1
|
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
|
2
|
Walker E, van Niekerk S, Hanning K, Kelton W, Hicks J. Mechanisms of host manipulation by Neisseria gonorrhoeae. Front Microbiol 2023; 14:1119834. [PMID: 36819065 PMCID: PMC9935845 DOI: 10.3389/fmicb.2023.1119834] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
Neisseria gonorrhoeae (also known as gonococcus) has been causing gonorrhoea in humans since ancient Egyptian times. Today, global gonorrhoea infections are rising at an alarming rate, in concert with an increasing number of antimicrobial-resistant strains. The gonococcus has concurrently evolved several intricate mechanisms that promote pathogenesis by evading both host immunity and defeating common therapeutic interventions. Central to these adaptations is the ability of the gonococcus to manipulate various host microenvironments upon infection. For example, the gonococcus can survive within neutrophils through direct regulation of both the oxidative burst response and maturation of the phagosome; a concerning trait given the important role neutrophils have in defending against invading pathogens. Hence, a detailed understanding of how N. gonorrhoeae exploits the human host to establish and maintain infection is crucial for combating this pathogen. This review summarizes the mechanisms behind host manipulation, with a central focus on the exploitation of host epithelial cell signaling to promote colonization and invasion of the epithelial lining, the modulation of the host immune response to evade both innate and adaptive defenses, and the manipulation of host cell death pathways to both assist colonization and combat antimicrobial activities of innate immune cells. Collectively, these pathways act in concert to enable N. gonorrhoeae to colonize and invade a wide array of host tissues, both establishing and disseminating gonococcal infection.
Collapse
Affiliation(s)
- Emma Walker
- Te Huataki Waiora, School of Health, University of Waikato, Hamilton, New Zealand
| | - Stacy van Niekerk
- Te Huataki Waiora, School of Health, University of Waikato, Hamilton, New Zealand
| | - Kyrin Hanning
- Te Huataki Waiora, School of Health, University of Waikato, Hamilton, New Zealand
| | - William Kelton
- Te Huataki Waiora, School of Health, University of Waikato, Hamilton, New Zealand
- Te Aka Mātuatua School of Science, University of Waikato, Hamilton, New Zealand
| | - Joanna Hicks
- Te Huataki Waiora, School of Health, University of Waikato, Hamilton, New Zealand
| |
Collapse
|
3
|
Liu D, Wu C, Wang J, Zhang L, Sun Z, Chen S, Ding Y, Wang W. Transfer RNA-derived fragment 5'tRF-Gly promotes the development of hepatocellular carcinoma by direct targeting of carcinoembryonic antigen-related cell adhesion molecule 1. Cancer Sci 2022; 113:3476-3488. [PMID: 35879647 PMCID: PMC9530880 DOI: 10.1111/cas.15505] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 11/30/2022] Open
Abstract
Transfer RNA-derived fragments are a group of small noncoding single-stranded RNA that play essential roles in multiple diseases. However, their biological functions in carcinogenesis are not well understood. In this study, 5'tRF-Gly was found to have significantly high expression in hepatocellular carcinoma (HCC), and the upregulation of 5'tRF-Gly was positively correlated with tumor size and tumor metastasis. Overexpression of 5'tRF-Gly induced increased growth rate and metastasis in HCC cells in vitro and in nude mice, while knockdown showed the opposite effect. Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) was confirmed to be a direct target of 5'tRF-Gly in HCC. In addition, the cytological effect of CEACAM1 knockdown proved to be similar to the overexpression of 5'tRF-Gly. Moreover, attenuation of CEACAM1 expression rescued the 5'tRF-Gly-mediated promoting effects on HCC cells. These data show that 5'tRF-Gly is a new tumor-promoting factor and could be a potential diagnostic biomarker or new therapeutic target for HCC.
Collapse
Affiliation(s)
- Dekai Liu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhouChina
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang ProvinceHangzhouChina
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang UniversityHangzhouChina
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang ProvinceHangzhouChina
- Cancer CenterZhejiang UniversityHangzhouChina
| | - Chengdong Wu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhouChina
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang ProvinceHangzhouChina
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang UniversityHangzhouChina
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang ProvinceHangzhouChina
- Cancer CenterZhejiang UniversityHangzhouChina
| | - Jingjie Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhouChina
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang ProvinceHangzhouChina
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang UniversityHangzhouChina
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang ProvinceHangzhouChina
- Cancer CenterZhejiang UniversityHangzhouChina
| | - Lufei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhouChina
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang ProvinceHangzhouChina
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang UniversityHangzhouChina
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang ProvinceHangzhouChina
- Cancer CenterZhejiang UniversityHangzhouChina
| | - Zhongquan Sun
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhouChina
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang ProvinceHangzhouChina
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang UniversityHangzhouChina
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang ProvinceHangzhouChina
- Cancer CenterZhejiang UniversityHangzhouChina
| | - Shihong Chen
- Department of Clinical MedicineWenzhou Medical CollegeWenzhouChina
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhouChina
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang ProvinceHangzhouChina
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang UniversityHangzhouChina
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang ProvinceHangzhouChina
- Cancer CenterZhejiang UniversityHangzhouChina
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhouChina
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang ProvinceHangzhouChina
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang UniversityHangzhouChina
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang ProvinceHangzhouChina
- Cancer CenterZhejiang UniversityHangzhouChina
| |
Collapse
|
4
|
Hu W, Bagramyan K, Bhatticharya S, Hong T, Tapia A, Wong P, Kalkum M, Shively JE. Phosphorylation of human CEACAM1-LF by PKA and GSK3β promotes its interaction with β-catenin. J Biol Chem 2021; 297:101305. [PMID: 34656562 PMCID: PMC8564729 DOI: 10.1016/j.jbc.2021.101305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 10/05/2021] [Accepted: 10/12/2021] [Indexed: 12/26/2022] Open
Abstract
CEACAM1-LF, a homotypic cell adhesion adhesion molecule, transduces intracellular signals via a 72 amino acid cytoplasmic domain that contains two immunoreceptor tyrosine-based inhibitory motifs (ITIMs) and a binding site for β-catenin. Phosphorylation of Ser503 by PKC in rodent CEACAM1 was shown to affect bile acid transport or hepatosteatosis via the level of ITIM phosphorylation, but the phosphorylation of the equivalent residue in human CEACAM1 (Ser508) was unclear. Here we studied this analogous phosphorylation by NMR analysis of the 15N labeled cytoplasmic domain peptide. Incubation with a variety of Ser/Thr kinases revealed phosphorylation of Ser508 by GSK3bβ but not by PKC. The lack of phosphorylation by PKC is likely due to evolutionary sequence changes between the rodent and human genes. Phosphorylation site assignment by mass spectrometry and NMR revealed phosphorylation of Ser472, Ser461 and Ser512 by PKA, of which Ser512 is part of a conserved consensus site for GSK3β binding. We showed here that only after phosphorylation of Ser512 by PKA was GSK3β able to phosphorylate Ser508. Phosphorylation of Ser512 by PKA promoted a tight association with the armadillo repeat domain of β-catenin at an extended region spanning the ITIMs of CEACAM1. The kinetics of phosphorylation of the ITIMs by Src, as well dephosphorylation by SHP2, were affected by the presence of Ser508/512 phosphorylation, suggesting that PKA and GSK3β may regulate the signal transduction activity of human CEACAM1-LF. The interaction of CEACAM1-LF with β-catenin promoted by PKA is suggestive of a tight association between the two ITIMs of CEACAM1-LF.
Collapse
Affiliation(s)
- Weidong Hu
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Karine Bagramyan
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Supriyo Bhatticharya
- Department of Computational and Quantitative Medicine, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Teresa Hong
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Alonso Tapia
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Patty Wong
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Markus Kalkum
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - John E Shively
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, California, USA.
| |
Collapse
|
5
|
Allen SD, Liu X, Jiang J, Liao YP, Chang CH, Nel AE, Meng H. Immune checkpoint inhibition in syngeneic mouse cancer models by a silicasome nanocarrier delivering a GSK3 inhibitor. Biomaterials 2020; 269:120635. [PMID: 33422940 DOI: 10.1016/j.biomaterials.2020.120635] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 12/21/2020] [Accepted: 12/27/2020] [Indexed: 12/18/2022]
Abstract
Checkpoint blocking antibodies that interfere in the PD-1/PD-L1 axis provide effective cancer immunotherapy for tumors that are immune inflamed or induced to become "hot". It has also been demonstrated that a small molecule inhibitor of the signaling hub kinase GSK3 can interfere in the PD-1/PD-L1 axis in T-cells by suppressing PD-1 expression. This provides an alternative approach to intervening in the PD-1/PD-L1 axis to provide cancer immunotherapy. In this communication, we demonstrate the remote loading of GSK3 inhibitor AZD1080 into the porous interior of mesoporous silica nanoparticles coated with a lipid bilayer (a.k.a. silicasomes). In a MC38 colon cancer model, intravenous injection (IV) of silicasome-encapsulated AZD1080 significantly improved biodistribution and drug delivery to the tumor site. The improved drug delivery was accompanied by cytotoxic MC38 tumor cell killing by perforin-releasing CD8+ T-cells, exhibiting reduced PD-1 expression. IV injection of encapsulated AZD1080 also resulted in significant tumor shrinkage in other syngeneic mouse tumor models, including another colorectal tumor (CT26), as well as pancreas (KPC) and lung (LLC) cancer models. Not only was the therapeutic efficacy of encapsulated AZD1080 similar or better than anti-PD-1 antibody, but the treatment was devoid of treatment toxicity. These results provide proof-of-principal demonstration of the feasibility of using encapsulated delivery of a GSK3 inhibitor to provide cancer immunotherapy, with the possibility to be used as a monotherapy or in combination with chemotherapy or other immunomodulatory agents.
Collapse
Affiliation(s)
- Sean D Allen
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA
| | - Xiangsheng Liu
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA; California NanoSystems Institute, University of California, Los Angeles, CA, USA
| | - Jinhong Jiang
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA
| | - Yu-Pei Liao
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA
| | - Chong Hyun Chang
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA
| | - Andre E Nel
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA; California NanoSystems Institute, University of California, Los Angeles, CA, USA.
| | - Huan Meng
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA; California NanoSystems Institute, University of California, Los Angeles, CA, USA.
| |
Collapse
|
6
|
Muenzner P, Hauck CR. Neisseria gonorrhoeae Blocks Epithelial Exfoliation by Nitric-Oxide-Mediated Metabolic Cross Talk to Promote Colonization in Mice. Cell Host Microbe 2020; 27:793-808.e5. [PMID: 32289262 DOI: 10.1016/j.chom.2020.03.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/19/2019] [Accepted: 03/17/2020] [Indexed: 12/18/2022]
Abstract
Several pathogens suppress exfoliation, a key defense of epithelia against microbial colonization. Common among these pathogens, exemplified by Neisseria gonorrhoeae, is their ability to bind carcinoembryonic antigen-related cell adhesion molecules (CEACAMs). Gonococcal CEACAM engagement triggers the expression of CD105, which is necessary to block epithelial exfoliation, whereas homotypic CEACAM-CEACAM interactions or antibody-mediated CEACAM clustering does not lead to CD105 expression. Here, we show that CEACAM-associated bacteria release nitric oxide (NO) during anaerobic respiration, and membrane-permeable NO initiates a eukaryotic signaling pathway involving soluble guanylate cyclase (sGC), protein kinase G, and the transcription factor CREB to upregulate CD105 expression. A murine vaginal infection model with N. gonorrhoeae reveals this metabolic cross communication allows bacterial suppression of epithelial exfoliation to facilitate mucosal colonization. Disrupting NO-initiated responses in host cells re-establishes epithelial exfoliation and inhibits mouse genital tract colonization by N. gonorrhoeae, suggesting a host-directed approach to prevent bacterial infections.
Collapse
Affiliation(s)
- Petra Muenzner
- Lehrstuhl Für Zellbiologie, Fachbereich Biologie, Universität Konstanz, 78457 Konstanz, Germany
| | - Christof R Hauck
- Lehrstuhl Für Zellbiologie, Fachbereich Biologie, Universität Konstanz, 78457 Konstanz, Germany; Konstanz Research School Chemical Biology, Universität Konstanz, 78457 Konstanz, Germany.
| |
Collapse
|
7
|
Blache P, Canterel-Thouennon L, Busson M, Verdié P, Subra G, Ychou M, Prévostel C. A Short SOX9 Peptide Mimics SOX9 Tumor Suppressor Activity and Is Sufficient to Inhibit Colon Cancer Cell Growth. Mol Cancer Ther 2019; 18:1386-1395. [DOI: 10.1158/1535-7163.mct-18-1149] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 02/14/2019] [Accepted: 05/09/2019] [Indexed: 11/16/2022]
|
8
|
Jia L, Lv D, Zhang S, Wang Z, Zhou B. Astragaloside IV Inhibits the Progression of Non-Small Cell Lung Cancer Through the Akt/GSK-3β/β-Catenin Pathway. Oncol Res 2018; 27:503-508. [PMID: 30131090 PMCID: PMC7848426 DOI: 10.3727/096504018x15344989701565] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Astragaloside IV (AS-IV) is an active ingredient in Astragalus membranaceus and is involved in various biological processes, such as regulating the immune system, and counteracting inflammation and malignancy. The aim of this study was to explore the effect of AS-IV on non-small cell lung cancer (NSCLC) cells. Cell counting kit (CCK)-8 assay and flow cytometry were performed to investigate cell survival and cell death, and Western blotting was performed to assess protein expression. We found that AS-IV inhibited the migration and proliferation of NSCLC cells and caused a noticeable increase in cell death. Furthermore, the expression of Bax, a marker of cell death, was increased, whereas the expression of Bcl-2, an antiapoptotic protein, was reduced. AS-IV also promoted cleavage of caspase-3, another indication of apoptosis. Finally, the Akt/GSK-3β/β-catenin axis was suppressed in response to AS-IV. Taken together, these findings provide evidence that AS-IV inhibits NSCLC development via inhibition of the Akt/GSK-3β/β-catenin signaling axis. We therefore propose that AS-IV represents a promising novel agent for the treatment of NSCLC.
Collapse
Affiliation(s)
- Liwei Jia
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, P.R. China
| | - Dongying Lv
- Heilongjiang Environmental Monitoring Central Station, Harbin, Heilongjiang Province, P.R. China
| | - Shuang Zhang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, P.R. China
| | - Zhenyue Wang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, P.R. China
| | - Bo Zhou
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, P.R. China
| |
Collapse
|
9
|
Prognostic Impact of CEACAM1 in Node-Negative Ovarian Cancer Patients. DISEASE MARKERS 2018; 2018:6714287. [PMID: 30050594 PMCID: PMC6046165 DOI: 10.1155/2018/6714287] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 05/31/2018] [Indexed: 01/01/2023]
Abstract
The underlying mechanisms of ovarian cancer (OvCa) dissemination are still poorly understood, and novel molecular markers for this cancer type are urgently needed. In search of adhesion molecules with prognostic relevance in OvCa, we compared tumors with good outcome (alive > 3 years) and those with poor outcome (dead < 2 years) within data from The Cancer Genome Atlas (TCGA). The carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) turned out as the only gene with differential expression in these groups. In order to further investigation on its role in OvCa, we analyzed CEACAM1 mRNA levels extracted from TCGA microarray data (n = 517) as well as CEACAM1 protein expression by Western blot analysis in a cohort of 242 tumor samples. Further, CEACAM1 localization in tumour tissue was evaluated by immunohistochemistry and CEACAM1 splice variants by RT-PCR in representative tumours. In Kaplan–Meier analysis, high CEACAM1 mRNA levels significantly correlated with longer survival (p = 0.008). By Western blot analysis in the second cohort, similar associations of high CEACAM1 protein levels with longer recurrence-free survival (RFS, p = 0.035) and overall survival (OAS, p = 0.004) were observed. In multivariate Cox regression analysis including clinical prognostic parameters, CEACAM1 mRNA or protein expression turned out as independent prognostic markers. Stratified survival analysis showed that high CEACAM1 protein expression was prognostic in node-negative tumors (p = 0.045 and p = 0.0002 for DFS and OAS) but lost prognostic significance in node-positive carcinomas. Similarly, high CEACAM1 mRNA expression did not show prognostic relevance in tumors with lymphatic invasion (L1) but was associated with longer survival in cases without lymphovascular involvement. Further analysis showed a predominance of 4S and 4L isoforms and mostly membraneous CEACAM1 localization in ovarian tumours. Our results suggest that CEACAM1 might be an independent favorable prognostic marker in OvCa, especially in the subgroup of patients with solely intraperitoneal metastasis.
Collapse
|
10
|
Prévostel C, Rammah-Bouazza C, Trauchessec H, Canterel-Thouennon L, Busson M, Ychou M, Blache P. SOX9 is an atypical intestinal tumor suppressor controlling the oncogenic Wnt/ß-catenin signaling. Oncotarget 2018; 7:82228-82243. [PMID: 27429045 PMCID: PMC5347687 DOI: 10.18632/oncotarget.10573] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 06/26/2016] [Indexed: 01/10/2023] Open
Abstract
SOX9 inactivation is frequent in colorectal cancer (CRC) due to SOX9 gene mutations and/or to ectopic expression of MiniSOX9, a dominant negative inhibitor of SOX9. In the present study, we report a heterozygous L142P inactivating mutation of SOX9 in the DLD-1 CRC cell line and we demonstrate that the conditional expression of a wild type SOX9 in this cell line inhibits cell growth, clonal capacity and colonosphere formation while decreasing both the activity of the oncogenic Wnt/ß-catenin signaling pathway and the expression of the c-myc oncogene. This activity does not require SOX9 transcriptional function but, rather, involves an interaction of SOX9 with nuclear ß-catenin. Furthermore, we report that SOX9 inhibits tumor development when conditionally expressed in CRC cells injected either subcutaneous or intraperitoneous in BALB/c mice as an abdominal metastasis model. These observations argue in favor of a tumor suppressor activity for SOX9. As an siRNA targeting SOX9 paradoxically also inhibits DLD-1 and HCT116 CRC cell growth, we conclude that there is a critical level of endogenous active SOX9 needed to maintain CRC cell growth.
Collapse
Affiliation(s)
- Corinne Prévostel
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM, U1194, Montpellier, France.,Université de Montpellier, Montpellier, France.,Institut régional du Cancer de Montpellier, Montpellier, France
| | - Cyrine Rammah-Bouazza
- Université de Montpellier, UMR 5237, Centre de Recherche de Biochimie Macromoléculaire, CNRS, Montpellier, France
| | - Hélène Trauchessec
- Université de Montpellier, UMR 5237, Centre de Recherche de Biochimie Macromoléculaire, CNRS, Montpellier, France
| | - Lucile Canterel-Thouennon
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM, U1194, Montpellier, France.,Université de Montpellier, Montpellier, France.,Institut régional du Cancer de Montpellier, Montpellier, France
| | - Muriel Busson
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM, U1194, Montpellier, France.,Université de Montpellier, Montpellier, France.,Institut régional du Cancer de Montpellier, Montpellier, France
| | - Marc Ychou
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM, U1194, Montpellier, France.,Université de Montpellier, Montpellier, France.,Institut régional du Cancer de Montpellier, Montpellier, France.,Centre Hospitalier Régional Universitaire (CHU) de Montpellier, Montpellier, France.,Institut Régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
| | - Philippe Blache
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM, U1194, Montpellier, France.,Université de Montpellier, Montpellier, France.,Institut régional du Cancer de Montpellier, Montpellier, France
| |
Collapse
|
11
|
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
|
12
|
Zhang Y, Cai P, Li L, Shi L, Chang P, Liang T, Yang Q, Liu Y, Wang L, Hu L. Co-expression of TIM-3 and CEACAM1 promotes T cell exhaustion in colorectal cancer patients. Int Immunopharmacol 2016; 43:210-218. [PMID: 28038383 DOI: 10.1016/j.intimp.2016.12.024] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 12/19/2016] [Accepted: 12/19/2016] [Indexed: 12/12/2022]
Abstract
T-cell immunoglobulin domain and mucin domain-3(TIM-3) is an activation induced inhibitory molecule involved in immune tolerance and is recently reported to induce T cell exhaustion which is mediated by carcinoembryonic antigen cell adhesion molecule 1(CEACAM1), another well-known molecule expressed on activated T cells and involved in T cell inhibition. To investigate the expression of TIM-3 and CEACAM1 on circulating CD8+ T cells and tumor infiltrating lymphocytes (TILs), 65 diagnosed colorectal cancer (CRC) patients and 38 healthy controls were enrolled in this study and the results showed that TIM-3 and CEACAM1 were both highly expressed on circulating CD8+ T cells in CRC patients and elevated on TILs compared with paraneoplastic T cells. Furthermore, TIM-3+CEACAM1+ CD8+ T cells represented the most dysfunctional population with the least IFN-γ production. In addition, the expressions of TIM-3 and CEACAM1 were correlated with advanced stage and could be independent risk factors for CRC. We for the first time to our knowledge suggested that co-expression of TIM-3 and CEACAM1 can mediate T cell exhaustion and may be potential biomarkers for CRC prediction, highlighting the possibility of being immunotherapy targets.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Pengcheng Cai
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lei Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Liang Shi
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Panpan Chang
- Central Medical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tao Liang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qianqian Yang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yang Liu
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lin Wang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Lihua Hu
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
13
|
Rueckschloss U, Kuerten S, Ergün S. The role of CEA-related cell adhesion molecule-1 (CEACAM1) in vascular homeostasis. Histochem Cell Biol 2016; 146:657-671. [PMID: 27695943 DOI: 10.1007/s00418-016-1505-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2016] [Indexed: 12/11/2022]
Abstract
Carcinoembryonic antigen (CEA)-related cell adhesion molecules belong to the immunoglobulin superfamily, are expressed in a broad spectrum of tissues and cell types and exert context-dependent activating as well as inhibitory effects. Among these molecules, the CEA-related cell adhesion molecule-1 (CEACAM1) is a transmembrane molecule with an extracellular, a transmembrane and a cytoplasmic domain. The latter contains immunoreceptor tyrosine-based inhibitory motifs and functions as a signaling molecule. CEACAM1 can form homo- and heterodimers which is relevant for its signaling activities. CEACAM1 acts as co-receptor that modulates the activity of different receptor types including VEGFR-2, and B and T cell receptors. CEACAM1 is expressed in endothelial cells, in pericytes of developing and newly formed immature blood vessels and in angiogenically activated adult vessels, e.g., tumor blood vessels. However, it is either undetectable or only weakly expressed in quiescent blood vessels. Recent studies indicated that CEACAM1 is involved in the regulation of the endothelial barrier function. In CEACAM1 -/- mice, increased vascular permeability and development of small atherosclerotic lesions was observed in the aortae. CEACAM1 is also detectable in activated lymphatic endothelial cells and plays a role in tumor lymphangiogenesis. This review summarizes the vascular effects of CEACAM1 and focuses on its role in vascular morphogenesis and endothelial barrier regulation.
Collapse
Affiliation(s)
- Uwe Rueckschloss
- Institute of Anatomy and Cell Biology, University of Würzburg, Köllikerstrasse 6, 97070, Würzburg, Germany
| | - Stefanie Kuerten
- Institute of Anatomy and Cell Biology, University of Würzburg, Köllikerstrasse 6, 97070, Würzburg, Germany
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, University of Würzburg, Köllikerstrasse 6, 97070, Würzburg, Germany.
| |
Collapse
|
14
|
Florian W, Lenfert E, Gerstel D, von Ehrenstein L, Einhoff J, Schmidt G, Logsdon M, Brandner J, Tiegs G, Beauchemin N, Wagener C, Deppert W, Horst AK. CEACAM1 controls the EMT switch in murine mammary carcinoma in vitro and in vivo. Oncotarget 2016; 7:63730-63746. [PMID: 27572314 PMCID: PMC5325399 DOI: 10.18632/oncotarget.11650] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 08/08/2016] [Indexed: 12/29/2022] Open
Abstract
We analyzed the molecular basis for carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1)-controlled inhibition of epithelial-mesenchymal transition (EMT) in a mouse model for mammary adenocarcinoma (WAP-T mice). We demonstrate that silencing of CEACAM1 in WAP-T tumor-derived G-2 cells induces epithelial-mesenchymal plasticity (EMP), as evidenced by typical changes of gene expression, morphology and increased invasion. In contrast, reintroduction of CEACAM1 into G-2 cells reversed up-regulation of genes imposing mesenchymal transition, as well as cellular invasion. We identified the Wnt-pathway as target for CEACAM1-mediated repression of EMT. Importantly, β-catenin phosphorylation status and transcriptional activity strongly depend on CEACAM1 expression: CEACAM1high G-2 cells displayed enhanced phosphorylation of β-catenin at S33/S37/T41 and decreased phosphorylation at Y86, thereby inhibiting canonical Wnt/β-catenin signaling. We identified Src-homology 2 domain-containing phosphatase 2 (SHP-2) as a critical binding partner of CEACAM1 that could modulate β-catenin Y86 phosphorylation. Hence, CEACAM1 serves as a scaffold that controls membrane proximal β-catenin signaling. In vivo, mammary tumors of WAP-T/CEACAM1null mice displayed increased nuclear translocation of β-catenin and a dramatically enhanced metastasis rate compared to WAP-T mice. Hence, CEACAM1 controls EMT in vitro and in vivo by site-specific regulation of β-catenin phosphorylation. Survival analyses of human mammary carcinoma patients corroborated these data, indicating that CEACAM1 is a prognostic marker for breast cancer survival.
Collapse
Affiliation(s)
- Wegwitz Florian
- Clinic for General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Georg-August-University of Göttingen, D-37077 Göttingen, Germany
- Institute for Tumor Biology, University Medical Center-Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Eva Lenfert
- Institute for Tumor Biology, University Medical Center-Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Daniela Gerstel
- Center for Diagnostics, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Lena von Ehrenstein
- Institute for Tumor Biology, University Medical Center-Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Julia Einhoff
- Institute for Tumor Biology, University Medical Center-Hamburg-Eppendorf, D-20251 Hamburg, Germany
- Pharmaceutical Institute, Christian-Albrechts-University Kiel, D-24118 Kiel, Germany
| | - Geske Schmidt
- Clinic for General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Georg-August-University of Göttingen, D-37077 Göttingen, Germany
| | - Matthew Logsdon
- Clinic for General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Georg-August-University of Göttingen, D-37077 Göttingen, Germany
| | - Johanna Brandner
- Dermatology and Venerology Department and Clinic, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Gisa Tiegs
- Institute for Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Nicole Beauchemin
- Goodman Cancer Research Centre and Departments of Biochemistry, Medicine and Oncology, McGill University, Montreal, H3G1Y6, Canada
| | - Christoph Wagener
- Center for Diagnostics, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Wolfgang Deppert
- Institute for Tumor Biology, University Medical Center-Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Andrea Kristina Horst
- Institute for Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| |
Collapse
|
15
|
Arabzadeh A, Dupaul-Chicoine J, Breton V, Haftchenary S, Yumeen S, Turbide C, Saleh M, McGregor K, Greenwood CMT, Akavia UD, Blumberg RS, Gunning PT, Beauchemin N. Carcinoembryonic Antigen Cell Adhesion Molecule 1 long isoform modulates malignancy of poorly differentiated colon cancer cells. Gut 2016; 65:821-9. [PMID: 25666195 PMCID: PMC4826327 DOI: 10.1136/gutjnl-2014-308781] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 01/20/2015] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Nearly 20%-29% of patients with colorectal cancer (CRC) succumb to liver or lung metastasis and there is a dire need for novel targets to improve the survival of patients with metastasis. The long isoform of the Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1-L or CC1-L) is a key regulator of immune surveillance in primary CRC, but its role in metastasis remains largely unexplored. We have examined how CC1-L expression impacts on colon cancer liver metastasis. DESIGN Murine MC38 transfected with CC1-L were evaluated in vitro for proliferation, migration and invasion, and for in vivo experimental liver metastasis. Using shRNA silencing or pharmacological inhibition, we delineated the role in liver metastasis of Chemokine (C-C motif) Ligand 2 (CCL2) and Signal Transducer and Activator of Transcription 3 (STAT3) downstream of CC1-L. We further assessed the clinical relevance of these findings in a cohort of patients with CRC. RESULTS MC38-CC1-L-expressing cells exhibited significantly reduced in vivo liver metastasis and displayed decreased CCL2 chemokine secretion and reduced STAT3 activity. Down-modulation of CCL2 expression and pharmacological inhibition of STAT3 activity in MC38 cells led to reduced cell invasion capacity and decreased liver metastasis. The clinical relevance of our findings is illustrated by the fact that high CC1 expression in patients with CRC combined with some inflammation-regulated and STAT3-regulated genes correlate with improved 10-year survival. CONCLUSIONS CC1-L regulates inflammation and STAT3 signalling and contributes to the maintenance of a less-invasive CRC metastatic phenotype of poorly differentiated carcinomas.
Collapse
Affiliation(s)
- Azadeh Arabzadeh
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | | | - Valérie Breton
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Sina Haftchenary
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Sara Yumeen
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Claire Turbide
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Maya Saleh
- Complex Trait Group, McGill University, Montreal, Quebec, Canada
| | - Kevin McGregor
- Departments of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| | - Celia M T Greenwood
- Departments of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| | - Uri David Akavia
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Richard S Blumberg
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Patrick T Gunning
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Nicole Beauchemin
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
16
|
Arabzadeh A, Beauchemin N. Stromal CEACAM1 expression regulates colorectal cancer metastasis. Oncoimmunology 2014; 1:1205-1207. [PMID: 23170281 PMCID: PMC3494647 DOI: 10.4161/onci.20735] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Colorectal cancer metastasis to Ceacam1-/- livers is significantly impaired, compared with wild type livers, due to decreased endothelial cell survival, reduced tumor cell proliferation, diminished immune infiltration and altered chemokine expression. Ceacam1-/- myeloid-derived suppressor cells diminish metastatic burden, as confirmed by bone marrow transplantation and adoptive transfer experiments.
Collapse
Affiliation(s)
- Azadeh Arabzadeh
- Goodman Cancer Research Centre; McGill University; Montreal, QC Canada
| | | |
Collapse
|
17
|
Bockhorn J, Prat A, Chang YF, Liu X, Huang S, Shang M, Nwachukwu C, Gomez-Vega MJ, Harrell JC, Olopade OI, Perou CM, Liu H. Differentiation and loss of malignant character of spontaneous pulmonary metastases in patient-derived breast cancer models. Cancer Res 2014; 74:7406-17. [PMID: 25339353 DOI: 10.1158/0008-5472.can-14-1188] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Patient-derived human-in-mouse xenograft models of breast cancer (PDX models) that exhibit spontaneous lung metastases offer a potentially powerful model of cancer metastasis. In this study, we evaluated the malignant character of lung micrometastases that emerge in such models after orthotopic implantation of human breast tumor cells into the mouse mammary fat pad. Interestingly, relative to the parental primary breast tumors, the lung metastasis (met)-derived mammary tumors exhibited a slower growth rate and a reduced metastatic potential with a more differentiated epithelial status. Epigenetic correlates were determined by gene array analyses. Lung met-derived tumors displayed differential expression of negative regulators of cell proliferation and metabolism and positive regulators of mammary epithelial differentiation. Clinically, this signature correlated with breast tumor subtypes. We identified hsa-miR-138 (miR-138) as a novel regulator of invasion and epithelial-mesenchymal transition in breast cancer cells, acting by directly targeting the polycomb epigenetic regulator EZH2. Mechanistic investigations showed that GATA3 transcriptionally controlled miR-138 levels in lung metastases. Notably, the miR-138 activity signature served as a novel independent prognostic marker for patient survival beyond traditional pathologic variables, intrinsic subtypes, or a proliferation gene signature. Our results highlight the loss of malignant character in some lung micrometastatic lesions and the epigenetic regulation of this phenotype.
Collapse
Affiliation(s)
- Jessica Bockhorn
- The Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois. Stanford Cancer Institute, Stanford University, Stanford, California
| | - Aleix Prat
- Translational Genomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain. Department of Medical Oncology, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Ya-Fang Chang
- The Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois
| | - Xia Liu
- Deparment of Pathology, Case Comprehensive Cancer Center, and National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Simo Huang
- Deparment of Pathology, Case Comprehensive Cancer Center, and National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Meng Shang
- Illinois Institute of Technology, Chicago, Illinois
| | - Chika Nwachukwu
- Center for Clinical Cancer Genetics, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Maria J Gomez-Vega
- Center for Clinical Cancer Genetics, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - J Chuck Harrell
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Olufunmilayo I Olopade
- Center for Clinical Cancer Genetics, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Huiping Liu
- Deparment of Pathology, Case Comprehensive Cancer Center, and National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
18
|
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]
|
19
|
Tsai YH, Disson O, Bierne H, Lecuit M. Murinization of internalin extends its receptor repertoire, altering Listeria monocytogenes cell tropism and host responses. PLoS Pathog 2013; 9:e1003381. [PMID: 23737746 PMCID: PMC3667765 DOI: 10.1371/journal.ppat.1003381] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 04/09/2013] [Indexed: 01/17/2023] Open
Abstract
Listeria monocytogenes (Lm) is an invasive foodborne pathogen that leads to severe central nervous system and maternal-fetal infections. Lm ability to actively cross the intestinal barrier is one of its key pathogenic properties. Lm crosses the intestinal epithelium upon the interaction of its surface protein internalin (InlA) with its host receptor E-cadherin (Ecad). InlA-Ecad interaction is species-specific, does not occur in wild-type mice, but does in transgenic mice expressing human Ecad and knock-in mice expressing humanized mouse Ecad. To study listeriosis in wild-type mice, InlA has been “murinized” to interact with mouse Ecad. Here, we demonstrate that, unexpectedly, murinized InlA (InlAm) mediates not only Ecad-dependent internalization, but also N-cadherin-dependent internalization. Consequently, InlAm-expressing Lm targets not only goblet cells expressing luminally-accessible Ecad, as does Lm in humanized mice, but also targets villous M cells, which express luminally-accessible N-cadherin. This aberrant Lm portal of entry results in enhanced innate immune responses and intestinal barrier damage, both of which are not observed in wild-type Lm-infected humanized mice. Murinization of InlA therefore not only extends the host range of Lm, but also broadens its receptor repertoire, providing Lm with artifactual pathogenic properties. These results challenge the relevance of using InlAm-expressing Lm to study human listeriosis and in vivo host responses to this human pathogen. Co-evolution of microbes with their hosts can select stringently specific host-microbe interactions at the cell, tissue and species levels. Listeria monocytogenes (Lm) is a foodborne pathogen that causes a deadly systemic infection in humans. Lm crosses the intestinal epithelium upon the interaction of its surface protein InlA with E-cadherin (Ecad). InlA-Ecad interaction is species-specific, does not occur in wild-type mice, but does in transgenic mice expressing human Ecad and knock-in mice expressing humanized mouse Ecad. To study listeriosis in wild-type mice, InlA has been “murinized” to interact with mouse Ecad. Here, we demonstrate that in addition to interacting with mouse Ecad, InlAm also uses N-cadherin as a receptor, whereas InlA does not. This artifactual InlAm-N-cadherin interaction promotes bacterial translocation across villous M cells, a cell type which is not targeted by InlA-expressing bacteria. This leads to intestinal inflammation and intestinal barrier damage, both of which are not seen in humans and humanized mouse models permissive to InlA-Ecad interaction. These results challenge the relevance of using InlAm-expressing Lm as a model to study human listeriosis and host responses to this pathogen. They also illustrate that caution must be exercised before using “murinized” pathogens to study human infectious diseases.
Collapse
Affiliation(s)
- Yu-Huan Tsai
- Institut Pasteur, Biology of Infection Unit, Paris, France
- Inserm U1117, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Olivier Disson
- Institut Pasteur, Biology of Infection Unit, Paris, France
- Inserm U1117, Paris, France
| | - Hélène Bierne
- Institut Pasteur, Unité des Interactions Bactéries Cellules, Paris, France
- Inserm, U604, Paris, France
- INRA, USC2020, Paris, France
| | - Marc Lecuit
- Institut Pasteur, Biology of Infection Unit, Paris, France
- Inserm U1117, Paris, France
- Institut Pasteur, French National Reference Center and World Health Organization Collaborating Center on Listeria, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Institut Imagine, Paris, France
- Necker-Enfants Malades University Hospital, APHP, Division of Infectious Diseases and Tropical Medicine, Paris, France
- * E-mail:
| |
Collapse
|
20
|
Prager S, Singer BB, Bendix I, Schlager GW, Bertling F, Ceylan B, Keller M, Felderhoff-Mueser U, Ergün S. CEACAM1 expression in oligodendrocytes of the developing rat brain shows a spatiotemporal relation to myelination and is altered in a model of encephalopathy of prematurity. Dev Neurosci 2013; 35:226-40. [PMID: 23651919 DOI: 10.1159/000348436] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 01/28/2013] [Indexed: 11/19/2022] Open
Abstract
CEACAM1 is the founder molecule of the family of 'carcinoembryonic antigen-related cell adhesion molecules' and part of the immunoglobulin superfamily. Due to its role as a coreceptor to many other receptors (e.g. Toll-like receptor 2, Toll-like receptor 4, T-cell receptor, B-cell receptor, epidermal growth factor receptor and vascular endothelial growth factor receptor) and its different isoforms, CEACAM1 is a multifunctional protein with an impact on proliferation and differentiation of multiple cell types. Although different modes of action in other tissues are described, the role of CEACAM1 in the developing brain remains elusive. Here we report for the first time that CEACAM1 is expressed ontogenetically in oligodendrocytes of the developing rat brain, and that CEACAM1 expression has a spatiotemporal relation to myelination. In addition, CEACAM1 expression is altered in a model of hyperoxia- and inflammation-induced encephalopathy of prematurity, a myelination disorder of children born preterm. Furthermore, primary oligodendrocytes stimulated with CEACAM1 show increased myelination. Therefore, we postulate that CEACAM1 is, at least in part, involved in hyperoxia- and inflammation-induced disruption of myelination, but may also play a role in intact myelination as it is ontogenetically expressed in myelinating oligodendrocytes.
Collapse
Affiliation(s)
- Sebastian Prager
- Department of Pediatrics 1, Neonatology, University Hospital Essen, Essen, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Zhang H, Eisenried A, Zimmermann W, Shively JE. Role of CEACAM1 and CEACAM20 in an in vitro model of prostate morphogenesis. PLoS One 2013; 8:e53359. [PMID: 23358633 PMCID: PMC3554727 DOI: 10.1371/journal.pone.0053359] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 11/29/2012] [Indexed: 11/19/2022] Open
Abstract
CEACAM20, a novel member of the CEACAM1 gene family with expression limited to the lumen of small intestine, testes, and prostate, is co-expressed with CEACAM1 in adult prostate tissue and down-regulated to the same extent as CEACAM1 in prostate cancer. Since prostate cancer often involves loss of epithelial lumen formation, we hypothesized that CEACAM20 and CEACAM1 play important roles in lumen formation of normal prostate epithelium. When prostate cells were grown on Matrigel as a source of extracellular matrix (ECM), they differentiated into acinar structures with single tubules and well-defined lumina closely resembling embryonic prostate organoids. Confocal microscopic analysis revealed restriction of CEACAM20 to acini and CEACAM1 to tubule structures, respectively. Inhibition of CEACAM1 with antibodies or soluble CEACAM1 or antisense oligonucleotides inhibited tubule formation by over 50% while the remaining tubules were stunted. Inhibition of CEACAM20 with antisense oligonucleotides completely inhibited tubule formation and stunted the growth of acini. We conclude that CEACAM20 and CEACAM1 not only mark the lumina of adult prostate tissue but also play a critical role in the vitro generation of prostate organoids.
Collapse
Affiliation(s)
- Hui Zhang
- City of Hope Irell & Manella Graduate School of Biological Sciences, Duarte, California, United States of America
- Department of Immunology, Beckman Research Institute of City of Hope, Duarte, California, United States of America
| | - Andreas Eisenried
- Anästhesiologische Klinik, Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - John E. Shively
- Department of Immunology, Beckman Research Institute of City of Hope, Duarte, California, United States of America
- * E-mail:
| |
Collapse
|
22
|
Kammerer R, Rüttiger L, Riesenberg R, Schäuble C, Krupar R, Kamp A, Sunami K, Eisenried A, Hennenberg M, Grunert F, Bress A, Battaglia S, Schrewe H, Knipper M, Schneider MR, Zimmermann W. Loss of mammal-specific tectorial membrane component carcinoembryonic antigen cell adhesion molecule 16 (CEACAM16) leads to hearing impairment at low and high frequencies. J Biol Chem 2012; 287:21584-98. [PMID: 22544735 DOI: 10.1074/jbc.m111.320481] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The vertebrate-restricted carcinoembryonic antigen gene family evolves extremely rapidly. Among their widely expressed members, the mammal-specific, secreted CEACAM16 is exceptionally well conserved and specifically expressed in the inner ear. To elucidate a potential auditory function, we inactivated murine Ceacam16 by homologous recombination. In young Ceacam16(-/-) mice the hearing threshold for frequencies below 10 kHz and above 22 kHz was raised. This hearing impairment progressed with age. A similar phenotype is observed in hearing-impaired members of Family 1070 with non-syndromic autosomal dominant hearing loss (DFNA4) who carry a missense mutation in CEACAM16. CEACAM16 was found in interdental and Deiters cells and was deposited in the tectorial membrane of the cochlea between postnatal days 12 and 15, when hearing starts in mice. In cochlear sections of Ceacam16(-/-) mice tectorial membranes were significantly more often stretched out as compared with wild-type mice where they were mostly contracted and detached from the outer hair cells. Homotypic cell sorting observed after ectopic cell surface expression of the carboxyl-terminal immunoglobulin variable-like N2 domain of CEACAM16 indicated that CEACAM16 can interact in trans. Furthermore, Western blot analyses of CEACAM16 under reducing and non-reducing conditions demonstrated oligomerization via unpaired cysteines. Taken together, CEACAM16 can probably form higher order structures with other tectorial membrane proteins such as α-tectorin and β-tectorin and influences the physical properties of the tectorial membrane. Evolution of CEACAM16 might have been an important step for the specialization of the mammalian cochlea, allowing hearing over an extended frequency range.
Collapse
Affiliation(s)
- Robert Kammerer
- Instute of Immunology, Friedrich-Loeffler Institut, 17493 Greifswald-Insel Riems, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
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
|
24
|
LeBlanc S, Arabzadeh A, Benlolo S, Breton V, Turbide C, Beauchemin N, Nouvion AL. CEACAM1 deficiency delays important wound healing processes. Wound Repair Regen 2012; 19:745-52. [PMID: 22092845 DOI: 10.1111/j.1524-475x.2011.00742.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cutaneous wound healing is a complex process that requires the coordination of many cell types to achieve proper tissue repair. Four major overlapping processes have been identified in wound healing: hemostasis, inflammation, reepithelialization and granulation tissue formation, and tissue remodeling. Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is a glycoprotein expressed in epithelial, endothelial, lymphoid, and myeloid cells. Given its known roles in angiogenesis, cell migration, and immune functions, we hypothesized that CEACAM1 might also be involved in cutaneous wound healing and that a number of relevant CEACAM1-positive cell types might contribute to wound healing. To evaluate the role of CEACAM1 in these processes, 6-mm-diameter skin wounds were inflicted on Ceacam1(-/-) and wild-type mice. Herein, we demonstrate that CEACAM1 deletion indeed affects wound healing in three key ways. Infiltration of F4/80(+) macrophages was decreased in Ceacam1(-/-) wounds, altering inflammatory processes. Reepithelialization in Ceacam1(-/-) wounds was delayed. Furthermore, the vascular density of the granulation tissue in Ceacam1(-/-) wounds was significantly diminished. These results confirm CEACAM1's role as an important regulator of key processes in cutaneous wound healing, although whether this works via a specific cell type or alterations in the functioning of multiple processes remains to be determined.
Collapse
Affiliation(s)
- Sarah LeBlanc
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
25
|
Patel PR, Ramakrishnan SK, Kaw MK, Raphael CK, Ghosh S, Marino JS, Heinrich G, Lee SJ, Bourey RE, Hill JW, Jung DY, Morgan DA, Kim JK, Rahmouni SK, Najjar SM. Increased metabolic rate and insulin sensitivity in male mice lacking the carcino-embryonic antigen-related cell adhesion molecule 2. Diabetologia 2012; 55:763-72. [PMID: 22159884 PMCID: PMC3272352 DOI: 10.1007/s00125-011-2388-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 11/07/2011] [Indexed: 10/14/2022]
Abstract
AIMS/HYPOTHESIS The carcino-embryonic antigen-related cell adhesion molecule (CEACAM)2 is produced in many feeding control centres in the brain, but not in peripheral insulin-targeted tissues. Global Ceacam2 null mutation causes insulin resistance and obesity resulting from hyperphagia and hypometabolism in female Ceacam2 homozygous null mutant mice (Cc2 [also known as Ceacam2](-/-)) mice. Because male mice are not obese, the current study examined their metabolic phenotype. METHODS The phenotype of male Cc2(-/-) mice was characterised by body fat composition, indirect calorimetry, hyperinsulinaemic-euglycaemic clamp analysis and direct recording of sympathetic nerve activity. RESULTS Despite hyperphagia, total fat mass was reduced, owing to the hypermetabolic state in male Cc2(-/-) mice. In contrast to females, male mice also exhibited insulin sensitivity with elevated β-oxidation in skeletal muscle, which is likely to offset the effects of increased food intake. Males and females had increased brown adipogenesis. However, only males had increased activation of sympathetic tone regulation of adipose tissue and increased spontaneous activity. The mechanisms underlying sexual dimorphism in energy balance with the loss of Ceacam2 remain unknown. CONCLUSIONS/INTERPRETATION These studies identified a novel role for CEACAM2 in the regulation of metabolic rate and insulin sensitivity via effects on brown adipogenesis, sympathetic nervous outflow to brown adipose tissue, spontaneous activity and energy expenditure in skeletal muscle.
Collapse
Affiliation(s)
- P. R. Patel
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA
- Department of Physiology and Pharmacology at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| | - S. K. Ramakrishnan
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA
- Department of Physiology and Pharmacology at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| | - M. K. Kaw
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA
- Department of Physiology and Pharmacology at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| | - C. K. Raphael
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA
- Department of Physiology and Pharmacology at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| | - S. Ghosh
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA
- Department of Physiology and Pharmacology at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| | - J. S. Marino
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA
- Department of Physiology and Pharmacology at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| | - G. Heinrich
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA
- Department of Physiology and Pharmacology at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| | - S. J. Lee
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA
- Department of Physiology and Pharmacology at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| | - R. E. Bourey
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA
- Department of Internal Medicine at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| | - J. W. Hill
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA
- Department of Physiology and Pharmacology at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| | - D. Y. Jung
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA, USA
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - D. A. Morgan
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - J. K. Kim
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA, USA
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - S. K. Rahmouni
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - S. M. Najjar
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Health Science Campus, 3000 Arlington Avenue, Mail Stop 1009, Toledo, OH 43614, USA,
- Department of Physiology and Pharmacology at the College of Medicine and Life Sciences, University of Toledo, Health Science Campus, Toledo, OH, USA
| |
Collapse
|
26
|
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
|
27
|
Abstract
Tumor invasion and metastasis is a multi-step process that requires adaptation of cancer cells to conditions that they encounter during their journey to distant body sites. Understanding the molecular processes that underlie this adaptation is of exceeding importance because most cancer patients die because of metastases rather than primary tumors. In this review we assess genetically engineered mouse models (GEMMs) that have been established to investigate mechanisms of cancer invasion and metastasis.
Collapse
|
28
|
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
|
29
|
|
30
|
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
|
31
|
Müller MM, Klaile E, Vorontsova O, Singer BB, Obrink B. Homophilic adhesion and CEACAM1-S regulate dimerization of CEACAM1-L and recruitment of SHP-2 and c-Src. ACTA ACUST UNITED AC 2009; 187:569-81. [PMID: 19948503 PMCID: PMC2779222 DOI: 10.1083/jcb.200904150] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Carcinoembryonic antigen (CEA)-related cell adhesion molecule 1 (CAM1 [CEACAM1]) mediates homophilic cell adhesion and regulates signaling. Although there is evidence that CEACAM1 binds and activates SHP-1, SHP-2, and c-Src, knowledge about the mechanism of transmembrane signaling is lacking. To analyze the regulation of SHP-1/SHP-2/c-Src binding, we expressed various CFP/YFP-tagged CEACAM1 isoforms in epithelial cells. The supramolecular organization of CEACAM1 was examined by cross-linking, coclustering, coimmunoprecipitation, and fluorescence resonance energy transfer. SHP-1/SHP-2/c-Src binding was monitored by coimmunoprecipitation and phosphotyrosine-induced recruitment to CEACAM1-L in cellular monolayers. We find that trans-homophilic CEACAM1 binding induces cis-dimerization by an allosteric mechanism transmitted by the N-terminal immunoglobulin-like domain. The balance of SHP-2 and c-Src binding is dependent on the monomer/dimer equilibrium of CEACAM1-L and is regulated by trans-binding, whereas SHP-1 does not bind under physiological conditions. CEACAM1-L homodimer formation is reduced by coexpression of CEACAM1-S and modulated by antibody ligation. These data suggest that transmembrane signaling by CEACAM1 operates by alteration of the monomer/dimer equilibrium, which leads to changes in the SHP-2/c-Src-binding ratio.
Collapse
Affiliation(s)
- Mario M Müller
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | | | | | | |
Collapse
|
32
|
Klaile E, Vorontsova O, Sigmundsson K, Müller MM, Singer BB, Öfverstedt LG, Svensson S, Skoglund U, Öbrink B. The CEACAM1 N-terminal Ig domain mediates cis- and trans-binding and is essential for allosteric rearrangements of CEACAM1 microclusters. J Cell Biol 2009; 187:553-67. [PMID: 19948502 PMCID: PMC2779236 DOI: 10.1083/jcb.200904149] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Accepted: 10/19/2009] [Indexed: 11/22/2022] Open
Abstract
Cell adhesion molecules (CAMs) sense the extracellular microenvironment and transmit signals to the intracellular compartment. In this investigation, we addressed the mechanism of signal generation by ectodomains of single-pass transmembrane homophilic CAMs. We analyzed the structure and homophilic interactions of carcinoembryonic antigen (CEA)-related CAM 1 (CEACAM1), which regulates cell proliferation, apoptosis, motility, morphogenesis, and microbial responses. Soluble and membrane-attached CEACAM1 ectodomains were investigated by surface plasmon resonance-based biosensor analysis, molecular electron tomography, and chemical cross-linking. The CEACAM1 ectodomain, which is composed of four glycosylated immunoglobulin-like (Ig) domains, is highly flexible and participates in both antiparallel (trans) and parallel (cis) homophilic binding. Membrane-attached CEACAM1 ectodomains form microclusters in which all four Ig domains participate. Trans-binding between the N-terminal Ig domains increases formation of CEACAM1 cis-dimers and changes CEACAM1 interactions within the microclusters. These data suggest that CEACAM1 transmembrane signaling is initiated by adhesion-regulated changes of cis-interactions that are transmitted to the inner phase of the plasma membrane.
Collapse
Affiliation(s)
- Esther Klaile
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Olga Vorontsova
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Mario M. Müller
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Lars-Göran Öfverstedt
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Stina Svensson
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Ulf Skoglund
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Björn Öbrink
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
33
|
|
34
|
Nouvion AL, Beauchemin N. [CEACAM1 as a central modulator of metabolism, tumor progression, angiogenesis and immunity]. Med Sci (Paris) 2009; 25:247-52. [PMID: 19361387 DOI: 10.1051/medsci/2009253247] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
CEACAM1 (carcinoembryonic antigen cell adhesion molecule 1), a member of the immunoglobulin (Ig) superfamily, is a heavily glycosylated protein. This glycoprotein exhibits an intracytoplasmic region that can be either long (71-73 amino acids) with two inhibitory tyrosine-phosphorylated motifs and several phosphorylated serine residues, or short (10 amino acid). CEACAM1 is a multifunctional protein that plays a role in intercellular adhesion, as an inhibitor of tumor development, as a bacterial adhesin, and as a receptor for the mouse hepatitis virus. Moreover, CEACAM1 is an active regulator of cell signaling, modulating the insulin or EGF receptor pathways in epithelial cells or the Zap-70 pathway in hematopoietic cells. The recent development of genetically modified mouse models altering the Ceacam1 gene corroborates most of these data, but also highlights CEACAM1's functional complexity. Thus, in addition to the functions identified previously, CEACAM1 is an important regulator of lipid metabolism, of tumor progression as a regulator of the Wnt signaling pathway, of normal and tumor neo-angiogenesis and of immunity.
Collapse
Affiliation(s)
- Anne-Laure Nouvion
- Centre de cancérologie McGill, Département de biochimie, Université McGill, 3655 Promenade Sir-William-Osler, Montréal, Québec, H3G 1Y6, Canada
| | | |
Collapse
|
35
|
Current Opinion in Oncology. Current world literature. Curr Opin Oncol 2009; 21:386-92. [PMID: 19509503 DOI: 10.1097/cco.0b013e32832e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
36
|
Dupasquier S, Abdel-Samad R, Glazer RI, Bastide P, Jay P, Joubert D, Cavaillès V, Blache P, Quittau-Prévostel C. A new mechanism of SOX9 action to regulate PKCalpha expression in the intestine epithelium. J Cell Sci 2009; 122:2191-6. [PMID: 19509063 DOI: 10.1242/jcs.036483] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Variations of protein kinase C (PKC) expression greatly influence the proliferation-to-differentiation transition (PDT) of intestinal epithelial cells and might have an important impact on intestinal tumorigenesis. We demonstrate here that the expression of PKCalpha in proliferating intestinal epithelial cells is repressed both in vitro and in vivo by the SOX9 transcription factor. This repression does not require DNA binding of the SOX9 high-mobility group (HMG) domain but is mediated through a new mechanism of SOX9 action requiring the central and highly conserved region of SOXE members. Because SOX9 expression is itself upregulated by Wnt-APC signaling in intestinal epithelial cells, the present study points out this transcription factor as a molecular link between the Wnt-APC pathway and PKCalpha. These results provide a potential explanation for the decrease of PKCalpha expression in colorectal cancers with constitutive activation of the Wnt-APC pathway.
Collapse
|