1
|
Zaimi B, Bazzano MV, Rauh M, Solano ME, Kappelmeyer M, Köninger A. sCEACAM-1 levels in maternal blood in case of threatened preterm birth. Eur J Obstet Gynecol Reprod Biol 2025; 307:230-235. [PMID: 39970542 DOI: 10.1016/j.ejogrb.2025.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/02/2025] [Accepted: 02/11/2025] [Indexed: 02/21/2025]
Abstract
INTRODUCTION This study aims to investigate the role of CEACAM1 in preterm birth. Preterm birth is a phenomenon with numerous triggers, with the immune system hypothesized to play a significant role in the process, aligning with the concept of 'birth as an immunological rejection phenomenon'. There are several approaches to predict preterm birth, and the determination of sCEACAM1 levels, a member of the carcinoembryonic antigen family, may serve as a potential candidate biomarker. METHODS A single-center prospective case series study included 67 pregnant women aged 18 years or older who presented before 37 weeks of gestation with signs of preterm birth in the years 2021-2023. At the time of admission, CEACAM1 was determined in maternal blood. RESULTS The median sCEACAM1 levels were significantly higher in women who delivered preterm compared to those who delivered at term respectively, 5014 pg/ml (IQR: 3592-8826) vs. 3353 pg/ml (IQR: 2354-5049) (p = 0.016). The median sCEACAM1 level in the group with PPROM (premature preterm rupture of membranes) at 34 weeks' gestation was 7001 pg/ml (IQR: 5683-13509), while the median sCEACAM1 level in the group without PPROM at 34 weeks' gestation was 3884 pg/ml (IQR; 2461-4985) (p < 0.001). CONCLUSIONS Pregnant women with preterm birth and/or PPROM before 34 weeks' gestation have higher CEACAM1 levels compared to women with threatened preterm labor who finally had labot at term. The results suggest early activated immune system as a potential pathomechanism of preterm delivery.
Collapse
Affiliation(s)
- Bora Zaimi
- University Department of Obstetrics and Gynaecology, Clinic St. Hedwig of The Order of St. John, University of Regensburg, Steinmetzstr. 1-3, D-93049 Regensburg, Germany.
| | - Maria Victoria Bazzano
- University Department of Obstetrics and Gynaecology, Clinic St. Hedwig of The Order of St. John, University of Regensburg, Steinmetzstr. 1-3, D-93049 Regensburg, Germany; Laboratory of Translational Perinatology, University of Regensburg, Biopark 1-3, D-93053 Regensburg, Germany
| | - Maximilian Rauh
- University Department of Obstetrics and Gynaecology, Clinic St. Hedwig of The Order of St. John, University of Regensburg, Steinmetzstr. 1-3, D-93049 Regensburg, Germany
| | - Maria Emilia Solano
- University Department of Obstetrics and Gynaecology, Clinic St. Hedwig of The Order of St. John, University of Regensburg, Steinmetzstr. 1-3, D-93049 Regensburg, Germany; Laboratory of Translational Perinatology, University of Regensburg, Biopark 1-3, D-93053 Regensburg, Germany
| | - Maurice Kappelmeyer
- University Department of Obstetrics and Gynaecology, Clinic St. Hedwig of The Order of St. John, University of Regensburg, Steinmetzstr. 1-3, D-93049 Regensburg, Germany; Laboratory of Translational Perinatology, University of Regensburg, Biopark 1-3, D-93053 Regensburg, Germany
| | - Angela Köninger
- University Department of Obstetrics and Gynaecology, Clinic St. Hedwig of The Order of St. John, University of Regensburg, Steinmetzstr. 1-3, D-93049 Regensburg, Germany
| |
Collapse
|
2
|
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
|
3
|
Dery KJ, Najjar SM, Beauchemin N, Shively JE, Kupiec‐Weglinski JW. Mechanism and function of CEACAM1 splice isoforms. Eur J Clin Invest 2024; 54 Suppl 2:e14350. [PMID: 39674874 PMCID: PMC11646291 DOI: 10.1111/eci.14350] [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: 09/01/2024] [Accepted: 10/29/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Alternative splicing is a fundamental mechanism in the post-transcriptional regulation of genes. The multifunctional transmembrane glycoprotein receptor carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) undergoes extensive alternative splicing to allow for tunable functions in cell signalling, adhesion and modulation of immune and metabolic responses. Splice isoforms that differ in their ectodomain and short or long cytoplasmic tail (CEACAM1-S/CEACAM1-L) have distinct functional roles. The mechanisms that regulate CEACAM1 RNA splicing remain elusive. METHODS This narrative review summarizes the current knowledge of the mechanism and function of CEACAM1 splice isoforms. Historical perspectives address the biological significance of the glycosylated Ig domains, the variable exon 7, and phosphorylation events that dictate its signal transduction pathways. The use of small antisense molecules to target mis-spliced variable exon 7 is discussed. RESULTS The Ig variable-like N domain mediates cell adhesion and immune checkpoint inhibitory functions. Gly and Tyr residues in the transmembrane (TM) domain are essential for dimerization. Calmodulin, Calcium/Calmodulin-dependent protein kinase II delta (CamK2D), Actin and Annexin A2 are binding partners of CEACAM1-S. Homology studies of the muCEACAM1-S and huCEACAM1-S TM predict differences in their signal transduction pathways. Hypoxia-inducible factor 1-α (HIF-1-α) induces alternative splicing to produce CEACAM1-S under limited oxygen conditions. Antisense small molecules directed to exon 7 may correct faulty expression of the short and long cytoplasmic tail splicing isoforms. CONCLUSION More pre-clinical and clinical studies are needed to elucidate the precise mechanisms by which CEACAM1 RNA splicing may be exploited to develop targeted interventions towards novel therapeutic strategies.
Collapse
Affiliation(s)
- Kenneth J. Dery
- Department of SurgeryUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Sonia M. Najjar
- Department of Biomedical Sciences, Heritage College of Osteopathic MedicineOhio UniversityAthensOhioUSA
| | - Nicole Beauchemin
- Rosalind and Morris Goodman Cancer InstituteMcGill UniversityMontrealQuebecCanada
| | - John E. Shively
- Department of Theranostics and Immunology, Arthur D. Riggs Diabetes and Metabolism Research Institute, Beckman Research InstituteCity of Hope National Medical CenterDuarteCaliforniaUSA
| | | |
Collapse
|
4
|
Götz L, Rueckschloss U, Najjar SM, Ergün S, Kleefeldt F. Carcinoembryonic antigen-related cell adhesion molecule 1 in cancer: Blessing or curse? Eur J Clin Invest 2024; 54 Suppl 2:e14337. [PMID: 39451132 DOI: 10.1111/eci.14337] [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: 09/03/2024] [Accepted: 10/04/2024] [Indexed: 10/26/2024]
Abstract
The Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1, also CD66a), a transmembrane glycoprotein of the immunoglobulin superfamily, is a pivotal mediator of various physiological and pathological processes, including oncologic disorders. However, its precise role in tumorigenicity is contradictory discussed by several clinical studies. This review aims to elucidate the clinical significance of CEACAM1 in different cancer entities focusing on tumour formation, progression and metastasis as well as on CEACAM1-mediated treatment resistance. Furthermore, we discuss the contribution of CEACAM1 to cancer immunity and modulation of the inflammatory microenvironment and finally provide a comprehensive review of treatment regimens targeting this molecule.
Collapse
Affiliation(s)
- Lisa Götz
- Institute of Anatomy and Cell Biology, University of Wuerzburg, Wuerzburg, Germany
| | - Uwe Rueckschloss
- Institute of Anatomy and Cell Biology, University of Wuerzburg, Wuerzburg, Germany
| | - Sonia M Najjar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine Irvine Hall, Ohio University, Athens, Ohio, USA
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, University of Wuerzburg, Wuerzburg, Germany
| | - Florian Kleefeldt
- Institute of Anatomy and Cell Biology, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
5
|
Götz L, Rueckschloss U, Balk G, Pfeiffer V, Ergün S, Kleefeldt F. The role of carcinoembryonic antigen-related cell adhesion molecule 1 in cancer. Front Immunol 2023; 14:1295232. [PMID: 38077351 PMCID: PMC10704240 DOI: 10.3389/fimmu.2023.1295232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
The Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), also known as CD66a, is a member of the immunoglobulin superfamily. CEACAM1 was shown to be a prognostic marker in patients suffering from cancer. In this review, we summarize pre-clinical and clinical evidence linking CEACAM1 to tumorigenicity and cancer progression. Furthermore, we discuss potential CEACAM1-based mechanisms that may affect cancer biology.
Collapse
Affiliation(s)
- Lisa Götz
- Institute of Anatomy and Cell Biology, Julius‐Maximilians‐University Würzburg, Würzburg, Germany
| | - Uwe Rueckschloss
- Institute of Anatomy and Cell Biology, Julius‐Maximilians‐University Würzburg, Würzburg, Germany
| | - Gözde Balk
- Institute of Anatomy and Cell Biology, Julius‐Maximilians‐University Würzburg, Würzburg, Germany
| | - Verena Pfeiffer
- Institute of Anatomy and Cell Biology, Julius‐Maximilians‐University Würzburg, Würzburg, Germany
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius‐Maximilians‐University Würzburg, Würzburg, Germany
| | - Florian Kleefeldt
- Institute of Anatomy and Cell Biology, Julius‐Maximilians‐University Würzburg, Würzburg, Germany
- Harvard Stem Cell Institute, Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, United States
| |
Collapse
|
6
|
Zhang L, Zhang J, Xuan X, Wu D, Yu J, Wang P, Yang X, Zhang J, Gan W, He M, Liu XM, Zhou J, Wang D, Gu W, Li D. A p53/LINC00324 positive feedback loop suppresses tumor growth by counteracting SET-mediated transcriptional repression. Cell Rep 2023; 42:112833. [PMID: 37480565 DOI: 10.1016/j.celrep.2023.112833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 05/26/2023] [Accepted: 07/03/2023] [Indexed: 07/24/2023] Open
Abstract
The p53 tumor suppressor exerts antitumor functions through its ability to regulate the transcription of its downstream targets. Long noncoding RNAs (lncRNAs) act as oncogenes or tumor suppressors implicated in tumorigenesis and tumor progression. Here, we identify the lncRNA LINC00324 (long intergenic noncoding RNA 00324) as a direct p53 transcriptional target. Knockdown of LINC00324 expression promotes tumor growth by reducing p53 transcriptional activity, whereas ectopic LINC00324 expression demonstrates a reverse effect. Notably, LINC00324 is present in the endogenous p53 complex in tumor cells and directly binds to the C-terminal domain of p53 in vitro. Mechanistically, LINC00324 enables p53 transactivation by competitively disrupting the p53-SET interaction, resulting in an increase of p300/CBP-mediated H3K18 and H3K27 acetylation on the p53 target promoters. Lower LINC00324 expression is associated with more aggressive disease status and predicts worse overall survival of patients with cancer. Our study identifies a p53/LINC00324 positive feedback loop that suppresses tumor growth by counteracting SET-mediated transcriptional repression.
Collapse
Affiliation(s)
- Ling Zhang
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, 68 Jiyang West Road, Suzhou 215600, China
| | - Jun Zhang
- School of Life Science and Technology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Xiaofeng Xuan
- Department of Respiratory & Critical Care Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, 68 Jiyang West Road, Suzhou 215600, China
| | - Di Wu
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, 68 Jiyang West Road, Suzhou 215600, China
| | - Jianfeng Yu
- Department of Life Science and Technology, Changshu Institute of Technology, 99 South Third Ring Road, Suzhou 215500, China
| | - Peizhen Wang
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, 68 Jiyang West Road, Suzhou 215600, China
| | - Xiaomei Yang
- Department of Emergency, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, 68 Jiyang West Road, Suzhou 215600, China
| | - Jieru Zhang
- Department of Respiratory & Critical Care Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, 68 Jiyang West Road, Suzhou 215600, China
| | - Wenjuan Gan
- Department of Pathology, Dushu Lake Hospital Affiliated to Soochow University, 9 Chongwen Road, Suzhou 215300, China
| | - Mengfan He
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, 68 Jiyang West Road, Suzhou 215600, China
| | - Xiao-Min Liu
- School of Life Science and Technology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Jun Zhou
- School of Life Science and Technology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Donglai Wang
- State Key Laboratory of Medical Molecular Biology and Department of Medical Genetics, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Wei Gu
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center, College of Physicians & Surgeons, Columbia University, 1130 St. Nicholas Avenue, New York, NY 10032, USA
| | - Dawei Li
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, 68 Jiyang West Road, Suzhou 215600, China.
| |
Collapse
|
7
|
Lima K, Ribas GT, Riella LV, Borges TJ. Inhibitory innate receptors and their potential role in transplantation. Transplant Rev (Orlando) 2023; 37:100776. [PMID: 37451057 DOI: 10.1016/j.trre.2023.100776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023]
Abstract
The regulatory arm of the immune system plays a crucial role in maintaining immune tolerance and preventing excessive immune responses. Immune regulation comprises various regulatory cells and molecules that work together to suppress or regulate immune responses. The programmed cell death protein 1 (PD-1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4) are examples of inhibitory receptors that counteract activating signals and fine-tune immune responses. While most of the discoveries of immune regulation have been related to T cells and the adaptive immune system, the innate arm of the immune system also has a range of inhibitory receptors that can counteract activating signals and suppress the effector immune responses. Targeting these innate inhibitory receptors may provide a complementary therapeutic approach in several immune-related conditions, including transplantation. In this review, we will explore the potential role of innate inhibitory receptors in controlling alloimmunity during solid organ transplantation.
Collapse
Affiliation(s)
- Karina Lima
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Guilherme T Ribas
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Professional and Technological Education Sector, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Leonardo V Riella
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Thiago J Borges
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
8
|
CEACAMS 1, 5, and 6 in disease and cancer: interactions with pathogens. Genes Cancer 2023; 14:12-29. [PMID: 36741860 PMCID: PMC9891707 DOI: 10.18632/genesandcancer.230] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 01/20/2023] [Indexed: 02/04/2023] Open
Abstract
The CEA family comprises 18 genes and 11 pseudogenes located at chromosome 19q13.2 and is divided into two main groups: cell surface anchored CEA-related cell adhesion molecules (CEACAMs) and the secreted pregnancy-specific glycoproteins (PSGs). CEACAMs are highly glycosylated cell surface anchored, intracellular, and intercellular signaling molecules with diverse functions, from cell differentiation and transformation to modulating immune responses associated with infection, inflammation, and cancer. In this review, we explore current knowledge surrounding CEACAM1, CEACAM5, and CEACAM6, highlight their pathological significance in the areas of cancer biology, immunology, and inflammatory disease, and describe the utility of murine models in exploring questions related to these proteins.
Collapse
|
9
|
Madani J, Aghebati-Maleki L, Gharibeh N, Pourakbari R, Yousefi M. Fetus, as an allograft, evades the maternal immunity. Transpl Immunol 2022; 75:101728. [DOI: 10.1016/j.trim.2022.101728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 10/09/2022] [Accepted: 10/09/2022] [Indexed: 11/05/2022]
|
10
|
Carlsen L, Schorl C, Huntington K, Hernandez-Borrero L, Jhaveri A, Zhang S, Zhou L, El-Deiry WS. Pan-drug and drug-specific mechanisms of 5-FU, irinotecan (CPT-11), oxaliplatin, and cisplatin identified by comparison of transcriptomic and cytokine responses of colorectal cancer cells. Oncotarget 2021; 12:2006-2021. [PMID: 34611476 PMCID: PMC8487728 DOI: 10.18632/oncotarget.28075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 08/28/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) caused over 900,000 deaths worldwide in 2020. A majority of late-stage CRC patients are treated with 5-fluorouracil (5-FU) combined with either irinotecan (CPT-11), oxaliplatin, or both. Despite their widespread use, the mechanisms of efficacy and toxicity of these drugs remain incompletely understood. While previous work has investigated cellular responses to these agents individually, we directly compare the transcriptomic and cytokine profiles of HCT116 wild-type and p53-/- colorectal cancer cells treated with these drugs and report pan-drug, drug-specific, drug class-specific, p53-independent, and p53-dependent signatures. We observed downregulation of histone genes by 5-FU (that significantly correlates with improved survival in CRC patients) and upregulation of FOS and ATF3 by oxaliplatin (which may contribute to peripheral neuropathy). BTG2 was identified as a top gene upregulated by all four drugs, suggesting its critical role in the cellular response to chemotherapy in CRC. Soluble TRAILR2 (death receptor 5; DR5) is a decoy receptor for TRAIL, an apoptosis-inducing cytokine. TRAILR2 was down-regulated by oxaliplatin and 5-FU, was not affected by CPT-11, and was increased by cisplatin. There was an increase in IL-8 by oxaliplatin and increase in ferritin by cisplatin which may contribute to cancer cell survival. Novel drug-specific mechanisms of efficacy or toxicity identified in these signatures may be targeted with combination therapies or development of new targeted therapies. Together, the findings here contribute to our understanding of the molecular bases of efficacy and toxicity of chemotherapeutic agents often used for treatment of GI cancer such as CRC.
Collapse
Affiliation(s)
- Lindsey Carlsen
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, USA.,Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,Pathobiology Graduate Program, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Christoph Schorl
- The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, USA.,Department of Molecular Biology, Cell Biology and Biochemistry, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,Genomics Core Facility, Brown University, Providence, RI 02903, USA.,Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Kelsey Huntington
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, USA.,Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,Pathobiology Graduate Program, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Liz Hernandez-Borrero
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, USA.,Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,Pathobiology Graduate Program, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Aakash Jhaveri
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Shengliang Zhang
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, USA.,Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Lanlan Zhou
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, USA.,Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Wafik S El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, USA.,Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,Pathobiology Graduate Program, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,Hematology-Oncology Division, Department of Medicine, Rhode Island Hospital and Brown University, Providence, RI 02903, USA.,Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| |
Collapse
|
11
|
Gandhi AK, Sun ZYJ, Kim WM, Huang YH, Kondo Y, Bonsor DA, Sundberg EJ, Wagner G, Kuchroo VK, Petsko GA, Blumberg RS. Structural basis of the dynamic human CEACAM1 monomer-dimer equilibrium. Commun Biol 2021; 4:360. [PMID: 33742094 PMCID: PMC7979749 DOI: 10.1038/s42003-021-01871-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 02/18/2021] [Indexed: 02/07/2023] Open
Abstract
Human (h) carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) function depends upon IgV-mediated homodimerization or heterodimerization with host ligands, including hCEACAM5, hTIM-3, PD-1, and a variety of microbial pathogens. However, there is little structural information available on how hCEACAM1 transitions between monomeric and dimeric states which in the latter case is critical for initiating hCEACAM1 activities. We therefore mutated residues within the hCEACAM1 IgV GFCC' face including V39, I91, N97, and E99 and examined hCEACAM1 IgV monomer-homodimer exchange using differential scanning fluorimetry, multi-angle light scattering, X-ray crystallography and/or nuclear magnetic resonance. From these studies, we describe hCEACAM1 homodimeric, monomeric and transition states at atomic resolution and its conformational behavior in solution through NMR assignment of the wildtype (WT) hCEACAM1 IgV dimer and N97A mutant monomer. These studies reveal the flexibility of the GFCC' face and its important role in governing the formation of hCEACAM1 dimers and selective heterodimers.
Collapse
Affiliation(s)
- Amit K Gandhi
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Zhen-Yu J Sun
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Walter M Kim
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yu-Hwa Huang
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yasuyuki Kondo
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Department of Internal Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Daniel A Bonsor
- Institute of Human Virology, University of Maryland School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Eric J Sundberg
- Institute of Human Virology, University of Maryland School of Medicine, University of Maryland, Baltimore, MD, USA
- Department of Medicine, University of Maryland School of Medicine, University of Maryland, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, University of Maryland, Baltimore, MD, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Gerhard Wagner
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Gregory A Petsko
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Richard S Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
12
|
Hong L, Tang Y, Pan S, Xu M, Shi Y, Gao S, Sui C, He C, Zheng K, Tang R, Shi Z, Wang Q, Wang H. Interleukin 3-induced GITR promotes the activation of human basophils. Cytokine 2020; 136:155268. [PMID: 32889153 DOI: 10.1016/j.cyto.2020.155268] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/22/2020] [Accepted: 08/24/2020] [Indexed: 01/01/2023]
Abstract
Human basophils regulate allergic reactions by secreting histamine, interleukin 4 (IL-4) and IL-13 through key surface receptors FcεRI as well as IL-3R, which are constitutively expressed on basophils. IL-3/IL-3R signaling axis plays key roles in regulating the development and activation of basophils. We and others have shown that IL-3-induced surface receptors e.g. ST2, IL-17RB and IL-2 receptors regulate the biology of basophils. However, the expression and function of IL-3-induced surface proteins on human basophils remain to be elucidated. We in this study aimed to identify new basophil activation regulators by transcriptomic analysis of IL-3-stimulated basophils. Gene expression microarray analysis of IL-3-treated basophils revealed 2050 differentially expressed genes, of which 323 genes encoded surface proteins including GITR. We identified that GITR was preferentially induced by IL-3 rather than anti-IgE, IL-33, fMLP and C5a. IL-3-induced GITR was suppressed by inhibitors targeting JAK2, PI3K and MEK1/2. Stimulation of IL-3-treated basophils by GITR enhanced the expression of IL-4 and IL-13. Moreover, IgE-mediated degranulation was enhanced by GITRL in the presence of IL-3. This transcriptomic analysis of IL-3-activated basophils helps to identify novel activation regulator. IL-3-induced GITR promoted the activation of basophils, adding new evidence supporting GITR as an important player in Th2-associated immune responses.
Collapse
Affiliation(s)
- Li Hong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yangyang Tang
- Department of Pathology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Shuai Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Meizhen Xu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yanbiao Shi
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Sijia Gao
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Chao Sui
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Cheng He
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - KuiYang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Renxian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Zhixu Shi
- Xuzhou Red Cross Blood Center, Xuzhou, Jiangsu 221400, China
| | - Qingling Wang
- Department of Pathology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Hui Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
13
|
Liu J, Muturi HT, Khuder SS, Helal RA, Ghadieh HE, Ramakrishnan SK, Kaw MK, Lester SG, Al-Khudhair A, Conran PB, Chin KV, Gatto-Weis C, Najjar SM. Loss of Ceacam1 promotes prostate cancer progression in Pten haploinsufficient male mice. Metabolism 2020; 107:154215. [PMID: 32209360 PMCID: PMC7283002 DOI: 10.1016/j.metabol.2020.154215] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/10/2020] [Accepted: 03/19/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE PTEN haploinsufficiency plays an important role in prostate cancer development in men. However, monoallelic deletion of Pten gene failed to induce high prostate intraepithelial neoplasia (PIN) until Pten+/- mice aged or fed a high-calorie diet. Because CEACAM1, a cell adhesion molecule with a potential tumor suppression activity, is induced in Pten+/- prostates, the study aimed at examining whether the rise of CEACAM1 limited neoplastic progression in Pten+/- prostates. METHODS Pten+/- were crossbred with Cc1-/- mice harboring a null deletion of Ceacam1 gene to produce Pten+/-/Cc1-/- double mutants. Prostates from 7-month old male mice were analyzed histologically and biochemically for PIN progression. RESULTS Deleting Ceacam1 in Pten+/- mice caused an early development of high-grade PIN in parallel to hyperactivation of PI3 kinase/Akt and Ras/MAP kinase pathways, with an increase in cell proliferation, epithelial-to-mesenchymal transition, angiogenesis and inflammation relative to Pten+/- and Cc1-/- individual mutants. It also caused a remarkable increase in lipogenesis in prostate despite maintaining insulin sensitivity. Concomitant Ceacam1 deletion with Pten+/- activated the IL-6/STAT3 signaling pathways to suppress Irf-8 transcription that in turn, led to a decrease in the expression level of promyelocytic leukemia gene, a well characterized tumor suppressor in prostate. CONCLUSIONS Ceacam1 deletion accelerated high-grade prostate intraepithelial neoplasia in Pten haploinsufficient mice while preserving insulin sensitivity. This demonstrated that the combined loss of Ceacam1 and Pten advanced prostate cancer by increasing lipogenesis and modifying the STAT3-dependent inflammatory microenvironment of prostate.
Collapse
Affiliation(s)
- Jehnan Liu
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Harrison T Muturi
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Saja S Khuder
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Raghd Abu Helal
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Hilda E Ghadieh
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Sadeesh K Ramakrishnan
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Meenakshi K Kaw
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Sumona Ghosh Lester
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Ahmed Al-Khudhair
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Philip B Conran
- Department of Pathology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Khew-Voon Chin
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Cara Gatto-Weis
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; Department of Pathology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Sonia M Najjar
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA; Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA.
| |
Collapse
|
14
|
Xiong D, Wang Y, You M. Tumor intrinsic immunity related proteins may be novel tumor suppressors in some types of cancer. Sci Rep 2019; 9:10918. [PMID: 31358815 PMCID: PMC6662687 DOI: 10.1038/s41598-019-47382-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 07/11/2019] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint blockade therapy (ICBT) can unleash T-cell responses against cancer. However, only a small fraction of patients exhibited responses to ICBT. The role of immune checkpoints in cancer cells is not well understood. In this study, we analyzed T-cell coinhibitory/costimulatory genes across more than 1100 samples of the Cancer Cell Line Encyclopedia (CCLE). Nearly 90% of such genes were not expressed or had low expression across the CCLE cancer cell lines. Cell line screening showed the enrichment of cancer cells deprived of the expression of CD27, CEACAM1, CTLA4, LRIG1, PDCD1LG2, or TNFRSF18, suggesting their role as tumor suppressor. The metagene expression signature derived from these six genes - Immu6Metagene was associated with prolonged survival phenotypes. A common set of five oncogenic pathways were significantly inhibited in different types of tumors of the cancer patients with good survival outcome and high Immu6Metagene signature expression. These pathways were TGF-β signaling, angiogenesis, EMT, hypoxia and mitotic process. Our study showed that oncoimmunology related molecules especially the six genes of the Immu6Metagene signature may play the tumor suppressor role in certain cancers. Therefore, the ICBT targeting them should be considered in such context to improve the efficacy.
Collapse
Affiliation(s)
- Donghai Xiong
- Center for Disease Prevention Research and Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Yian Wang
- Center for Disease Prevention Research and Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Ming You
- Center for Disease Prevention Research and Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
15
|
Kim WM, Huang YH, Gandhi A, Blumberg RS. CEACAM1 structure and function in immunity and its therapeutic implications. Semin Immunol 2019; 42:101296. [PMID: 31604530 PMCID: PMC6814268 DOI: 10.1016/j.smim.2019.101296] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/01/2019] [Indexed: 12/13/2022]
Abstract
The type I membrane protein receptor carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) distinctively exhibits significant alternative splicing that allows for tunable functions upon homophilic binding. CEACAM1 is highly expressed in the tumor environment and is strictly regulated on lymphocytes such that its expression is restricted to activated cells where it is now recognized to function in tolerance pathways. CEACAM1 is also an important target for microbes which have co-opted these attributes of CEACAM1 for the purposes of invading the host and evading the immune system. These properties, among others, have focused attention on CEACAM1 as a unique target for immunotherapy in autoimmunity and cancer. This review examines recent structural information derived from the characterization of CEACAM1:CEACAM1 interactions and heterophilic modes of binding especially to microbes and how this relates to CEACAM1 function. Through this, we aim to provide insights into targeting CEACAM1 for therapeutic intervention.
Collapse
Affiliation(s)
- Walter M Kim
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Yu-Hwa Huang
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Amit Gandhi
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Richard S Blumberg
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.
| |
Collapse
|
16
|
Nichita L, Zurac S, Bastian A, Stinga P, Nedelcu R, Brinzea A, Turcu G, Ion D, Jilaveanu L, Sticlaru L, Popp C, Cioplea M. Comparative analysis of CEACAM1 expression in thin melanomas with and without regression. Oncol Lett 2019; 17:4149-4154. [PMID: 30944609 PMCID: PMC6444332 DOI: 10.3892/ol.2019.10067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/18/2018] [Indexed: 11/29/2022] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is a key molecule in several intracellular and intercellular signaling pathways, with multiple functional and structural roles. CEACAM1 expression in melanoma is often described in the invading part of the tumor and has been associated with increased melanoma cells invasion and migration. We studied CEACAM1 expression in regressing versus non-regressing thin melanomas, knowing that phenomenon of regression represents a valuable model for understanding tumor immunity. In melanoma, through homophilic interactions, CEACAM1 inhibits natural killer cell activity, inhibits effector functions of tumor infiltrating lymphocytes, such as cytotoxicity and interferon-γ release. We present a retrospective study including 53 consecutive cases of thin melanoma, 21 with regression and 32 without regression. Comparative analysis of CEACAM1 expression in regressed and non-regressed areas from melanomas with regression and in non-regressed melanomas was performed. We used three different clones of CEACAM1: AA 1-428, extracellular domain, rabbit; AA 1-428, mouse, clone 8B6E2F4; and AA 1-468, full length, mouse, clone 2F6. All three clones had similar reactivity. We identified membrane positivity of tumor cells in non-regressed melanomas and in non-regressed areas in melanomas with regression. Remaining tumor cells in regressed areas were mostly negative for CEACAM1. In non-regressed lesions, there was a stronger positivity of CEACAM1 in the deep invasive front. In thin melanomas, CEACAM1 overexpression is related with invasiveness, suggesting that CEACAM1-positive melanomas are more aggressive. Also, in areas of regression tumor cells lose CEACAM1 expression, probably correlated with the presence of natural killer cells.
Collapse
Affiliation(s)
- Luciana Nichita
- Department of Pathology, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 010221 Bucharest, Romania.,Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
| | - Sabina Zurac
- Department of Pathology, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 010221 Bucharest, Romania.,Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
| | - Alexandra Bastian
- Department of Pathology, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 010221 Bucharest, Romania.,Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
| | - Patricia Stinga
- Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
| | - Roxana Nedelcu
- Department of Physiopathology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Alice Brinzea
- Department of Physiopathology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Gabriela Turcu
- Department of Physiopathology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Dermatology, Colentina University Hospital, 020125 Bucharest, Romania
| | - Daniela Ion
- Department of Physiopathology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Lucia Jilaveanu
- Department of Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT 208028, USA
| | - Liana Sticlaru
- Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
| | - Cristiana Popp
- Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
| | - Mirela Cioplea
- Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
| |
Collapse
|
17
|
Calinescu A, Turcu G, Nedelcu RI, Brinzea A, Hodorogea A, Antohe M, Diaconu C, Bleotu C, Pirici D, Jilaveanu LB, Ion DA, Badarau IA. On the Dual Role of Carcinoembryonic Antigen-Related Cell Adhesion Molecule 1 (CEACAM1) in Human Malignancies. J Immunol Res 2018; 2018:7169081. [PMID: 30406153 PMCID: PMC6204181 DOI: 10.1155/2018/7169081] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 09/05/2018] [Indexed: 11/26/2022] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is a glycoprotein belonging to the carcinoembryonic antigen (CEA) family that is expressed on a wide variety of cells and holds a complex role in inflammation through its alternate splicing and generation of various isoforms, mediating intricate mechanisms of modulation and dysregulation. Initially regarded as a tumor suppressor as its expression shows considerable downregulation within the epithelia in the early phases of many solid cancers, CEACAM1 has been linked lately to the progression of malignancy and metastatic spread as various papers point to its role in tumor progression, angiogenesis, and invasion. We reviewed the literature and discussed the various expression patterns of CEACAM1 in different types of tumors, describing its structure and general biologic functions and emphasizing the most significant findings that link this molecule to poor prognosis. The importance of understanding the role of CEACAM1 in cell transformation stands not only in this adhesion molecule's value as a prognostic factor but also in its promising premise as a potential new molecular target that could be exploited as a specific cancer therapy.
Collapse
Affiliation(s)
- Andreea Calinescu
- Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Dermatology 1 Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Gabriela Turcu
- Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Dermatology 1 Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
- Derma 360° Clinic, 011273 Bucharest, Romania
| | - Roxana I. Nedelcu
- Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Derma 360° Clinic, 011273 Bucharest, Romania
- National Institute for Infectious Diseases Prof. Dr. Matei Balș, 021105 Bucharest, Romania
| | - Alice Brinzea
- Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- National Institute for Infectious Diseases Prof. Dr. Matei Balș, 021105 Bucharest, Romania
| | - Anastasia Hodorogea
- Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Dermatology 1 Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Mihaela Antohe
- Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Derma 360° Clinic, 011273 Bucharest, Romania
| | - Carmen Diaconu
- Stefan Nicolau Institute of Virology, 030304 Bucharest, Romania
| | - Coralia Bleotu
- Stefan Nicolau Institute of Virology, 030304 Bucharest, Romania
| | - Daniel Pirici
- University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Lucia B. Jilaveanu
- Department of Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT 208028, USA
| | - Daniela A. Ion
- Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Ioana A. Badarau
- Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
18
|
McLeod RL, Angagaw MH, Baral TN, Liu L, Moniz RJ, Laskey J, Hsieh S, Lee M, Han JH, Issafras H, Javaid S, Loboda A, Sadekova S, O'Connor JA, Tse A, Punnonen J. Characterization of murine CEACAM1 in vivo reveals low expression on CD8 + T cells and no tumor growth modulating activity by anti-CEACAM1 mAb CC1. Oncotarget 2018; 9:34459-34470. [PMID: 30349641 PMCID: PMC6195382 DOI: 10.18632/oncotarget.26108] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 08/27/2018] [Indexed: 12/17/2022] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) has been reported to mediate both tumorigenic and anti-tumor effects in vivo. Blockade of the CEACAM1 signaling pathway has recently been implicated as a novel mechanism for cancer immunotherapy. CC1, a mouse anti-CEACAM1 monoclonal antibody (mAb), has been widely used as a pharmacological tool in preclinical studies to inform on CEACAM1 pathway biology although limited data are available on its CEACAM1 blocking characteristics or pharmacodynamic-pharmacokinetic profiles. We sought to investigate CEACAM1 expression on mouse tumor and immune cells, characterize CC1 mAb binding, and evaluate CC1 in syngeneic mouse oncology models as a monotherapy and in combination with an anti-PD-1 mAb. CEACAM1 expression was observed at high levels on neutrophils, NK cells and myeloid-derived suppressor cells (MDSCs), while the expression on tumor-infiltrating CD8+ T cells was low. Unexpectedly, rather than blocking, CC1 facilitated binding of soluble CEACAM1 to CEACAM1 expressing cells. No anti-tumor effects were observed in CT26, MBT2 or A20 models when tested up to 30 mg/kg dose, a dose that was estimated to achieve >90% target engagement in vivo. Taken together, tumor infiltrating CD8+ T cells express low levels of CEACAM1 and CC1 Ab mediates no or minimal anti-tumor effects in vivo, as a monotherapy or in combination with anti-PD-1 treatment.
Collapse
Affiliation(s)
- Robbie L McLeod
- Merck & Co., Inc., Boston, MA, USA.,Merck & Co., Inc., Kenilworth, NJ, USA.,Merck & Co., Inc., Palo Alto, CA, USA
| | - Minilik H Angagaw
- Merck & Co., Inc., Boston, MA, USA.,Merck & Co., Inc., Kenilworth, NJ, USA.,Merck & Co., Inc., Palo Alto, CA, USA
| | - Toya Nath Baral
- Merck & Co., Inc., Boston, MA, USA.,Merck & Co., Inc., Kenilworth, NJ, USA.,Merck & Co., Inc., Palo Alto, CA, USA
| | - Liming Liu
- Merck & Co., Inc., Boston, MA, USA.,Merck & Co., Inc., Kenilworth, NJ, USA.,Merck & Co., Inc., Palo Alto, CA, USA
| | - Raymond Joseph Moniz
- Merck & Co., Inc., Boston, MA, USA.,Merck & Co., Inc., Kenilworth, NJ, USA.,Merck & Co., Inc., Palo Alto, CA, USA
| | - Jason Laskey
- Merck & Co., Inc., Boston, MA, USA.,Merck & Co., Inc., Kenilworth, NJ, USA.,Merck & Co., Inc., Palo Alto, CA, USA
| | - SuChun Hsieh
- Merck & Co., Inc., Boston, MA, USA.,Merck & Co., Inc., Kenilworth, NJ, USA.,Merck & Co., Inc., Palo Alto, CA, USA
| | - Mike Lee
- Merck & Co., Inc., Boston, MA, USA.,Merck & Co., Inc., Kenilworth, NJ, USA.,Merck & Co., Inc., Palo Alto, CA, USA
| | - Jin-Hwan Han
- Merck & Co., Inc., Boston, MA, USA.,Merck & Co., Inc., Kenilworth, NJ, USA.,Merck & Co., Inc., Palo Alto, CA, USA
| | - Hassan Issafras
- Merck & Co., Inc., Boston, MA, USA.,Merck & Co., Inc., Kenilworth, NJ, USA.,Merck & Co., Inc., Palo Alto, CA, USA
| | - Sarah Javaid
- Merck & Co., Inc., Boston, MA, USA.,Merck & Co., Inc., Kenilworth, NJ, USA.,Merck & Co., Inc., Palo Alto, CA, USA
| | - Andrey Loboda
- Merck & Co., Inc., Boston, MA, USA.,Merck & Co., Inc., Kenilworth, NJ, USA.,Merck & Co., Inc., Palo Alto, CA, USA
| | - Svetlana Sadekova
- Merck & Co., Inc., Boston, MA, USA.,Merck & Co., Inc., Kenilworth, NJ, USA.,Merck & Co., Inc., Palo Alto, CA, USA
| | - Joann A O'Connor
- Merck & Co., Inc., Boston, MA, USA.,Merck & Co., Inc., Kenilworth, NJ, USA.,Merck & Co., Inc., Palo Alto, CA, USA
| | - Archie Tse
- Merck & Co., Inc., Boston, MA, USA.,Merck & Co., Inc., Kenilworth, NJ, USA.,Merck & Co., Inc., Palo Alto, CA, USA
| | - Juha Punnonen
- Merck & Co., Inc., Boston, MA, USA.,Merck & Co., Inc., Kenilworth, NJ, USA.,Merck & Co., Inc., Palo Alto, CA, USA
| |
Collapse
|
19
|
Abouelghar A, Hasnah R, Taouk G, Saad M, Karam M. Prognostic values of the mRNA expression of natural killer receptor ligands and their association with clinicopathological features in breast cancer patients. Oncotarget 2018; 9:27171-27196. [PMID: 29930758 PMCID: PMC6007477 DOI: 10.18632/oncotarget.25506] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/14/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Natural killer (NK) cells are lymphocytes of the innate immune system that have potent cytotoxic activity against tumor cells. NK cell recognition and activity towards cancer cells are regulated by an integrated interplay between numerous inhibitory and activating receptors acting in concert to eliminate tumor cells expressing cognate ligands. Despite strong evidence supporting the role of NK cells in breast cancer (BC) control, BC still develops and progresses to form large tumors and metastases. A major mechanism of BC escape from NK immunity is the alteration of the expression of NK receptor ligands. The aim of this study was to determine whether NK receptor ligands' mRNA expression might influence prognosis in BC patients and whether these effects differ by molecular subtypes and clinicopathological features. METHODS We used the KM plotter platform to analyze the correlation between mRNA expression of 32 NK receptor ligands and relapse-free survival (RFS) and overall survival (OS) in 3951 and 1402 BC patients, respectively. The association with tumor subtypes and clinicopathological features was determined. BC samples were split into high and low expression groups according to the best cutoff value and the two patient cohorts were compared by Kaplan-Meier survival plots. The hazard ratios with 95% confidence intervals and log rank P values were calculated and FDR-adjusted for multiple testing correction. The data was considered to be statistically significant when FDR-adjusted P value < 0.05. RESULTS High mRNA expression of around 80% of ligands for NK activating and inhibitory receptors associated with better RFS, which correlated with longer OS for only about half of the NK-activating ligands but for most NK-inhibitory ligands. Also, five NK-activating ligands correlated with worse prognosis. These prognostic values were differentially associated with the BC clinical criteria. In addition, the favorable prognostic influence of NK-activating ligands' upregulation, as a whole, was mainly significantly associated with HER2-positive and basal-like subtypes, lymph node positive phenotype, and high-grade tumors. CONCLUSIONS NK receptor ligands appear to play an important role in defining BC patient prognosis. Identification of a group of patients with worse prognosis expressing high levels of NK-activating ligands and low levels of NK-inhibitory ligands makes them ideal potential candidates for NK-based immunotherapy to eliminate residual tumor cells, prevent relapse and improve patient survival.
Collapse
Affiliation(s)
- Ali Abouelghar
- Cancer Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Reem Hasnah
- Department of Biological Sciences, Carnegie Mellon University in Qatar, Doha, Qatar
| | - Ghina Taouk
- Cancer Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Mohamad Saad
- Qatar Computing Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Manale Karam
- Cancer Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| |
Collapse
|
20
|
Knockdown of CEACAM19 suppresses human gastric cancer through inhibition of PI3K/Akt and NF-κB. Surg Oncol 2018; 27:495-502. [PMID: 30217308 DOI: 10.1016/j.suronc.2018.05.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 05/02/2018] [Indexed: 12/15/2022]
Abstract
Gastric cancer directly affects the quality of human life worldwide. Some members, which belong to carcinoembryonic antigen-related cell adhesion molecule (CEACAM) subfamily, are deregulated in tumors. Of the subfamily, CEACAM19, a new member was the research object. Our study sought to explore the potential role of CEACAM19 in gastric cancer. According to the immunohistochemistry (IHC), RT-PCR and Western blot, CEACAM19 was over-expressed in gastric cancer tissues and cells. Moreover, the Western blot analysis showed that the expression of MMP2 and MMP9 was inhibited in CEACAM19 knockdown gastric cancer cells. Meanwhile, in SGC-7901 and MGC-803 cells, the knockdown of CEACAM19 reduced proliferation, migration and invasion. Additionally, the Western blot assay revealed that the phosphorylation levels of Akt and p65 were declined by the knockdown of CEACAM19. Furthermore, the influence of CEACAM19 knockdown was confirmed by the studies in vivo. Collectively, our results revealed that the CEACAM19 knockdown prevented the gastric cancer progression likely related to inactivating the PI3K/Akt and NF-κB signaling pathways. Our findings provided insights into a promising biomarker of gastric cancer and the potential molecule clues for the prevention of gastric cancer.
Collapse
|
21
|
Prévostel C, Blache P. The dose-dependent effect of SOX9 and its incidence in colorectal cancer. Eur J Cancer 2017; 86:150-157. [DOI: 10.1016/j.ejca.2017.08.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 08/24/2017] [Accepted: 08/30/2017] [Indexed: 10/18/2022]
|
22
|
Simonetti O, Lucarini G, Rubini C, Zizzi A, Aspriello SD, Di Primio R, Offidani AM. Correlation between immunohistochemical staining of CEACAM1 and clinicopathological findings in oral pre-neoplastic lesions and squamous cell carcinoma. Med Mol Morphol 2017; 51:41-47. [PMID: 28887602 DOI: 10.1007/s00795-017-0169-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 08/21/2017] [Indexed: 12/11/2022]
Abstract
Squamous cell carcinoma of the oral cavity represents the sixth most common cancer worldwide and it is often preceded by pre-neoplastic lesions. Sometimes it is still difficult for pathologists to make objective differential diagnoses only on histological characteristics. Tumorigenesis is accompanied by altered expression of cell adhesion molecules, like carcinoembryonic antigen cell adhesion molecule (CEACAM)1. We wanted to investigative CEACAM1 in oral dysplastic lesions, carcinoma in situ (CIS) and oral squamous cell carcinoma (OSCC). We examined immunohistochemical CEACAM1 expression in 50 OSCC, 30 oral CIS and 40 pre-neoplastic lesions and assessed its correlation with clinical and pathological parameters. CEACAM1 was not expressed in normal mucosa, significantly expressed in CIS while it was negative in all the dysplastic lesions. In OSCC, high CEACAM1 expression was associated with tumor grade and inversely correlated with both overall and disease-specific 5-year survival. We showed that CEACAM1 expression is very dynamic: absent in dysplastic lesions, up-regulated in CIS and OSCC. We suggest that CEACAM1 could be a prognostic marker of OSCC and oral CIS. Our most important finding was that it could help pathologists diagnosing oral carcinoma in situ.
Collapse
Affiliation(s)
- Oriana Simonetti
- Department of Dermatology, Polytechnic University of Marche, Torrette, Ancona, Italy
| | - Guendalina Lucarini
- Department of Clinic and Molecular Sciences, Histology, Polytechnic University of Marche, Via Tronto 10/a, Torrette, 60020, Ancona, Italy.
| | - Corrado Rubini
- Department of Biomedical Sciences and Public Health, Section of Pathologic Anatomy and Histopathology, Polytechnic University of Marche, Torrette, Ancona, Italy
| | - Antonio Zizzi
- Department of Biomedical Sciences and Public Health, Section of Pathologic Anatomy and Histopathology, Polytechnic University of Marche, Torrette, Ancona, Italy
| | | | - Roberto Di Primio
- Department of Clinic and Molecular Sciences, Histology, Polytechnic University of Marche, Via Tronto 10/a, Torrette, 60020, Ancona, Italy
| | - Anna Maria Offidani
- Department of Dermatology, Polytechnic University of Marche, Torrette, Ancona, Italy
| |
Collapse
|
23
|
Yang C, Cao M, Liu Y, He Y, Yang C, Du Y, Wang W, Zhang G, Wu M, Zhou M, Gao F. Inhibition of cell invasion and migration by CEACAM1-4S in breast cancer. Oncol Lett 2017; 14:4758-4766. [PMID: 29085477 PMCID: PMC5649695 DOI: 10.3892/ol.2017.6791] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 07/05/2017] [Indexed: 11/05/2022] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), a cell-cell adhesion molecule, has been revealed to perform an important role in tumor progression. Although there are a number of studies on CEACAM1 in patients with breast cancer, there is limited information on the roles of CEACAM1 in breast cancer metastasis. The present study aimed to identify whether CEACAM1 is involved in breast cancer development and to investigate the underlying mechanisms. First, the expression of CEACAM1 was observed in patients with breast cancer, and the association between CEACAM1 expression levels and migration and invasion of breast cancer cells was analyzed. As there are 12 isoforms of CEACAM1, of which CEACAM1-4S dominates in the human breast epithelium, subsequent study focused on CEACAM1-4S as a representative of all the isoforms. Results of the present study demonstrated that CEACAM1-4S suppresses breast cancer cell invasion and migration in a manner that is dependent on the balance between matrix metalloproteinase 2/tissue inhibitor of metalloproteinase 2 and E-/N-cadherin expression. In addition, CEACAM1-4S was likely to cause reversal of epithelial-mesenchymal transition of breast cancer cells through repressing Smad2 and signal transducer and phosphorylation of activator of transcription 3. In conclusion, the present study demonstrated that CEACAM1-4S performs an inhibitory role in breast cancer metastasis, and restoring CEACAM1-4S expression may provide a novel strategy for therapy of patients with metastatic breast cancer.
Collapse
Affiliation(s)
- Changcheng Yang
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Manlin Cao
- Department of Rehabilitation Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Yiwen Liu
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Yiqing He
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Cuixia Yang
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Yan Du
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Wenjuan Wang
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Guoliang Zhang
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Man Wu
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Muqing Zhou
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Feng Gao
- Department of Molecular Biology and Clinical Laboratory, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| |
Collapse
|
24
|
The Roles of Carcinoembryonic Antigen in Liver Metastasis and Therapeutic Approaches. Gastroenterol Res Pract 2017; 2017:7521987. [PMID: 28588612 PMCID: PMC5447280 DOI: 10.1155/2017/7521987] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 04/16/2017] [Indexed: 12/22/2022] Open
Abstract
Metastasis is a highly complicated and sequential process in which primary cancer spreads to secondary organic sites. Liver is a well-known metastatic organ from colorectal cancer. Carcinoembryonic antigen (CEA) is expressed in most gastrointestinal, breast, and lung cancer cells. Overexpression of CEA is closely associated with liver metastasis, which is the main cause of death from colorectal cancer. CEA is widely used as a diagnostic and prognostic tumor marker in cancer patients. It affects many steps of liver metastasis from colorectal cancer cells. CEA inhibits circulating cancer cell death. CEA also binds to heterogeneous nuclear RNA binding protein M4 (hnRNP M4), a Kupffer cell receptor protein, and activates Kupffer cells to secrete various cytokines that change the microenvironments for the survival of colorectal cancer cells in the liver. CEA also activates cell adhesion-related molecules. The close correlation between CEA and cancer has spurred the exploration of many CEA-targeted approaches as anticancer therapeutics. Understanding the detailed functions and mechanisms of CEA in liver metastasis will provide great opportunities for the improvement of anticancer approaches against colorectal cancers. In this report, the roles of CEA in liver metastasis and CEA-targeting anticancer modalities are reviewed.
Collapse
|
25
|
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
|
26
|
Jumper N, Hodgkinson T, Paus R, Bayat A. Site-specific gene expression profiling as a novel strategy for unravelling keloid disease pathobiology. PLoS One 2017; 12:e0172955. [PMID: 28257480 PMCID: PMC5336271 DOI: 10.1371/journal.pone.0172955] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 02/13/2017] [Indexed: 12/13/2022] Open
Abstract
Keloid disease (KD) is a fibroproliferative cutaneous tumour characterised by heterogeneity, excess collagen deposition and aggressive local invasion. Lack of a validated animal model and resistance to a multitude of current therapies has resulted in unsatisfactory clinical outcomes of KD management. In order to address KD from a new perspective, we applied for the first time a site-specific in situ microdissection and gene expression profiling approach, through combined laser capture microdissection and transcriptomic array. The aim here was to analyse the utility of this approach compared with established methods of investigation, including whole tissue biopsy and monolayer cell culture techniques. This study was designed to approach KD from a hypothesis-free and compartment-specific angle, using state-of-the-art microdissection and gene expression profiling technology. We sought to characterise expression differences between specific keloid lesional sites and elucidate potential contributions of significantly dysregulated genes to mechanisms underlying keloid pathobiology, thus informing future explorative research into KD. Here, we highlight the advantages of our in situ microdissection strategy in generating expression data with improved sensitivity and accuracy over traditional methods. This methodological approach supports an active role for the epidermis in the pathogenesis of KD through identification of genes and upstream regulators implicated in epithelial-mesenchymal transition, inflammation and immune modulation. We describe dermal expression patterns crucial to collagen deposition that are associated with TGFβ-mediated signalling, which have not previously been examined in KD. Additionally, this study supports the previously proposed presence of a cancer-like stem cell population in KD and explores the possible contribution of gene dysregulation to the resistance of KD to conventional therapy. Through this innovative in situ microdissection gene profiling approach, we provide better-defined gene signatures of distinct KD regions, thereby addressing KD heterogeneity, facilitating differential diagnosis with other cutaneous fibroses via transcriptional fingerprinting, and highlighting key areas for future KD research.
Collapse
Affiliation(s)
- N. Jumper
- Plastic and Reconstructive Surgery Research, University of Manchester, Oxford Rd, Manchester, United Kingdom
| | - T. Hodgkinson
- Plastic and Reconstructive Surgery Research, University of Manchester, Oxford Rd, Manchester, United Kingdom
- Centre for Tissue Injury and Repair, University of Manchester, and MAHSC, Manchester, United Kingdom
| | - R. Paus
- Centre for Dermatology Research, University of Manchester, and MAHSC, Manchester, United Kingdom
| | - A. Bayat
- Plastic and Reconstructive Surgery Research, University of Manchester, Oxford Rd, Manchester, United Kingdom
- Centre for Dermatology Research, University of Manchester, and MAHSC, Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
27
|
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
|
28
|
Mollah MMH, Jamal R, Mokhtar NM, Harun R, Mollah MNH. A Hybrid One-Way ANOVA Approach for the Robust and Efficient Estimation of Differential Gene Expression with Multiple Patterns. PLoS One 2015; 10:e0138810. [PMID: 26413858 PMCID: PMC4587675 DOI: 10.1371/journal.pone.0138810] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 09/03/2015] [Indexed: 11/22/2022] Open
Abstract
Background Identifying genes that are differentially expressed (DE) between two or more conditions with multiple patterns of expression is one of the primary objectives of gene expression data analysis. Several statistical approaches, including one-way analysis of variance (ANOVA), are used to identify DE genes. However, most of these methods provide misleading results for two or more conditions with multiple patterns of expression in the presence of outlying genes. In this paper, an attempt is made to develop a hybrid one-way ANOVA approach that unifies the robustness and efficiency of estimation using the minimum β-divergence method to overcome some problems that arise in the existing robust methods for both small- and large-sample cases with multiple patterns of expression. Results The proposed method relies on a β-weight function, which produces values between 0 and 1. The β-weight function with β = 0.2 is used as a measure of outlier detection. It assigns smaller weights (≥ 0) to outlying expressions and larger weights (≤ 1) to typical expressions. The distribution of the β-weights is used to calculate the cut-off point, which is compared to the observed β-weight of an expression to determine whether that gene expression is an outlier. This weight function plays a key role in unifying the robustness and efficiency of estimation in one-way ANOVA. Conclusion Analyses of simulated gene expression profiles revealed that all eight methods (ANOVA, SAM, LIMMA, EBarrays, eLNN, KW, robust BetaEB and proposed) perform almost identically for m = 2 conditions in the absence of outliers. However, the robust BetaEB method and the proposed method exhibited considerably better performance than the other six methods in the presence of outliers. In this case, the BetaEB method exhibited slightly better performance than the proposed method for the small-sample cases, but the the proposed method exhibited much better performance than the BetaEB method for both the small- and large-sample cases in the presence of more than 50% outlying genes. The proposed method also exhibited better performance than the other methods for m > 2 conditions with multiple patterns of expression, where the BetaEB was not extended for this condition. Therefore, the proposed approach would be more suitable and reliable on average for the identification of DE genes between two or more conditions with multiple patterns of expression.
Collapse
Affiliation(s)
- Mohammad Manir Hossain Mollah
- Institut Perubatan Molekul UKM (UMBI), University Kebangsaan Malaysia (UKM), Jalan Ya’acob Latiff, Bandar Tun Razak, Cheras 56000 Kuala Lumpur, Malaysia
- * E-mail:
| | - Rahman Jamal
- Institut Perubatan Molekul UKM (UMBI), University Kebangsaan Malaysia (UKM), Jalan Ya’acob Latiff, Bandar Tun Razak, Cheras 56000 Kuala Lumpur, Malaysia
| | - Norfilza Mohd Mokhtar
- Institut Perubatan Molekul UKM (UMBI), University Kebangsaan Malaysia (UKM), Jalan Ya’acob Latiff, Bandar Tun Razak, Cheras 56000 Kuala Lumpur, Malaysia
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Roslan Harun
- Institut Perubatan Molekul UKM (UMBI), University Kebangsaan Malaysia (UKM), Jalan Ya’acob Latiff, Bandar Tun Razak, Cheras 56000 Kuala Lumpur, Malaysia
| | - Md. Nurul Haque Mollah
- Laboratory of Bioinformatics, Department of Statistics, University of Rajshahi, Rajshahi-6205, Bangladesh
| |
Collapse
|
29
|
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]
|
30
|
Yamamoto N, Yokoyama S, Ieda J, Mitani Y, Yamaguchi S, Takifuji K, Hotta T, Matsuda K, Watanabe T, Shively JE, Yamaue H. CEACAM1 and hollow spheroid formation modulate the chemosensitivity of colorectal cancer to 5-fluorouracil. Cancer Chemother Pharmacol 2015; 75:421-430. [PMID: 25547406 DOI: 10.1007/s00280-014-2662-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Accepted: 12/22/2014] [Indexed: 01/18/2023]
Abstract
PURPOSE Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) re-expressed and promoted hollow spheroid (HS) formation beyond the invasion front of colorectal cancer. The aim of the present study was to clarify whether CEACAM1 cytoplasmic domain isoform balance and HS are associated with resistance to 5-fluorouracil (5FU). METHODS Two-dimensional (D) or 3D culture systems were employed to evaluate the effects of CEACAM1 cytoplasmic isoform balance and HS formation on the chemosensitivity of colorectal cancer cells to 5FU. The risk factors for postoperative recurrence were calculated based on the presence of HS and various clinicopathological characteristics in 82 patients with Stage III colorectal cancer who had undergone curative surgery followed by 5FU-based chemotherapy. RESULTS CEACAM1-4L-transfected HT29 and CEACAM1-4L and 4S expressing parental LS174T cells had significantly higher resistance to 5FU in comparison with CEACAM1-4S- or vector control-transfected cells. In 3D culture, HS formation induced by CEACAM1-4L induced chemoresistance to 5FU, whereas the solid spheres formed in response to CEACAM1-4S were destroyed by 5FU treatment. HS was identified as an independent factor for recurrence of Stage III colorectal cancer after curative resection followed by 5FU-based chemotherapy. Kaplan-Meier survival curves demonstrated that patients with HS had lower recurrence-free survival rate. CONCLUSIONS CEACAM1 long cytoplasmic domain isoform dominance and HS formation are phenotypes associated with chemoresistance to 5FU.
Collapse
Affiliation(s)
- Naoyuki Yamamoto
- Second Department of Surgery, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Liu JN, Wang HB, Zhou CC, Hu SY. CEACAM5 has different expression patterns in gastric non-neoplastic and neoplastic lesions and cytoplasmic staining is a marker for evaluation of tumor progression in gastric adenocarcinoma. Pathol Res Pract 2014; 210:686-93. [PMID: 25042385 DOI: 10.1016/j.prp.2014.06.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Revised: 05/01/2014] [Accepted: 06/19/2014] [Indexed: 01/08/2023]
Abstract
OBJECTIVE The aim of this study was to investigate the expression patterns of CEACAM5 in non-neoplastic and neoplastic gastric lesions, as well as its application in the differential diagnosis and its relationship with tumor progression. METHODS CEACAM5 expression was detected by immunohistochemical staining in the serial sections of the gastric neoplastic and non-neoplastic lesions. The impacts of CEACAM5 expression patterns on tumor progression were evaluated by statistics, the clinical and pathological data included sex, age, tumor extension, lymph node involvement and tumor staging. RESULTS There was no CEACAM5 expression in normal gastric epithelial cells. In hyperplastic polyps, CEACAM5 was expressed with apical membranous staining in the hyperplastic and prolonged gastric pit adjacent to the surface. Intestinal metaplasia (IM) expressed CEACAM5 mainly with membranous pattern, and some cases showed membranous staining mixed with cytoplasmic staining. GIN expressed CEACAM5 mainly with membranous staining, but the mixed staining of cytoplasmic and membranous patterns increased, and especially in the high grade GIN, cytoplasmic staining of CEACAM5 began to occur. Compared with IM and GIN, CEACAM5 expression patterns of hyperplastic polyp showed a significant difference (P=0.000). IM, low grade GIN and the whole GIN showed no significant difference in CEACAM5 expression patterns (P=0.355), but IM and high grade GIN showed a significant difference (P=0.027). There was a significant difference between low and high grade GIN (P=0.002). GIN and well-differentiated carcinomas showed no significant difference (P=0.070), but low grade GIN and well differentiated carcinomas showed a significant difference (P=0.006). In gastric adenocarcinomas, CEACAM5 expression patterns showed a significant difference in tumor grading (P=0.010) and Laurén classification (P=0.001). In histological grading, well differentiated carcinomas showed more membranous staining than moderately and poorly differentiated, and more cytoplasmic CEACAM5 staining was detected in moderately and poorly differentiated carcinomas. Similar to that, in Laurén classification, intestinal carcinomas showed more membranous staining, and diffuse carcinomas showed more cytoplasmic staining. Moreover, CEACAM5 expression patterns showed a significant difference in tumor extension (P=0.012), lymph node involvement (P=0.015) and tumor staging (P=0.002), suggesting that CEACAM5 should be involved in tumor progression. In advanced carcinomas, CEACAM5 was expressed with more cytoplasmic staining regardless of the histological classification. CONCLUSION CEACAM5 had different expression patterns in gastric non-neoplastic and neoplastic lesions. The CEACAM5 expression patterns were associated with tumor progression. Membranous staining of CEACAM5 might be a marker of premalignancy in gastric lesions, and cytoplasmic CEACAM5 might enhance tumor invasion and migration and be an evaluated marker for progressive and advanced gastric cancer. Also, it might be useful for the differential diagnosis of gastric premalignant lesions.
Collapse
Affiliation(s)
- Jia-Ning Liu
- Department of General Surgery, The Second Hospital, Shandong University, 247#, BeiYuan Street, Jinan, Shandong 250033, PR China
| | - Hong-Bo Wang
- Department of Digestive Disease, The Second Hospital, Shandong University, 247#, BeiYuan Street, Jinan, Shandong 250033, PR China
| | - Cheng-Cheng Zhou
- Department of Pathology, The Second Hospital, Shandong University, 247#, BeiYuan Street, Jinan, Shandong 250033, PR China
| | - San-Yuan Hu
- Qilu Hospital, Shandong University, 107#, Wenhua Xi Road, Jinan, Shandong 250012, PR China.
| |
Collapse
|
32
|
Smith DH, Christensen IJ, Jensen NF, Markussen B, Müller S, Nielsen HJ, Brünner N, Nielsen KV. An explorative analysis of ERCC1-19q13 copy number aberrations in a chemonaive stage III colorectal cancer cohort. BMC Cancer 2013; 13:489. [PMID: 24144331 PMCID: PMC4015772 DOI: 10.1186/1471-2407-13-489] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 10/08/2013] [Indexed: 11/10/2022] Open
Abstract
Background Platinum-based chemotherapy has long been used in the treatment of a variety of cancers and functions by inducing DNA damage. ERCC1 and ERCC4 are involved in the removal of this damage and have previously been implicated in resistance to platinum compounds. The aim of the current investigation is to determine the presence, frequency and prognostic impact of ERCC1 or ERCC4 gene copy number alterations in colorectal cancer (CRC). Methods Fluorescent in situ hybridization probes directed at ERCC1 and ERCC4 with relevant reference probes were constructed. Probes were tested in a CRC cell line panel and in tumor sections from 152 stage III CRC chemonaive patients. Relationships between biomarker status and clinical endpoints (overall survival, time to recurrence, and local recurrence in rectal cancer) were analyzed by survival statistics. Results ERCC1-19q13 copy number alterations were observed in a single cell line metaphase (HT29). In patient material, ERCC1-19q13 copy number gains (ERCC1-19q13/CEN-2 ≥ 1.5) were detected in 27.0% of specimens, whereas ERCC1-19q13 deletions (ERCC1-19q13/CEN-2 < 0.8) were only detected in 1.3%. ERCC1-19q13 gain was significantly associated with longer survival (multivariate analysis, HR: 0.45, 95% CI: 0.20-1.00, p = 0.049) in patients with colon tumors, but not rectal tumors. No ERCC4 aberrations were detected and scoring was discontinued after 50 patients. Conclusions ERCC1-19q13 copy number gains occur frequently in stage III CRC and influences survival in patients with colon tumors. Future studies will investigate the effect of ERCC1-19q13 aberrations in a platinum-treated patient population with the aim of developing a predictive biomarker profile for oxaliplatin sensitivity in CRC.
Collapse
|
33
|
Gene conversions are under purifying selection in the carcinoembryonic antigen immunoglobulin gene families of primates. Genomics 2013; 102:301-9. [DOI: 10.1016/j.ygeno.2013.07.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 06/25/2013] [Accepted: 07/08/2013] [Indexed: 11/20/2022]
|
34
|
Patel PC, Lee HSW, Ming AYK, Rath A, Deber CM, Yip CM, Rocheleau JV, Gray-Owen SD. Inside-out signaling promotes dynamic changes in the carcinoembryonic antigen-related cellular adhesion molecule 1 (CEACAM1) oligomeric state to control its cell adhesion properties. J Biol Chem 2013; 288:29654-69. [PMID: 24005674 DOI: 10.1074/jbc.m113.504639] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Cell-cell contacts are fundamental to multicellular organisms and are subject to exquisite levels of control. The carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) can engage in both cis-homophilic (parallel) oligomerization and trans-homophilic (anti-parallel) binding. In this study, we establish that the CEACAM1 transmembrane domain has a propensity to form cis-dimers via the transmembrane-embedded (432)GXXXG(436) motif and that this basal state is overcome when activated calmodulin binds to the CEACAM1 cytoplasmic domain. Although mutation of the (432)GXXXG(436) motif reduced CEACAM1 oligomerization, it did not affect surface localization of the receptor or influence CEACAM1-dependent cellular invasion by the pathogenic Neisseria. The mutation did, however, have a striking effect on CEACAM1-dependent cellular aggregation, increasing both the kinetics of cell-cell association and the size of cellular aggregates formed. CEACAM1 association with tyrosine kinase c-Src and tyrosine phosphatases SHP-1 and SHP-2 was not affected by the (432)GXXXG(436) mutation, consistent with their association with the monomeric form of wild type CEACAM1. Collectively, our results establish that a dynamic oligomer-to-monomer shift in surface-expressed CEACAM1 facilitates trans-homophilic binding and downstream effector signaling.
Collapse
|
35
|
Beauchemin N, Arabzadeh A. Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) in cancer progression and metastasis. Cancer Metastasis Rev 2013; 32:643-71. [DOI: 10.1007/s10555-013-9444-6] [Citation(s) in RCA: 288] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
36
|
MICHAELIDOU KLEITA, TZOVARAS ALEXANDROS, MISSITZIS IOANNIS, ARDAVANIS ALEXANDROS, SCORILAS ANDREAS. The expression of the CEACAM19 gene, a novel member of the CEA family, is associated with breast cancer progression. Int J Oncol 2013; 42:1770-7. [DOI: 10.3892/ijo.2013.1860] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 09/10/2012] [Indexed: 11/05/2022] Open
|
37
|
Neumaier C, Nittka S, Neumaier M. Loss of expression of the tumor suppressor CEACAM1 links different hereditary colorectal carcinoma subtypes to the genesis of sporadic colorectal carcinoma. ACTA ACUST UNITED AC 2012; 35:563-8. [PMID: 23038226 DOI: 10.1159/000342673] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
According to their carcinogenesis, colorectal cancer (CRC) subtypes show distinct molecular parameters. Hereditary non-polypous colorectal cancer (HNPCC) is the most common inherited CRC characterized by clinical criteria and confirmed microsatellite instability (MSI). Interestingly, a recently identified subtype, familial colorectal cancer type X (FCC-X), shows the same clinical criteria but microsatellite stability (MSS). CEACAM1 is a known tumor suppressor that regulates apoptosis in colon cells, and its loss is one of the most frequent events in early tumorigenesis of CRC. Therefore its loss may characterize precursor colon cells prior to neoplastic transformation. We analyzed tumor specimens of HNPCC and FCC-X patients in order to investigate whether there is a loss of CEACAM1 expression analogous to sporadic CRC and whether the expression of CEACAM1 would distinguish between these tumor entities. No differences in CEACAM1 expression were noted between HNPPC (n = 38) and FCC-X (n = 30) tumors. CEACAM1 was reduced in near-identical frequencies in 36/38 (95%) HNPCC and 29/30 (97%) FCC-X. This is the first report to demonstrate the loss of CEACAM1 expression in hereditary CRC. There was no difference between HNPCC and FCC-X. The frequency of expression loss was comparable to sporadic CRC, indicating that loss of CEACAM1 is an early event in colorectal tumorigenesis linking the genesis of sporadic and hereditary CRC.
Collapse
Affiliation(s)
- Christian Neumaier
- Clinic of Radiation Oncology, Universitätsmedizin Mannheim, University of Heidelberg, Mannheim, Germany
| | | | | |
Collapse
|
38
|
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
|
39
|
Lu R, Kujawski M, Pan H, Shively JE. Tumor angiogenesis mediated by myeloid cells is negatively regulated by CEACAM1. Cancer Res 2012; 72:2239-50. [PMID: 22406619 DOI: 10.1158/0008-5472.can-11-3016] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bv8 (prokineticin 2) expressed by Gr1(+)CD11b(+) myeloid cells is critical for VEGF-independent tumor angiogenesis. Although granulocyte colony-stimulating factor (G-CSF) has been shown to be a key inducer of Bv8 expression, the basis for Bv8 production in driving tumor angiogenesis is undefined. Because the cell adhesion molecule CEACAM1, which is highly expressed on Gr1(+)CD11b(+) myeloid cells, is known to regulate G-CSF receptor (G-CSFR) signaling, we hypothesized that CEACAM1 would regulate Bv8 production in these cells. In support of this hypothesis, we found that Bv8 expression was elevated in Gr1(+)CD11b(+) cells from Ceacam1-deficient mice implanted with B16 melanoma, increasing the infiltration of Gr1(+)CD11b(+) myeloid cells in melanoma tumors and enhancing their growth and angiogenesis. Furthermore, treatment with anti-Gr1 or anti-Bv8 or anti-G-CSF monoclonal antibody reduced myeloid cell infiltration, tumor growth, and angiogenesis to levels observed in tumor-bearing wild-type (WT) mice. Reconstitution of CEACAM1-deficient mice with WT bone marrow cells restored tumor infiltration of Gr1(+)CD11b(+) cells along with tumor growth and angiogenesis to WT levels. Treatment of tumor-bearing WT mice with anti-CEACAM1 antibody limited tumor outgrowth and angiogenesis, albeit to a lesser extent. Tumor growth in Ceacam1-deficient mice was not affected significantly in Rag(-/-) background, indicating that CEACAM1 expression in T and B lymphocytes had a negligible role in this pathway. Together, our findings show that CEACAM1 negatively regulates Gr1(+)CD11b(+) myeloid cell-dependent tumor angiogenesis by inhibiting the G-CSF-Bv8 signaling pathway.
Collapse
Affiliation(s)
- Rongze Lu
- City of Hope Irell & Manella Graduate School of Biological Sciences, Duarte, California, USA
| | | | | | | |
Collapse
|
40
|
Microarray Analyses of Genes Differentially Expressed by Diet (Black Beans and Soy Flour) during Azoxymethane-Induced Colon Carcinogenesis in Rats. J Nutr Metab 2012; 2012:351796. [PMID: 22496968 PMCID: PMC3306975 DOI: 10.1155/2012/351796] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Accepted: 10/25/2011] [Indexed: 02/06/2023] Open
Abstract
We previously demonstrated that black bean (BB) and soy flour (SF)-based diets inhibit azoxymethane (AOM)-induced colon cancer. The objective of this study was to identify genes altered by carcinogen treatment in normal-appearing colonic mucosa and those attenuated by bean feeding. Ninety-five male F344 rats were fed control (AIN) diets upon arrival. At 4 and 5 weeks, rats were injected with AOM (15 mg/kg) or saline and one week later administered an AIN, BB-, or SF-based diet. Rats were sacrificed after 31 weeks, and microarrays were conducted on RNA isolated from the distal colonic mucosa. AOM treatment induced a number of genes involved in immunity, including several MHC II-associated antigens and innate defense genes (RatNP-3, Lyz2, Pla2g2a). BB- and SF-fed rats exhibited a higher expression of genes involved in energy metabolism and water and sodium absorption and lower expression of innate (RatNP-3, Pla2g2a, Tlr4, Dmbt1) and cell cycle-associated (Cdc2, Ccnb1, Top2a) genes. Genes involved in the extracellular matrix (Col1a1, Fn1) and innate immunity (RatNP-3, Pla2g2a) were induced by AOM in all diets, but to a lower extent in bean-fed animals. This profile suggests beans inhibit colon carcinogenesis by modulating cellular kinetics and reducing inflammation, potentially by preserving mucosal barrier function.
Collapse
|
41
|
Hasselbalch HC, Skov V, Larsen TS, Thomassen M, Riley CH, Jensen MK, Bjerrum OW, Kruse TA. High expression of carcinoembryonic antigen-related cell adhesion molecule (CEACAM) 6 and 8 in primary myelofibrosis. Leuk Res 2011; 35:1330-4. [DOI: 10.1016/j.leukres.2011.03.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2011] [Revised: 03/07/2011] [Accepted: 03/08/2011] [Indexed: 12/20/2022]
|
42
|
Ieda J, Yokoyama S, Tamura K, Takifuji K, Hotta T, Matsuda K, Oku Y, Nasu T, Kiriyama S, Yamamoto N, Nakamura Y, Shively JE, Yamaue H. Re-expression of CEACAM1 long cytoplasmic domain isoform is associated with invasion and migration of colorectal cancer. Int J Cancer 2011; 129:1351-1361. [PMID: 21413011 DOI: 10.1002/ijc.26072] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 02/15/2011] [Indexed: 11/10/2022]
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is known to be downregulated at the transcriptional level in adenoma and carcinoma. Recent reports have shown that CEACAM1 is overexpressed at protein level in colorectal cancer and correlated with clinical stage. The reason why colorectal cancer cells re-expressed CEACAM1 remains unclear. The aim of our study was to clarify the implication of CEACAM1 re-expression in colorectal cancer. Immunohistochemical analyses were conducted with CEACAM1 long (CEACAM1-L) or short (CEACAM1-S) cytoplasmic domain-specific antibodies on clinical samples from 164 patients with colorectal cancer. The risk factors for metastasis and survival were calculated for clinical implication of CEACAM1 re-expression. Invasion chamber and wound healing assays were performed for the effect of CEACAM1 expression on invasion and migration of colorectal cancer cells. CEACAM1-L and CEACAM1-S stained with greater intensity at the invasion front than at the luminal surface of tumors. Differences between the long and short cytoplasmic isoform expression levels were observed at the invasion front. Multivariate analysis showed that CEACAM1-L dominance was an independent risk factor for lymph node metastasis, hematogenous metastasis and short survival. The Kaplan-Meier evaluation demonstrated that CEACAM1-L dominance was associated with shorter survival time (p < 0.0001). In the invasion chamber and wound healing assays, CEACAM1-L promoted invasion and migration. Re-expression of CEACAM1 is observed at the invasion front of colorectal cancer. CEACAM1-L dominance is associated with metastasis and shorter survival of the patients with colorectal cancer. CEACAM1-L dominance is important for colorectal cancer cells invasion and migration.
Collapse
Affiliation(s)
- Junji Ieda
- Second Department of Surgery, Wakayama Medical University School of Medicine, Wakayama, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Transcriptional changes in human Caco-2 colon cancer cells following exposure to a recurrent non-toxic dose of polyphenol-rich chokeberry juice. GENES AND NUTRITION 2011; 2:111-3. [PMID: 18850155 DOI: 10.1007/s12263-007-0026-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Berries and red fruits are important dietary sources of polyphenols [1]. In vitro and animal studies have demonstrated the bioavailability and the anti-proliferative and anticarcinogenic properties of these fruits or of their phenolic components [2, 3]. Consumption of berries may contribute to the reduction of colon cancer by mechanisms not yet understood. Gene expression analysis using microarrays allows for a more comprehensive study of the possible molecular mechanisms by which food or food components may prevent certain cancers of the gastrointestinal tract [4]. The aim of this research is to investigate the anti-proliferative effects of a polyphenol-rich berry juice on a human model of colon cancer cells and its association to transcriptional changes in relation to colon cancer.
Collapse
|
44
|
Lu SX, Kappel LW, Charbonneau-Allard AM, Atallah R, Holland AM, Turbide C, Hubbard VM, Rotolo JA, Smith M, Suh D, King C, Rao UK, Yim N, Bautista JL, Jenq RR, Penack O, Na IK, Liu C, Murphy G, Alpdogan O, Blumberg RS, Macian F, Holmes KV, Beauchemin N, van den Brink MRM. Ceacam1 separates graft-versus-host-disease from graft-versus-tumor activity after experimental allogeneic bone marrow transplantation. PLoS One 2011; 6:e21611. [PMID: 21760897 PMCID: PMC3130781 DOI: 10.1371/journal.pone.0021611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 06/02/2011] [Indexed: 11/19/2022] Open
Abstract
Background Allogeneic bone marrow transplantation (allo-BMT) is a potentially curative therapy for a variety of hematologic diseases, but benefits, including graft-versus-tumor (GVT) activity are limited by graft-versus-host-disease (GVHD). Carcinoembryonic antigen related cell adhesion molecule 1 (Ceacam1) is a transmembrane glycoprotein found on epithelium, T cells, and many tumors. It regulates a variety of physiologic and pathological processes such as tumor biology, leukocyte activation, and energy homeostasis. Previous studies suggest that Ceacam1 negatively regulates inflammation in inflammatory bowel disease models. Methods We studied Ceacam1 as a regulator of GVHD and GVT after allogeneic bone marrow transplantation (allo-BMT) in mouse models. In vivo, Ceacam1−/− T cells caused increased GVHD mortality and GVHD of the colon, and greater numbers of donor T cells were positive for activation markers (CD25hi, CD62Llo). Additionally, Ceacam1−/− CD8 T cells had greater expression of the gut-trafficking integrin α4β7, though both CD4 and CD8 T cells were found increased numbers in the gut post-transplant. Ceacam1−/− recipients also experienced increased GVHD mortality and GVHD of the colon, and alloreactive T cells displayed increased activation. Additionally, Ceacam1−/− mice had increased mortality and decreased numbers of regenerating small intestinal crypts upon radiation exposure. Conversely, Ceacam1-overexpressing T cells caused attenuated target-organ and systemic GVHD, which correlated with decreased donor T cell numbers in target tissues, and mortality. Finally, graft-versus-tumor survival in a Ceacam1+ lymphoma model was improved in animals receiving Ceacam1−/− vs. control T cells. Conclusions We conclude that Ceacam1 regulates T cell activation, GVHD target organ damage, and numbers of donor T cells in lymphoid organs and GVHD target tissues. In recipients of allo-BMT, Ceacam1 may also regulate tissue radiosensitivity. Because of its expression on both the donor graft and host tissues, this suggests that targeting Ceacam1 may represent a potent strategy for the regulation of GVHD and GVT after allogeneic transplantation.
Collapse
Affiliation(s)
- Sydney X Lu
- Department of Immunology and Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Dery KJ, Gaur S, Gencheva M, Yen Y, Shively JE, Gaur RK. Mechanistic control of carcinoembryonic antigen-related cell adhesion molecule-1 (CEACAM1) splice isoforms by the heterogeneous nuclear ribonuclear proteins hnRNP L, hnRNP A1, and hnRNP M. J Biol Chem 2011; 286:16039-51. [PMID: 21398516 PMCID: PMC3091213 DOI: 10.1074/jbc.m110.204057] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Revised: 02/14/2011] [Indexed: 11/06/2022] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule-1 (CEACAM1) is expressed in a variety of cell types and is implicated in carcinogenesis. Alternative splicing of CEACAM1 pre-mRNA generates two cytoplasmic domain splice variants characterized by the inclusion (L-isoform) or exclusion (S-isoform) of exon 7. Here we show that the alternative splicing of CEACAM1 pre-mRNA is regulated by novel cis elements residing in exon 7. We report the presence of three exon regulatory elements that lead to the inclusion or exclusion of exon 7 CEACAM1 mRNA in ZR75 breast cancer cells. Heterologous splicing reporter assays demonstrated that the maintenance of authentic alternative splicing mechanisms were independent of the CEACAM1 intron sequence context. We show that forced expression of these exon regulatory elements could alter CEACAM1 splicing in HEK-293 cells. Using RNA affinity chromatography, three members of the heterogeneous nuclear ribonucleoprotein family (hnRNP L, hnRNP A1, and hnRNP M) were identified. RNA immunoprecipitation of hnRNP L and hnRNP A1 revealed a binding motif located central and 3' to exon 7, respectively. Depletion of hnRNP A1 or L by RNAi in HEK-293 cells promoted exon 7 inclusion, whereas overexpression led to exclusion of the variable exon. By contrast, overexpression of hnRNP M showed exon 7 inclusion and production of CEACAM1-L mRNA. Finally, stress-induced cytoplasmic accumulation of hnRNP A1 in MDA-MB-468 cells dynamically alters the CEACAM1-S:CEACAM1:L ratio in favor of the l-isoform. Thus, we have elucidated the molecular factors that control the mechanism of splice-site recognition in the alternative splicing regulation of CEACAM1.
Collapse
Affiliation(s)
| | - Shikha Gaur
- Clinical and Molecular Pharmacology Beckman Research Institute of the City of Hope, Duarte, California 91010
| | | | - Yun Yen
- Clinical and Molecular Pharmacology Beckman Research Institute of the City of Hope, Duarte, California 91010
| | | | | |
Collapse
|
46
|
Nonaka M, Ma BY, Imaeda H, Kawabe K, Kawasaki N, Hodohara K, Kawasaki N, Andoh A, Fujiyama Y, Kawasaki T. Dendritic cell-specific intercellular adhesion molecule 3-grabbing non-integrin (DC-SIGN) recognizes a novel ligand, Mac-2-binding protein, characteristically expressed on human colorectal carcinomas. J Biol Chem 2011; 286:22403-13. [PMID: 21515679 DOI: 10.1074/jbc.m110.215301] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Dendritic cell (DC)-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN) is a type II transmembrane C-type lectin expressed on DCs such as myeloid DCs and monocyte-derived DCs (MoDCs). Recently, we have reported that DC-SIGN interacts with carcinoembryonic antigen (CEA) expressed on colorectal carcinoma cells. CEA is one of the most widely used tumor markers for gastrointestinal cancers such as colorectal cancer. On the other hand, other groups have reported that the level of Mac-2-binding protein (Mac-2BP) increases in patients with pancreatic, breast, and lung cancers, virus infections such as human immunodeficiency virus and hepatitis C virus, and autoimmune diseases. Here, we first identified Mac-2BP expressed on several colorectal carcinoma cell lines as a novel DC-SIGN ligand through affinity chromatography and mass spectrometry. Interestingly, we found that DC-SIGN selectively recognizes Mac-2BP derived from some colorectal carcinomas but not from the other ones. Furthermore, we found that the α1-3,4-fucose moieties of Le glycans expressed on DC-SIGN-binding Mac-2BP were important for recognition. DC-SIGN-dependent cellular interactions between immature MoDCs and colorectal carcinoma cells significantly inhibited MoDC functional maturation, suggesting that Mac-2BP may provide a tolerogenic microenvironment for colorectal carcinoma cells through DC-SIGN-dependent recognition. Importantly, Mac-2BP was detected as a predominant DC-SIGN ligand expressed on some primary colorectal cancer tissues from certain parts of patients in comparison with CEA from other parts, suggesting that DC-SIGN-binding Mac-2BP bearing tumor-associated Le glycans may become a novel potential colorectal cancer biomarker for some patients instead of CEA.
Collapse
Affiliation(s)
- Motohiro Nonaka
- Research Center for Glycobiotechnology, Ritsumeikan University, Shiga 525-0058, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Edvardsson K, Ström A, Jonsson P, Gustafsson JÅ, Williams C. Estrogen receptor β induces antiinflammatory and antitumorigenic networks in colon cancer cells. Mol Endocrinol 2011; 25:969-79. [PMID: 21493669 DOI: 10.1210/me.2010-0452] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Several studies suggest estrogen to be protective against the development of colon cancer. Estrogen receptor β (ERβ) is the predominant estrogen receptor expressed in colorectal epithelium and is the main candidate to mediate the protective effects. We have previously shown that expression of ERβ reduces growth of colorectal cancer in xenografts. Little is known of the actions of ERβ and its effect on gene transcription in colon cancers. To dissect the processes that ERβ mediates and to investigate cell-specific mechanisms, we reexpressed ERβ in three colorectal cancer cell lines (SW480, HT29, and HCT-116) and conducted genome-wide expression studies in combination with gene-pathway analyses and cross-correlation to ERβ-chromatin-binding sites. Although induced gene regulation was cell specific, overrepresentation analysis of functional classes indicated that the same biological themes, including apoptosis, cell differentiation, and regulation of the cell cycle, were affected in all three cell lines. Novel findings include a strong ERβ-mediated down-regulation of IL-6 and downstream networks with significant implications for inflammatory mechanisms involved in colon carcinogenesis. We also discovered cross talk between the suggested nuclear receptor coregulator PROX1 and ERβ, demonstrating that ERβ both regulates and shares target genes with PROX1. The influence of ERβ on apoptosis was further explored using functional studies, which suggested an increased DNA-repair capacity. We conclude that reexpression of ERβ induces transcriptome changes that, through several parallel pathways, converge into antitumorigenic capabilities in all three cell lines. We propose that enhancing ERβ action has potential as a novel therapeutic approach for prevention and/or treatment of colon cancer.
Collapse
Affiliation(s)
- Karin Edvardsson
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas 77204-5056, USA
| | | | | | | | | |
Collapse
|
48
|
Song JH, Cao Z, Yoon JH, Nam SW, Kim SY, Lee JY, Park WS. Genetic alterations and expression pattern of CEACAM1 in colorectal adenomas and cancers. Pathol Oncol Res 2011; 17:67-74. [PMID: 20524097 DOI: 10.1007/s12253-010-9282-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Accepted: 05/19/2010] [Indexed: 11/26/2022]
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is expressed on epithelial cells throughout the intestinal tract and is a negative regulator of tumor cell growth, suggesting that it may function as a tumor suppressor. In this study, to determine whether the CEACAM1 is involved in colorectal tumorigenesis, we have investigated the genetic alterations, including mutations and allelic loss, of the CEACAM1 gene in 17 colonic adenomas and 123 sporadic colorectal cancers. In addition, the expression pattern of the CEACAM1 protein was examined in 60 colonic adenomas and 123 sporadic colorectal adenocarcinomas. No mutation was found in colonic adenomas, but four somatic missense mutations, L36F, T312I, V398I and A445V, were detected in colorectal cancers. Interestingly, all of the mutations were found in left-side colon cancers of the patients with clinical stage III. In LOH analysis, nine adenomas were informative for at least one of the markers and five (55.6%) showed allelic loss. Thirty-eight cancers were informative at D19S211 and D19S872 markers and 21 (56.3%) showed LOH at these markers. Statistically, the frequency of allelic loss at the CEACAM1 locus was not associated with clinicopathologic parameters (P > 0.05). In immunohistochemical analysis, loss of expression of CEACAM1 protein was detected in nine (15.0%) and 30 (24.4%) of 60 colorectal adenomas and 123 colorectal cancers. Statistically, there was no significant relationship between loss of CEACAM1 expression and clinicopathologic parameters, including clinical stage, tumor location, tumor size, lymph node metastasis and 5-year survival (P > 0.05). These data suggest that genetic alteration and loss of expression of the CEACAM1 may contribute to the development of colorectal cancers, as an early event.
Collapse
Affiliation(s)
- Jae Hwi Song
- Department of Pathology, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul 137-701, Korea
| | | | | | | | | | | | | |
Collapse
|
49
|
Musteanu M, Blaas L, Mair M, Schlederer M, Bilban M, Tauber S, Esterbauer H, Mueller M, Casanova E, Kenner L, Poli V, Eferl R. Stat3 is a negative regulator of intestinal tumor progression in Apc(Min) mice. Gastroenterology 2010; 138:1003-11.e1-5. [PMID: 19962983 DOI: 10.1053/j.gastro.2009.11.049] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2009] [Revised: 10/16/2009] [Accepted: 11/19/2009] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS The transcription factor signal transducer and activator of transcription 3 (Stat3) has been considered to promote progression and metastasis of intestinal cancers. METHODS We investigated the role of Stat3 in intestinal tumors using mice with conditional ablation of Stat3 in intestinal epithelial cells (Stat3(DeltaIEC)). RESULTS In the Apc(Min) mouse model of intestinal cancer, genetic ablation of Stat3 reduced the multiplicity of early adenomas. However, loss of Stat3 promoted tumor progression at later stages, leading to formation of invasive carcinomas, which significantly shortened the lifespan of Stat3(DeltaIEC)Apc(Min/+) mice. Interestingly, loss of Stat3 in tumors of Apc(Min/+) mice had no significant impact on cell survival and angiogenesis, but promoted cell proliferation. A genome-wide expression analysis of Stat3-deficient tumors suggested that Stat3 might negatively regulate intestinal cancer progression via the cell adhesion molecule CEACAM1. CONCLUSIONS Our data suggest that Stat3 impairs invasiveness of intestinal tumors. Therefore, therapeutic targeting of the Stat3 signaling pathway in intestinal cancer should be evaluated for adverse effects on tumor progression.
Collapse
Affiliation(s)
- Monica Musteanu
- Ludwig Boltzmann Institute for Cancer Research, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Kammerer R, Zimmermann W. Coevolution of activating and inhibitory receptors within mammalian carcinoembryonic antigen families. BMC Biol 2010; 8:12. [PMID: 20132533 PMCID: PMC2832619 DOI: 10.1186/1741-7007-8-12] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Accepted: 02/04/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Most rapidly evolving gene families are involved in immune responses and reproduction, two biological functions which have been assigned to the carcinoembryonic antigen (CEA) gene family. To gain insights into evolutionary forces shaping the CEA gene family we have analysed this gene family in 27 mammalian species including monotreme and marsupial lineages. RESULTS Phylogenetic analysis provided convincing evidence that the primordial CEA gene family in mammals consisted of five genes, including the immune inhibitory receptor-encoding CEACAM1 (CEA-related cell adhesion molecule) ancestor. Our analysis of the substitution rates within the nucleotide sequence which codes for the ligand binding domain of CEACAM1 indicates that the selection for diversification is, perhaps, a consequence of the exploitation of CEACAM1 by a variety of viral and bacterial pathogens as their cellular receptor. Depending on the extent of the amplification of an ancestral CEACAM1, the number of CEACAM1-related genes varies considerably between mammalian species from less than five in lagomorphs to more than 100 in bats. In most analysed species, ITAM (immunoreceptor tyrosine-based activation motifs) or ITAM-like motif-containing proteins exist which contain Ig-V-like, ligand binding domains closely related to that of CEACAM1. Human CEACAM3 is one such protein which can function as a CEACAM1 decoy receptor in granulocytes by mediating the uptake and destruction of specific bacterial pathogens via its ITAM-like motif. The close relationship between CEACAM1 and its ITAM-encoding relatives appears to be maintained by gene conversion and reciprocal recombination. Surprisingly, secreted CEACAMs resembling immunomodulatory CEACAM1-related trophoblast-specific pregnancy-specific glycoproteins (PSGs) found in humans and rodents evolved only in a limited set of mammals. The appearance of PSG-like genes correlates with invasive trophoblast growth in these species. CONCLUSIONS These phylogenetic studies provide evidence that pathogen/host coevolution and a possible participation in fetal-maternal conflict processes led to a highly species-specific diversity of mammalian CEA gene families.
Collapse
Affiliation(s)
- Robert Kammerer
- Tumor Immunology Laboratory, LIFE Center, Klinikum Grosshadern, Ludwig-Maximilians-University, Marchioninistrasse 23, 81377 Munich, Germany.
| | | |
Collapse
|