biology, The Medical Centre of Postgraduate Education, Warsaw, Poland
| | | | - Alicja Nauman
- Department of Biochemistry and Molecular Biology, The Medical Centre of Postgraduate Education, Warsaw, Poland
- * E-mail:
| |
Collapse
|
28
|
Generation of human CEACAM1 transgenic mice and binding of Neisseria Opa protein to their neutrophils. PLoS One 2010; 5:e10067. [PMID: 20404914 PMCID: PMC2852402 DOI: 10.1371/journal.pone.0010067] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Accepted: 03/12/2010] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Human CEACAM1 is a cell-cell adhesion molecule with multiple functions including insulin clearance in the liver, vasculogenesis in endothelial cells, lumen formation in the mammary gland, and binding of certain human pathogens. PRINCIPAL FINDINGS Three genomic BAC clones containing the human CEACAM1 gene were microinjected into pronuclei of fertilized FVB mouse oocytes. The embryos were implanted in the oviducts of pseudopregnant females and allowed to develop to term. DNA from newborn mice was evaluated by PCR for the presence of the human CEACAM1 gene. Feces of the PCR positive offspring screened for expression of human CEACAM1. Using this assay, one out of five PCR positive lines was positive for human CEACAM1 expression and showed stable transmission to the F1 generation with the expected transmission frequency (0.5) for heterozygotes. Liver, lung, intestine, kidney, mammary gland, and prostate were strongly positive for the dual expression of both murine and human CEACAM1 and mimic that seen in human tissue. Peripheral blood and bone marrow granulocytes stained strongly for human CEACAM1 and bound Neisseria Opa proteins similar to that in human neutrophils. CONCLUSION These transgenic animals may serve as a model for the binding of human pathogens to human CEACAM1.
Collapse
|
29
|
Singer BB, Scheffrahn I, Kammerer R, Suttorp N, Ergun S, Slevogt H. Deregulation of the CEACAM expression pattern causes undifferentiated cell growth in human lung adenocarcinoma cells. PLoS One 2010; 5:e8747. [PMID: 20090913 PMCID: PMC2807459 DOI: 10.1371/journal.pone.0008747] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2009] [Accepted: 12/23/2009] [Indexed: 01/05/2023] Open
Abstract
CEACAM1, CEA/CEACAM5, and CEACAM6 are cell adhesion molecules (CAMs) of the carcinoembryonic antigen (CEA) family that have been shown to be deregulated in lung cancer and in up to 50% of all human cancers. However, little is known about the functional impact of these molecules on undifferentiated cell growth and tumor progression. Here we demonstrate that cell surface expression of CEACAM1 on confluent A549 human lung adenocarcinoma cells plays a critical role in differentiated, contact-inhibited cell growth. Interestingly, CEACAM1-L, but not CEACAM1-S, negatively regulates proliferation via its ITIM domain, while in proliferating cells no CEACAM expression is detectable. Furthermore, we show for the first time that CEACAM6 acts as an inducer of cellular proliferation in A549 cells, likely by interfering with the contact-inhibiting signal triggered by CEACAM1-4L, leading to undifferentiated anchorage-independent cell growth. We also found that A549 cells expressed significant amounts of non-membrane anchored variants of CEACAM5 and CEACAM6, representing a putative source for the increased CEACAM5/6 serum levels frequently found in lung cancer patients. Taken together, our data suggest that post-confluent contact inhibition is established and maintained by CEACAM1-4L, but disturbances of CEACAM1 signalling by CEACAM1-4S and other CEACAMs lead to undifferentiated cell growth and malignant transformation.
Collapse
|
30
|
Nouvion AL, Beauchemin N. [CEACAM1 as a central modulator of metabolism, tumor progression, angiogenesis and immunity]. Med Sci (Paris) 2009; 25:247-52. [PMID: 19361387 DOI: 10.1051/medsci/2009253247] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
CEACAM1 (carcinoembryonic antigen cell adhesion molecule 1), a member of the immunoglobulin (Ig) superfamily, is a heavily glycosylated protein. This glycoprotein exhibits an intracytoplasmic region that can be either long (71-73 amino acids) with two inhibitory tyrosine-phosphorylated motifs and several phosphorylated serine residues, or short (10 amino acid). CEACAM1 is a multifunctional protein that plays a role in intercellular adhesion, as an inhibitor of tumor development, as a bacterial adhesin, and as a receptor for the mouse hepatitis virus. Moreover, CEACAM1 is an active regulator of cell signaling, modulating the insulin or EGF receptor pathways in epithelial cells or the Zap-70 pathway in hematopoietic cells. The recent development of genetically modified mouse models altering the Ceacam1 gene corroborates most of these data, but also highlights CEACAM1's functional complexity. Thus, in addition to the functions identified previously, CEACAM1 is an important regulator of lipid metabolism, of tumor progression as a regulator of the Wnt signaling pathway, of normal and tumor neo-angiogenesis and of immunity.
Collapse
Affiliation(s)
- Anne-Laure Nouvion
- Centre de cancérologie McGill, Département de biochimie, Université McGill, 3655 Promenade Sir-William-Osler, Montréal, Québec, H3G 1Y6, Canada
| | | |
Collapse
|