1
|
Zhao L, Li T, Zhou Y, Wang P, Luo L. Monoclonal antibody targeting CEACAM1 enhanced the response to anti-PD1 immunotherapy in non-small cell lung cancer. Int Immunopharmacol 2024; 143:113395. [PMID: 39426236 DOI: 10.1016/j.intimp.2024.113395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 09/27/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
Carcinoembryonic antigen-related cellular adhesion molecule 1 (CEACAM1), an extensively studied cell surface molecule, mainly expressed by certain epithelial, endothelial, lymphoid and myeloid cells, and is an attractive target for cancer immunotherapy. Here, to investigate the anti-tumor effects and mechanisms of CEACAM1 antibody, we prepared the antibody and explored its anti-tumor effects on Non-small Cell Lung Cancer (NSCLC) in vitro and in vivo. Firstly, antigen of human CEACAM1 recombinant protein was immunized on BALB/c mice and the high-affinity mouse anti-human monoclonal antibody 3C11 was selected by hybridoma technique. Next, ELISA was applied to detect the blocking effects of 3C11 on CEACAM1-CEACAM1 and CEACAM1-CEACAM5. Then, cell assays and ELISA were used to evaluate the role of 3C11 in blocking CEACAM1-CEACAM1 immunosuppressive signal transduction between dendritic cells (DCs) and T cells or natural killer cells (NK) and tumor cells. Finally, the synergistic anti-tumor effect of 3C11 combined with anti-PD-1 antibody was evaluated through cell stimulation assays and NCI-H358-induced tumor models in mice. The results showed the EC50 of 3C11 binding to NCI-H358 or exhausted T cells were 0.04971 μg/mL and 0.03475 μg/mL, respectively. 3C11 activated the exhausted T cells and enhanced the killing effect of NK by blocking CEACAM1-CEACAM1. In addition, the combination of 3C11 and anti-PD1 antibody produced synergistic anti-tumor effect on NSCLC. Its improved tumor growth inhibition value (TGI) of anti-PD-1 from 18 % to 85 % in vivo. These findings suggest that 3C11 can be considered an effective immunotherapy drug for NSCLC.
Collapse
Affiliation(s)
- Lianqi Zhao
- School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, PR China; Ouyue Biotech. Inc, Suzhou, Jiangsu 215400, PR China; In Vivo Pharmacology, Frontage Laboratories, Suzhou, Jiangsu 215400, PR China
| | - Tingting Li
- School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, PR China; Ouyue Biotech. Inc, Suzhou, Jiangsu 215400, PR China
| | - Yinwei Zhou
- School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, PR China
| | - Pengbo Wang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, PR China.
| | - Lin Luo
- School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, PR China.
| |
Collapse
|
2
|
Bogdan C, Islam NAK, Barinberg D, Soulat D, Schleicher U, Rai B. The immunomicrotope of Leishmania control and persistence. Trends Parasitol 2024; 40:788-804. [PMID: 39174373 DOI: 10.1016/j.pt.2024.07.013] [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: 06/20/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 08/24/2024]
Abstract
Leishmania is an intracellular protozoan transmitted by sand fly vectors; it causes cutaneous, mucocutaneous, or visceral disease. Its growth and survival are impeded by type 1 T helper cell responses, which entail interferon (IFN)-γ-mediated macrophage activation. Leishmania partially escapes this host defense by triggering immune cell and cytokine responses that favor parasite replication rather than killing. Novel methods for in situ analyses have revealed that the pathways of immune control and microbial evasion are strongly influenced by the tissue context, the micro milieu factors, and the metabolism at the site of infection, which we collectively term the 'immunomicrotope'. Understanding the components and the impact of the immunomicrotope will enable the development of novel strategies for the treatment of chronic leishmaniasis.
Collapse
Affiliation(s)
- Christian Bogdan
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany; FAU Profile Center Immunomedicine, FAU Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany.
| | - Noor-A-Kasida Islam
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany
| | - David Barinberg
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany
| | - Didier Soulat
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany; FAU Profile Center Immunomedicine, FAU Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany
| | - Ulrike Schleicher
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany; FAU Profile Center Immunomedicine, FAU Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany
| | - Baplu Rai
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany
| |
Collapse
|
3
|
L’Estrange-Stranieri E, Gottschalk TA, Wright MD, Hibbs ML. The dualistic role of Lyn tyrosine kinase in immune cell signaling: implications for systemic lupus erythematosus. Front Immunol 2024; 15:1395427. [PMID: 39007135 PMCID: PMC11239442 DOI: 10.3389/fimmu.2024.1395427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 06/17/2024] [Indexed: 07/16/2024] Open
Abstract
Systemic lupus erythematosus (SLE, lupus) is a debilitating, multisystem autoimmune disease that can affect any organ in the body. The disease is characterized by circulating autoantibodies that accumulate in organs and tissues, which triggers an inflammatory response that can cause permanent damage leading to significant morbidity and mortality. Lyn, a member of the Src family of non-receptor protein tyrosine kinases, is highly implicated in SLE as remarkably both mice lacking Lyn or expressing a gain-of-function mutation in Lyn develop spontaneous lupus-like disease due to altered signaling in B lymphocytes and myeloid cells, suggesting its expression or activation state plays a critical role in maintaining tolerance. The past 30 years of research has begun to elucidate the role of Lyn in a duplicitous signaling network of activating and inhibitory immunoreceptors and related targets, including interactions with the interferon regulatory factor family in the toll-like receptor pathway. Gain-of-function mutations in Lyn have now been identified in human cases and like mouse models, cause severe systemic autoinflammation. Studies of Lyn in SLE patients have presented mixed findings, which may reflect the heterogeneity of disease processes in SLE, with impairment or enhancement in Lyn function affecting subsets of SLE patients that may be a means of stratification. In this review, we present an overview of the phosphorylation and protein-binding targets of Lyn in B lymphocytes and myeloid cells, highlighting the structural domains of the protein that are involved in its function, and provide an update on studies of Lyn in SLE patients.
Collapse
Affiliation(s)
- Elan L’Estrange-Stranieri
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Timothy A. Gottschalk
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Mark D. Wright
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Margaret L. Hibbs
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
4
|
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
|
5
|
Yiong CS, Lin TP, Lim VY, Toh TB, Yang VS. Biomarkers for immune checkpoint inhibition in sarcomas - are we close to clinical implementation? Biomark Res 2023; 11:75. [PMID: 37612756 PMCID: PMC10463641 DOI: 10.1186/s40364-023-00513-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 07/26/2023] [Indexed: 08/25/2023] Open
Abstract
Sarcomas are a group of diverse and complex cancers of mesenchymal origin that remains poorly understood. Recent developments in cancer immunotherapy have demonstrated a potential for better outcomes with immune checkpoint inhibition in some sarcomas compared to conventional chemotherapy. Immune checkpoint inhibitors (ICIs) are key agents in cancer immunotherapy, demonstrating improved outcomes in many tumor types. However, most patients with sarcoma do not benefit from treatment, highlighting the need for identification and development of predictive biomarkers for response to ICIs. In this review, we first discuss United States (US) Food and Drug Administration (FDA)-approved and European Medicines Agency (EMA)-approved biomarkers, as well as the limitations of their use in sarcomas. We then review eight potential predictive biomarkers and rationalize their utility in sarcomas. These include gene expression signatures (GES), circulating neutrophil-to-lymphocyte ratio (NLR), indoleamine 2,3-dioxygenase (IDO), lymphocyte activation gene 3 (LAG-3), T cell immunoglobin and mucin domain-containing protein 3 (TIM-3), TP53 mutation status, B cells, and tertiary lymphoid structures (TLS). Finally, we discuss the potential for TLS as both a predictive and prognostic biomarker for ICI response in sarcomas to be implemented in the clinic.
Collapse
Affiliation(s)
- Chin Sern Yiong
- Translational Precision Oncology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
- Department of Pharmacy, National University of Singapore, Singapore, 117544, Singapore
| | - Tzu Ping Lin
- Translational Precision Oncology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
- Department of Pharmacy, National University of Singapore, Singapore, 117544, Singapore
| | - Vivian Yujing Lim
- Translational Precision Oncology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
| | - Tan Boon Toh
- The N.1 Institute for Health, National University of Singapore, Singapore, Singapore
- The Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, Singapore
| | - Valerie Shiwen Yang
- Translational Precision Oncology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore.
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore.
- Duke-NUS Medical School, Oncology Academic Clinical Program, Singapore, 169857, Singapore.
| |
Collapse
|
6
|
Dulal D, Boring A, Terrero D, Johnson T, Tiwari AK, Raman D. Tackling of Immunorefractory Tumors by Targeting Alternative Immune Checkpoints. Cancers (Basel) 2023; 15:2774. [PMID: 37345111 PMCID: PMC10216651 DOI: 10.3390/cancers15102774] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/09/2023] [Accepted: 05/12/2023] [Indexed: 06/23/2023] Open
Abstract
Physiologically, well known or traditional immune checkpoints (ICs), such as CTLA-4 and PD-1, are in place to promote tolerance to self-antigens and prevent generation of autoimmunity. In cancer, the ICs are effectively engaged by the tumor cells or stromal ells from the tumor microenvironment through expression of cognate ligands for the ICs present on the cell surface of CD8+ T lymphocytes. The ligation of ICs on CD8+ T lymphocytes triggers inhibitory signaling pathways, leading to quiescence or an exhaustion of CD8+ T lymphocytes. This results in failure of immunotherapy. To overcome this, several FDA-approved therapeutic antibodies are available, but the clinical outcome is quite variable due to the resistance encountered through upregulated expression of alternate ICs such as VISTA, LAG-3, TIGIT and TIM-3. This review focuses on the roles played by the traditional as well as alternate ICs and the contribution of associated signaling pathways in generating such resistance to immunotherapy. Combinatorial targeting of traditional and alternate ICs might be beneficial for immune-refractory tumors.
Collapse
Affiliation(s)
- Dharmindra Dulal
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614, USA; (D.D.); (A.B.); (A.K.T.)
| | - Andrew Boring
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614, USA; (D.D.); (A.B.); (A.K.T.)
| | - David Terrero
- Department of Pharmacology & Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo Main Campus, Toledo, OH 43614, USA
| | - Tiffany Johnson
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614, USA; (D.D.); (A.B.); (A.K.T.)
| | - Amit K. Tiwari
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614, USA; (D.D.); (A.B.); (A.K.T.)
- Department of Pharmacology & Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo Main Campus, Toledo, OH 43614, USA
| | - Dayanidhi Raman
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614, USA; (D.D.); (A.B.); (A.K.T.)
| |
Collapse
|
7
|
Matsumoto H, Fujita Y, Onizawa M, Saito K, Sumichika Y, Yoshida S, Temmoku J, Matsuoka N, Yashiro-Furuya M, Asano T, Sato S, Suzuki E, Machida T, Watanabe H, Migita K. Increased CEACAM1 expression on peripheral blood neutrophils in patients with rheumatoid arthritis. Front Immunol 2022; 13:978435. [PMID: 36591283 PMCID: PMC9794574 DOI: 10.3389/fimmu.2022.978435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Altered expression of adhesion molecules in immune cells has been demonstrated in rheumatoid arthritis (RA). Carcinoembryonic-antigen-related cell-adhesion molecule 1 (CEACAM1) is an adhesion molecule that acts as a coinhibitory receptor in the immune system. We investigated the role of CEACAM1 in immune cell subsets of patients with RA. Peripheral blood was obtained from 37 patients with RA and 20 healthy controls (HC). The expression of CEACAM1 and T-cell immunoglobulin mucin domain molecule (TIM) -3 on peripheral blood mononuclear cells and neutrophils was analyzed by flow cytometry. Intracellular TIM-3 expression was analyzed using cellular lysates by Western blot analysis. Serum levels of soluble CEACAM1 (sCEACAM1) were estimated by an enzyme-linked immunosorbent assay. CEACAM1 expression was not detected in peripheral blood mononuclear cells, including in CD14(+) monocytes and CD3(+) lymphocytes isolated from patients with RA or HC. However, substantial cell-surface expression of CEACAM1 was detected in peripheral blood neutrophils, and it was significantly elevated in samples from patients with RA without remission compared to those in remission. There was no significant difference in serum levels of sCEACAM1 between patients with RA and HC. Cell-surface expression of TIM-3 was not detected in peripheral blood neutrophils from patients with RA or HC but was seen in CD14(+) monocytes. However, there was no significant difference in TIM-3 expression on monocytes between patients with RA and HC. Our data indicate that cell-surface expression of CEACAM1 on peripheral blood neutrophils are higher in patients with RA and that it is associated with rheumatoid inflammation. Further studies are needed to explore the potential role of CEACAM1 in rheumatoid inflammatory pathways.
Collapse
Affiliation(s)
- Haruki Matsumoto
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Yuya Fujita
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Michio Onizawa
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Kenji Saito
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Yuya Sumichika
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Shuhei Yoshida
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Jumpei Temmoku
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Naoki Matsuoka
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Makiko Yashiro-Furuya
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Tomoyuki Asano
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Shuzo Sato
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Eiji Suzuki
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Takeshi Machida
- Department of Immunology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Hiroshi Watanabe
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
| | - Kiyoshi Migita
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan,*Correspondence: Kiyoshi Migita,
| |
Collapse
|
8
|
Gomes de Morais AL, Cerdá S, de Miguel M. New Checkpoint Inhibitors on the Road: Targeting TIM-3 in Solid Tumors. Curr Oncol Rep 2022; 24:651-658. [PMID: 35218498 DOI: 10.1007/s11912-022-01218-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2021] [Indexed: 12/28/2022]
Abstract
PURPOSE OF REVIEW Even though checkpoint inhibitors have become a recent milestone for the treatment of many different tumor types, eventually, most part of patients will develop resistance mechanisms and their disease will progress. New generations of checkpoint inhibitors, as the ones directed to TIM-3, are on research. RECENT FINDINGS TIM-3 expression has been associated with more advanced stages and shorter survival in several tumor types, due to its association with T-cell dysfunction, and has become an interesting target to explore. Early phase clinical trials with different anti-TIM-3 monoclonal antibodies have shown a safe toxicity profile, as cobolimab, LY3321367, or sabatolimab; however, the general antitumor activity remains to be determined and further investigations are needed. TIM-3 is implicated in resistance to immunotherapy due to its role in T cell exhaustion. However, the TIM-3 pathway is highly complex in terms of non-canonical signaling, broad expression by different immune cells and multiple ligands. Different anti-TIM-3 inhibitors are currently on research, either as monotherapy or in combination with other immunotherapies or chemotherapy, aiming to overcome resistance.
Collapse
Affiliation(s)
- Ana Luiza Gomes de Morais
- START Madrid-Fundación Jiménez Díaz (FJD) Early Phase Program, Fundación Jiménez Díaz Hospital, Madrid, Spain
| | - Sara Cerdá
- START Madrid-HM Centro Integral Oncológico Clara Campal (CIOCC) Early Phase Program, HM Sanchinarro University Hospital, Calle Oña, 10, 28050, Madrid, Spain
| | - Maria de Miguel
- START Madrid-HM Centro Integral Oncológico Clara Campal (CIOCC) Early Phase Program, HM Sanchinarro University Hospital, Calle Oña, 10, 28050, Madrid, Spain.
| |
Collapse
|
9
|
Immune Regulatory Processes of the Tumor Microenvironment under Malignant Conditions. Int J Mol Sci 2021; 22:ijms222413311. [PMID: 34948104 PMCID: PMC8706102 DOI: 10.3390/ijms222413311] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/03/2021] [Accepted: 12/05/2021] [Indexed: 02/07/2023] Open
Abstract
The tumor microenvironment (TME) is a critical regulator of tumor growth, progression, and metastasis. Since immune cells represent a large fraction of the TME, they play a key role in mediating pro- and anti-tumor immune responses. Immune escape, which suppresses anti-tumor immunity, enables tumor cells to maintain their proliferation and growth. Numerous mechanisms, which have been intensively studied in recent years, are involved in this process and based on these findings, novel immunotherapies have been successfully developed. Here, we review the composition of the TME and the mechanisms by which immune evasive processes are regulated. In detail, we describe membrane-bound and soluble factors, their regulation, and their impact on immune cell activation in the TME. Furthermore, we give an overview of the tumor/antigen presentation and how it is influenced under malignant conditions. Finally, we summarize novel TME-targeting agents, which are already in clinical trials for different tumor entities.
Collapse
|
10
|
Moon J, Oh YM, Ha SJ. Perspectives on immune checkpoint ligands: expression, regulation, and clinical implications. BMB Rep 2021. [PMID: 34078531 PMCID: PMC8411045 DOI: 10.5483/bmbrep.2021.54.8.054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In the tumor microenvironment, immune checkpoint ligands (ICLs) must be expressed in order to trigger the inhibitory signal via immune checkpoint receptors (ICRs). Although ICL expression frequently occurs in a manner intrinsic to tumor cells, extrinsic factors derived from the tumor microenvironment can fine-tune ICL expression by tumor cells or prompt non-tumor cells, including immune cells. Considering the extensive interaction between T cells and other immune cells within the tumor microenvironment, ICL expression on immune cells can be as significant as that of ICLs on tumor cells in promoting anti-tumor immune responses. Here, we introduce various regulators known to induce or suppress ICL expression in either tumor cells or immune cells, and concise mechanisms relevant to their induction. Finally, we focus on the clinical significance of understanding the mechanisms of ICLs for an optimized immunotherapy for individual cancer patients.
Collapse
Affiliation(s)
- Jihyun Moon
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul 03722, Korea
| | - Yoo Min Oh
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul 03722, Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
11
|
Gioseffi A, Edelmann MJ, Kima PE. Intravacuolar Pathogens Hijack Host Extracellular Vesicle Biogenesis to Secrete Virulence Factors. Front Immunol 2021; 12:662944. [PMID: 33959131 PMCID: PMC8093443 DOI: 10.3389/fimmu.2021.662944] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/30/2021] [Indexed: 12/18/2022] Open
Abstract
Extracellular vesicles (EVs) have garnered significant interest in recent years due to their contributions to cell-to-cell communication and disease processes. EVs are composed of a complex profile of bioactive molecules, which include lipids, nucleic acids, metabolites, and proteins. Although the biogenesis of EVs released by cells under various normal and abnormal conditions has been well-studied, there is incomplete knowledge about how infection influences EV biogenesis. EVs from infected cells contain specific molecules of both host and pathogen origin that may contribute to pathogenesis and the elicitation of the host immune response. Intracellular pathogens exhibit diverse lifestyles that undoubtedly dictate the mechanisms by which their molecules enter the cell’s exosome biogenesis schemes. We will discuss the current understanding of the mechanisms used during infection to traffic molecules from their vacuolar niche to host EVs by selected intravacuolar pathogens. We initially review general exosome biogenesis schemes and then discuss what is known about EV biogenesis in Mycobacterium, Plasmodium, Toxoplasma, and Leishmania infections, which are pathogens that reside within membrane delimited compartments in phagocytes at some time in their life cycle within mammalian hosts. The review includes discussion of the need for further studies into the biogenesis of EVs to better understand the contributions of these vesicles to host-pathogen interactions, and to uncover potential therapeutic targets to control these pathogens.
Collapse
Affiliation(s)
- Anna Gioseffi
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Mariola J Edelmann
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Peter E Kima
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| |
Collapse
|
12
|
Zeidan AM, Komrokji RS, Brunner AM. TIM-3 pathway dysregulation and targeting in cancer. Expert Rev Anticancer Ther 2021; 21:523-534. [PMID: 33334180 DOI: 10.1080/14737140.2021.1865814] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Dysfunction of the immune system is a hallmark of cancer. Through increased understanding of the complex interactions between immunity and cancer, immunotherapy has emerged as a treatment modality for different types of cancer. Promising activity with immunotherapy has been reported in numerous malignancies, but challenges such as limited response rates and treatment resistance remain. Furthermore, outcomes with this therapeutic approach in hematologic malignancies are even more limited than in solid tumors. T-cell immunoglobulin domain and mucin domain 3 (TIM-3) has emerged as a potential immune checkpoint target in both solid tumors and hematologic malignancies. TIM-3 has been shown to promote immune tolerance, and overexpression of TIM-3 is associated with more aggressive or advanced disease and poor prognosis. AREAS COVERED This review examines what is currently known regarding the biology of TIM-3 and clinical implications of targeting TIM-3 in cancer. Particular focus is given to myeloid malignancies. EXPERT OPINION The targeting of TIM-3 is a promising therapeutic approach in cancers, including hematologic cancers such as myeloid malignancies which have not benefited much from current immunotherapeutic treatment approaches. We anticipate that with further clinical evaluation, TIM-3 blockade will emerge as an important treatment strategy in myeloid malignancies.
Collapse
Affiliation(s)
- Amer M Zeidan
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT, USA
| | - Rami S Komrokji
- Malignant Hematology Department, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Andrew M Brunner
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Ding QQ, Chauvin JM, Zarour HM. Targeting novel inhibitory receptors in cancer immunotherapy. Semin Immunol 2020; 49:101436. [PMID: 33288379 DOI: 10.1016/j.smim.2020.101436] [Citation(s) in RCA: 7] [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: 10/07/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/24/2022]
Abstract
T cells play a critical role in promoting tumor regression in both experimental models and humans. Yet, T cells that are chronically exposed to tumor antigen during cancer progression can become dysfunctional/exhausted and fail to induce tumor destruction. Such tumor-induced T cell dysfunction may occur via multiple mechanisms. In particular, immune checkpoint inhibitory receptors that are upregulated by tumor-infiltrating lymphocytes in many cancers limit T cell survival and function. Overcoming this inhibitory receptor-mediated T cell dysfunction has been a central focus of recent developments in cancer immunotherapy. Immunotherapies targeting inhibitory receptor pathways such as programmed cell death 1 (PD-1)/programmed death ligand 1 and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), alone or in combination, confer significant clinical benefits in multiple tumor types. However, many patients with cancer do not respond to immune checkpoint blockade, and dual PD-1/CTLA-4 blockade may cause serious adverse events, which limits its indications. Targeting novel non-redundant inhibitory receptor pathways contributing to tumor-induced T cell dysfunction in the tumor microenvironment may prove efficacious and non-toxic. This review presents preclinical and clinical findings supporting the roles of two key pathways-T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) and T cell immunoreceptor with Ig and ITIM domain (TIGIT)/CD226/CD96/CD112R-in cancer immunotherapy.
Collapse
Affiliation(s)
- Quan-Quan Ding
- Department of Medicine and Division of Hematology/Oncology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Joe-Marc Chauvin
- Department of Medicine and Division of Hematology/Oncology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Hassane M Zarour
- Department of Medicine and Division of Hematology/Oncology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
14
|
Gioseffi A, Hamerly T, Van K, Zhang N, Dinglasan RR, Yates PA, Kima PE. Leishmania-infected macrophages release extracellular vesicles that can promote lesion development. Life Sci Alliance 2020; 3:3/12/e202000742. [PMID: 33122174 PMCID: PMC7652379 DOI: 10.26508/lsa.202000742] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 10/06/2020] [Accepted: 10/08/2020] [Indexed: 12/12/2022] Open
Abstract
Macrophages infected with Leishmania donovani release extracellular vesicles that are composed of parasite and host-derived molecules that have the potential to induce vascular changes in tissues. Leishmania donovani infection of macrophages results in quantitative and qualitative changes in the protein profile of extracellular vesicles (EVs) released by the infected host cells. We confirmed mass spectrometry results orthogonally by performing Western blots for several Leishmania-infected macrophage-enriched EVs (LieEVs) molecules. Several host cell proteins in LieEVs have been implicated in promoting vascular changes in other systems. We also identified 59 parasite-derived proteins in LieEVs, including a putative L. donovani homolog of mammalian vasohibins (LdVash), which in mammals promotes angiogenesis. We developed a transgenic parasite that expressed an endogenously tagged LdVash/mNeonGreen (mNG) and confirmed that LdVash/mNG is indeed expressed in infected macrophages and in LieEVs. We further observed that LieEVs induce endothelial cells to release angiogenesis promoting mediators including IL-8, G-CSF/CSF-3, and VEGF-A. In addition, LieEVs induce epithelial cell migration and tube formation by endothelial cells in surrogate angiogenesis assays. Taken together, these studies show that Leishmania infection alters the composition of EVs from infected cells and suggest that LieEVs may play a role in the promotion of vascularization of Leishmania infections.
Collapse
Affiliation(s)
- Anna Gioseffi
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, USA
| | - Tim Hamerly
- Emerging Pathogens Institute and Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL, USA
| | - Kha Van
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, USA
| | - Naixin Zhang
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, USA
| | - Rhoel R Dinglasan
- Emerging Pathogens Institute and Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL, USA
| | - Phillip A Yates
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, OR, USA
| | - Peter E Kima
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, USA
| |
Collapse
|
15
|
Acharya N, Sabatos-Peyton C, Anderson AC. Tim-3 finds its place in the cancer immunotherapy landscape. J Immunother Cancer 2020; 8:e000911. [PMID: 32601081 PMCID: PMC7326247 DOI: 10.1136/jitc-2020-000911] [Citation(s) in RCA: 233] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2020] [Indexed: 02/06/2023] Open
Abstract
The blockade of immune checkpoint receptors has made great strides in the treatment of major cancers, including melanoma, Hodgkin's lymphoma, renal, and lung cancer. However, the success rate of immune checkpoint blockade is still low and some cancers, such as microsatellite-stable colorectal cancer, remain refractory to these treatments. This has prompted investigation into additional checkpoint receptors. T-cell immunoglobulin and mucin domain 3 (Tim-3) is a checkpoint receptor expressed by a wide variety of immune cells as well as leukemic stem cells. Coblockade of Tim-3 and PD-1 can result in reduced tumor progression in preclinical models and can improve antitumor T-cell responses in cancer patients. In this review, we will discuss the basic biology of Tim-3, its role in the tumor microenvironment, and the emerging clinical trial data that point to its future application in the field of immune-oncology.
Collapse
Affiliation(s)
- Nandini Acharya
- Department of Neurology, Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Catherine Sabatos-Peyton
- Exploratory Immuno-oncology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | - Ana Carrizosa Anderson
- Department of Neurology, Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
16
|
Abstract
T cell immunoglobulin and mucin domain-containing protein 3 (TIM3), a member of the TIM family, was originally identified as a receptor expressed on interferon-γ-producing CD4+ and CD8+ T cells. Initial data indicated that TIM3 functioned as a 'co-inhibitory' or 'checkpoint' receptor, but due to the lack of a definable inhibitory signalling motif, it was also suggested that TIM3 might act as a co-stimulatory receptor. Recent studies have shown that TIM3 is part of a module that contains multiple co-inhibitory receptors (checkpoint receptors), which are co-expressed and co-regulated on dysfunctional or 'exhausted' T cells in chronic viral infections and cancer. Furthermore, co-blockade of TIM3 and programmed cell death 1 (PD1) can result in tumour regression in preclinical models and can improve anticancer T cell responses in patients with advanced cancers. Here, we highlight the developments in understanding TIM3 biology, including novel ligand identification and the discovery of loss-of-function mutations associated with human disease. In addition, we summarize emerging data from human clinical trials showing that TIM3 indeed acts as a 'checkpoint' receptor and that inhibition of TIM3 enhances the antitumour effect of PD1 blockade.
Collapse
|
17
|
Leishmania Infection Induces Macrophage Vascular Endothelial Growth Factor A Production in an ARNT/HIF-Dependent Manner. Infect Immun 2019; 87:IAI.00088-19. [PMID: 31451620 PMCID: PMC6803331 DOI: 10.1128/iai.00088-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 08/12/2019] [Indexed: 12/22/2022] Open
Abstract
Cutaneous leishmaniasis is characterized by vascular remodeling. Following infection with Leishmania parasites, the vascular endothelial growth factor A (VEGF-A)/VEGF receptor 2 (VEGFR-2) signaling pathway mediates lymphangiogenesis, which is critical for lesion resolution. Therefore, we investigated the cellular and molecular mediators involved in VEGF-A/VEGFR-2 signaling using a murine model of infection. We found that macrophages are the predominant cell type expressing VEGF-A during Leishmania major infection. Given that Leishmania parasites activate hypoxia-inducible factor 1α (HIF-1α) and this transcription factor can drive VEGF-A expression, we analyzed the expression of HIF-1α during infection. We showed that macrophages were also the major cell type expressing HIF-1α during infection and that infection-induced VEGF-A production is mediated by ARNT/HIF activation. Furthermore, mice deficient in myeloid ARNT/HIF signaling exhibited larger lesions without differences in parasite numbers. These data show that L. major infection induces macrophage VEGF-A production in an ARNT/HIF-dependent manner and suggest that ARNT/HIF signaling may limit inflammation by promoting VEGF-A production and, thus, lymphangiogenesis during infection.
Collapse
|
18
|
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: 57] [Impact Index Per Article: 11.4] [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
|
19
|
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: 3.8] [Reference Citation Analysis] [Abstract] [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
|
20
|
Schatz V, Neubert P, Rieger F, Jantsch J. Hypoxia, Hypoxia-Inducible Factor-1α, and Innate Antileishmanial Immune Responses. Front Immunol 2018. [PMID: 29520262 PMCID: PMC5827161 DOI: 10.3389/fimmu.2018.00216] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Low oxygen environments and accumulation of hypoxia-inducible factors (HIFs) are features of infected and inflamed tissues. Here, we summarize our current knowledge on oxygen levels found in Leishmania-infected tissues and discuss which mechanisms potentially contribute to local tissue oxygenation in leishmanial lesions. Moreover, we review the role of hypoxia and HIF-1 on innate antileishmanial immune responses.
Collapse
Affiliation(s)
- Valentin Schatz
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg, University of Regensburg, Regensburg, Germany
| | - Patrick Neubert
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg, University of Regensburg, Regensburg, Germany
| | - Franz Rieger
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg, University of Regensburg, Regensburg, Germany
| | - Jonathan Jantsch
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg, University of Regensburg, Regensburg, Germany
| |
Collapse
|
21
|
Pober JS, Merola J, Liu R, Manes TD. Antigen Presentation by Vascular Cells. Front Immunol 2017; 8:1907. [PMID: 29312357 PMCID: PMC5744398 DOI: 10.3389/fimmu.2017.01907] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/14/2017] [Indexed: 01/21/2023] Open
Abstract
Antigen presentation by cells of the vessel wall may initiate rapid and localized memory immune responses in peripheral tissues. Peptide antigens displayed on major histocompatibility complex (MHC) molecules on the surface of endothelial cells (ECs) can be recognized by T cell receptors on circulating effector memory T cells (TEM), triggering both transendothelial migration and activation. The array of co-stimulatory receptors, adhesion molecules, and cytokines expressed by ECs serves to modulate T cell activation responses. While the effects of these interactions vary among species, vascular beds, and vascular segments within the same tissue, they are capable of triggering allograft rejection without direct involvement of professional antigen-presenting cells and may play a similar role in host defense against infections and in autoimmunity. Once across the endothelium, extravasating TEM then contact mural cells of the vessel wall, including pericytes or vascular smooth muscle cells, which may also present antigens and provide signals that further regulate T cell responses. Collectively, these interactions provide an unexplored opportunity in which targeting of vascular cells can be used to modulate immune responses. In organ transplantation, targeting ECs with siRNA to reduce expression of MHC molecules may additionally mitigate perioperative injuries by preformed alloantibodies, further reducing the risk of graft rejection. Similarly, genetic manipulation of vascular cells to minimize antigen-dependent responses can be used to increase perfusion of tissue engineered organs without triggering rejection.
Collapse
Affiliation(s)
- Jordan S Pober
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
| | - Jonathan Merola
- Department of Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Rebecca Liu
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
| | - Thomas D Manes
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
22
|
Arabzadeh A, McGregor K, Breton V, Van Der Kraak L, Akavia UD, Greenwood CMT, Beauchemin N. EphA2 signaling is impacted by carcinoembryonic antigen cell adhesion molecule 1-L expression in colorectal cancer liver metastasis in a cell context-dependent manner. Oncotarget 2017; 8:104330-104346. [PMID: 29262644 PMCID: PMC5732810 DOI: 10.18632/oncotarget.22236] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 10/05/2017] [Indexed: 12/12/2022] Open
Abstract
We have shown that carcinoembryonic antigen cell adhesion molecule 1 long isoform (CEACAM1-L) expression in MC38 metastatic colorectal cancer (CRC) cells results in liver metastasis inhibition via CCL2 and STAT3 signaling. But other molecular mechanisms orchestrating CEACAM1-L-mediated metastasis inhibition remain to be defined. We screened a panel of mouse and human CRC cells and evaluated their metastatic outcome after CEACAM1 overexpression or downregulation. An unbiased transcript profiling and a phospho-receptor tyrosine kinase screen comparing MC38 CEACAM1-L-expressing and non-expressing (CT) CRC cells revealed reduced ephrin type-A receptor 2 (EPHA2) expression and activity. An EPHA2-specific inhibitor reduced EPHA2 downstream signaling in CT cells similar to that in CEACAM1-L cells with decreased proliferation and migration. Human CRC patients exhibiting high CEACAM1 in combination with low EPHA2 expression benefited from longer time to first recurrence/metastasis compared to those with high EPHA2 expression. With the added interaction of CEACAM6, we denoted that CEACAM1 high- and EPHA2 low-expressing patient samples with lower CEACAM6 expression also exhibited a longer time to first recurrence/metastasis. In HT29 human CRC cells, down-regulation of CEACAM1 along with CEA and CEACAM6 up-regulation led to higher metastatic burden. Overall, CEACAM1-L expression in poorly differentiated CRC can inhibit liver metastasis through cell context-dependent EPHA2-mediated signaling. However, CEACAM1’s role should be considered in the presence of other CEACAM family members.
Collapse
Affiliation(s)
- Azadeh Arabzadeh
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | - Kevin McGregor
- Department of Epidemiology, Biostatistics & Occupational Health, McGill University, Montreal, QC, Canada
| | - Valérie Breton
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | - Lauren Van Der Kraak
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Uri David Akavia
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Celia M T Greenwood
- Department of Epidemiology, Biostatistics & Occupational Health, McGill University, Montreal, QC, Canada.,Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada.,Departments of Oncology and Human Genetics, McGill University, Montreal, QC, Canada
| | - Nicole Beauchemin
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada.,Departments of Medicine and Oncology, McGill University, Montreal, QC, Canada
| |
Collapse
|
23
|
Dankner M, Gray-Owen SD, Huang YH, Blumberg RS, Beauchemin N. CEACAM1 as a multi-purpose target for cancer immunotherapy. Oncoimmunology 2017; 6:e1328336. [PMID: 28811966 PMCID: PMC5543821 DOI: 10.1080/2162402x.2017.1328336] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/03/2017] [Accepted: 05/05/2017] [Indexed: 02/06/2023] Open
Abstract
CEACAM1 is an extensively studied cell surface molecule with established functions in multiple cancer types, as well as in various compartments of the immune system. Due to its multi-faceted role as a recently appreciated immune checkpoint inhibitor and tumor marker, CEACAM1 is an attractive target for cancer immunotherapy. Herein, we highlight CEACAM1's function in various immune compartments and cancer types, including in the context of metastatic disease. This review outlines CEACAM1's role as a therapeutic target for cancer treatment in light of these properties.
Collapse
Affiliation(s)
- Matthew Dankner
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | - Scott D Gray-Owen
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Yu-Hwa Huang
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Richard S Blumberg
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicole Beauchemin
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| |
Collapse
|
24
|
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: 2.0] [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
|
25
|
Solano ME, Kowal MK, O'Rourke GE, Horst AK, Modest K, Plösch T, Barikbin R, Remus CC, Berger RG, Jago C, Ho H, Sass G, Parker VJ, Lydon JP, DeMayo FJ, Hecher K, Karimi K, Arck PC. Progesterone and HMOX-1 promote fetal growth by CD8+ T cell modulation. J Clin Invest 2015; 125:1726-38. [PMID: 25774501 DOI: 10.1172/jci68140] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/29/2015] [Indexed: 12/20/2022] Open
Abstract
Intrauterine growth restriction (IUGR) affects up to 10% of pregnancies in Western societies. IUGR is a strong predictor of reduced short-term neonatal survival and impairs long-term health in children. Placental insufficiency is often associated with IUGR; however, the molecular mechanisms involved in the pathogenesis of placental insufficiency and IUGR are largely unknown. Here, we developed a mouse model of fetal-growth restriction and placental insufficiency that is induced by a midgestational stress challenge. Compared with control animals, pregnant dams subjected to gestational stress exhibited reduced progesterone levels and placental heme oxygenase 1 (Hmox1) expression and increased methylation at distinct regions of the placental Hmox1 promoter. These stress-triggered changes were accompanied by an altered CD8+ T cell response, as evidenced by a reduction of tolerogenic CD8+CD122+ T cells and an increase of cytotoxic CD8+ T cells. Using progesterone receptor- or Hmox1-deficient mice, we identified progesterone as an upstream modulator of placental Hmox1 expression. Supplementation of progesterone or depletion of CD8+ T cells revealed that progesterone suppresses CD8+ T cell cytotoxicity, whereas the generation of CD8+CD122+ T cells is supported by Hmox1 and ameliorates fetal-growth restriction in Hmox1 deficiency. These observations in mice could promote the identification of pregnancies at risk for IUGR and the generation of clinical interventional strategies.
Collapse
|
26
|
Khairnar V, Duhan V, Maney SK, Honke N, Shaabani N, Pandyra AA, Seifert M, Pozdeev V, Xu HC, Sharma P, Baldin F, Marquardsen F, Merches K, Lang E, Kirschning C, Westendorf AM, Häussinger D, Lang F, Dittmer U, Küppers R, Recher M, Hardt C, Scheffrahn I, Beauchemin N, Göthert JR, Singer BB, Lang PA, Lang KS. CEACAM1 induces B-cell survival and is essential for protective antiviral antibody production. Nat Commun 2015; 6:6217. [PMID: 25692415 PMCID: PMC4346637 DOI: 10.1038/ncomms7217] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 01/07/2015] [Indexed: 01/03/2023] Open
Abstract
B cells are essential for antiviral immune defence because they produce neutralizing antibodies, present antigen and maintain the lymphoid architecture. Here we show that intrinsic signalling of CEACAM1 is essential for generating efficient B-cell responses. Although CEACAM1 exerts limited influence on the proliferation of B cells, expression of CEACAM1 induces survival of proliferating B cells via the BTK/Syk/NF-κB-axis. The absence of this signalling cascade in naive Ceacam1−/− mice limits the survival of B cells. During systemic infection with cytopathic vesicular stomatitis virus, Ceacam1−/− mice can barely induce neutralizing antibody responses and die early after infection. We find, therefore, that CEACAM1 is a crucial regulator of B-cell survival, influencing B-cell numbers and protective antiviral antibody responses. Antibody responses are regulated by selective survival of B cells with proper antigen specificity. Here the authors show that CEACAM1 is critical for B-cell survival during homeostasis and antiviral responses.
Collapse
Affiliation(s)
- Vishal Khairnar
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Vikas Duhan
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Sathish Kumar Maney
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany
| | - Nadine Honke
- 1] Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany [2] Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany
| | - Namir Shaabani
- 1] Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany [2] Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany
| | - Aleksandra A Pandyra
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Marc Seifert
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Virchowstrasse 173, Essen 45122, Germany
| | - Vitaly Pozdeev
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany
| | - Haifeng C Xu
- 1] Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany [2] Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany
| | - Piyush Sharma
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Fabian Baldin
- Clinic for Primary Immunodeficiency, Medical Outpatient Unit and Immunodeficiency Laboratory, Department of Biomedicine, University Hospital, Basel 4031, Switzerland
| | - Florian Marquardsen
- Clinic for Primary Immunodeficiency, Medical Outpatient Unit and Immunodeficiency Laboratory, Department of Biomedicine, University Hospital, Basel 4031, Switzerland
| | - Katja Merches
- 1] Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany [2] Department of Physiology I, University of Tuebingen, Gmelinstrasse 5, Tuebingen 72076, Germany
| | - Elisabeth Lang
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany
| | - Carsten Kirschning
- Institute of Medical Microbiology, Faculty of Medicine, University Hospital Essen, Hufelandstrasse 55, Essen 45122, Germany
| | - Astrid M Westendorf
- Institute of Medical Microbiology, Faculty of Medicine, University Hospital Essen, Hufelandstrasse 55, Essen 45122, Germany
| | - Dieter Häussinger
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany
| | - Florian Lang
- Department of Physiology I, University of Tuebingen, Gmelinstrasse 5, Tuebingen 72076, Germany
| | - Ulf Dittmer
- Institute of Virology, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Ralf Küppers
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Virchowstrasse 173, Essen 45122, Germany
| | - Mike Recher
- Clinic for Primary Immunodeficiency, Medical Outpatient Unit and Immunodeficiency Laboratory, Department of Biomedicine, University Hospital, Basel 4031, Switzerland
| | - Cornelia Hardt
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Inka Scheffrahn
- Clinic of Gastroenterology and Hepatology, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Nicole Beauchemin
- Rosalind and Morris Goodman Cancer Centre, Departments of Biochemistry, Medicine and Oncology, McIntyre Medical Science Building, Montreal, Quebec, Canada H3G 1Y6
| | - Joachim R Göthert
- Department of Hematology, West German Cancer Center (WTZ), University Hospital Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Bernhard B Singer
- Institute of Anatomy, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany
| | - Philipp A Lang
- 1] Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany [2] Department of Molecular Medicine II, Heinrich Heine University Düsseldorf, Universitätsstrasse 1, Düsseldorf 40225, Germany
| | - Karl S Lang
- 1] Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, Essen 45147, Germany [2] Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, Düsseldorf 40225, Germany
| |
Collapse
|
27
|
Kwiatkowski M, Wurlitzer M, Omidi M, Ren L, Kruber S, Nimer R, Robertson WD, Horst A, Miller RJD, Schlüter H. Desorption durch impulsive Anregung intramolekularer Vibrationszustände – eine Methode zur schnellen Extraktion von Proteinen aus intakten Geweben. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201407669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Marcel Kwiatkowski
- Universitätsklinikum Hamburg‐Eppendorf, Institut für Klinische Chemie, Martinistraße 52, Hamburg, 20246 (Deutschland)
| | - Marcus Wurlitzer
- Universitätsklinikum Hamburg‐Eppendorf, Institut für Klinische Chemie, Martinistraße 52, Hamburg, 20246 (Deutschland)
| | - Maryam Omidi
- Universitätsklinikum Hamburg‐Eppendorf, Institut für Klinische Chemie, Martinistraße 52, Hamburg, 20246 (Deutschland)
| | - Ling Ren
- Max‐Planck‐Institut für Struktur und Dynamik der Materie, CFEL (Gebäude 99), Luruper Chaussee 149, Hamburg, 22761 (Deutschland)
| | - Sebastian Kruber
- Max‐Planck‐Institut für Struktur und Dynamik der Materie, CFEL (Gebäude 99), Luruper Chaussee 149, Hamburg, 22761 (Deutschland)
| | - Refat Nimer
- Universitätsklinikum Hamburg‐Eppendorf, Institut für Klinische Chemie, Martinistraße 52, Hamburg, 20246 (Deutschland)
| | - Wesley D. Robertson
- Max‐Planck‐Institut für Struktur und Dynamik der Materie, CFEL (Gebäude 99), Luruper Chaussee 149, Hamburg, 22761 (Deutschland)
| | - Andrea Horst
- Universitätsklinikum Hamburg‐Eppendorf, Institut für Klinische Chemie, Martinistraße 52, Hamburg, 20246 (Deutschland)
| | - R. J. Dwayne Miller
- Max‐Planck‐Institut für Struktur und Dynamik der Materie, CFEL (Gebäude 99), Luruper Chaussee 149, Hamburg, 22761 (Deutschland)
| | - Hartmut Schlüter
- Universitätsklinikum Hamburg‐Eppendorf, Institut für Klinische Chemie, Martinistraße 52, Hamburg, 20246 (Deutschland)
| |
Collapse
|
28
|
Heckt T, Bickert T, Jeschke A, Seitz S, Schulze J, Ito WD, Zimmermann W, Amling M, Schinke T, Horst AK, Keller J. Increased osteoclastogenesis in mice lacking the carcinoembryonic antigen-related cell adhesion molecule 1. PLoS One 2014; 9:e114360. [PMID: 25490771 PMCID: PMC4260834 DOI: 10.1371/journal.pone.0114360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 11/06/2014] [Indexed: 02/02/2023] Open
Abstract
Alterations in bone remodeling are a major public health issue, as therapeutic options for widespread bone disorders such as osteoporosis and tumor-induced osteolysis are still limited. Therefore, a detailed understanding of the regulatory mechanism governing bone cell differentiation in health and disease are of utmost clinical importance. Here we report a novel function of carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), a member of the immunoglobulin superfamily involved in inflammation and tumorigenesis, in the physiologic regulation of bone remodeling. Assessing the expression of all members of the murine Ceacam family in bone tissue and marrow, we found CEACAM1 and CEACAM10 to be differentially expressed in both bone-forming osteoblasts and bone-resorbing osteoclasts. While Ceacam10-deficient mice displayed no alteration in structural bone parameters, static histomorphometry demonstrated a reduced trabecular bone mass in mice lacking CEACAM1. Furthermore, cellular and dynamic histomorphometry revealed an increased osteoclast formation in Ceacam1-deficient mice, while osteoblast parameters and the bone formation rate remained unchanged. In line with these findings, we detected accelerated osteoclastogenesis in Ceacam1-deficient bone marrow cells, while osteoblast differentiation, as determined by mineralization and alkaline phosphatase assays, was not affected. Therefore, our results provide in vivo and in vitro evidence for a physiologic role of CEACAM1 in the regulation of osteoclastogenesis.
Collapse
Affiliation(s)
- Timo Heckt
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Thomas Bickert
- Institute of Clinical Chemistry, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Anke Jeschke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Sebastian Seitz
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Jochen Schulze
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Wulf D. Ito
- Cardiovascular Center Oberallgäu-Kempten, Im Stillen 3, Immenstadt 87509, Germany
| | - Wolfgang Zimmermann
- Tumor Immunology Laboratory, LIFE-Center, Klinikum Grosshadern, Ludwig-Maximilians-University Munich, Marchionistraße 15, Munich 81377, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Andrea Kristina Horst
- Institute of Clinical Chemistry, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Johannes Keller
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| |
Collapse
|
29
|
Kwiatkowski M, Wurlitzer M, Omidi M, Ren L, Kruber S, Nimer R, Robertson WD, Horst A, Miller RJD, Schlüter H. Ultrafast Extraction of Proteins from Tissues Using Desorption by Impulsive Vibrational Excitation. Angew Chem Int Ed Engl 2014; 54:285-8. [DOI: 10.1002/anie.201407669] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Indexed: 01/07/2023]
Affiliation(s)
- Marcel Kwiatkowski
- University Medical Center Hamburg‐Eppendorf, Department of Clinical Chemistry, Martinistrasse 52, Hamburg, 20246 (Germany)
| | - Marcus Wurlitzer
- University Medical Center Hamburg‐Eppendorf, Department of Clinical Chemistry, Martinistrasse 52, Hamburg, 20246 (Germany)
| | - Maryam Omidi
- University Medical Center Hamburg‐Eppendorf, Department of Clinical Chemistry, Martinistrasse 52, Hamburg, 20246 (Germany)
| | - Ling Ren
- Max Planck Institute for the Structure and Dynamics of Matter, CFEL (Building 99), Luruper Chaussee 149, 22761 Hamburg (Germany)
| | - Sebastian Kruber
- Max Planck Institute for the Structure and Dynamics of Matter, CFEL (Building 99), Luruper Chaussee 149, 22761 Hamburg (Germany)
| | - Refat Nimer
- University Medical Center Hamburg‐Eppendorf, Department of Clinical Chemistry, Martinistrasse 52, Hamburg, 20246 (Germany)
| | - Wesley D. Robertson
- Max Planck Institute for the Structure and Dynamics of Matter, CFEL (Building 99), Luruper Chaussee 149, 22761 Hamburg (Germany)
| | - Andrea Horst
- University Medical Center Hamburg‐Eppendorf, Department of Clinical Chemistry, Martinistrasse 52, Hamburg, 20246 (Germany)
| | - R. J. Dwayne Miller
- Max Planck Institute for the Structure and Dynamics of Matter, CFEL (Building 99), Luruper Chaussee 149, 22761 Hamburg (Germany)
| | - Hartmut Schlüter
- University Medical Center Hamburg‐Eppendorf, Department of Clinical Chemistry, Martinistrasse 52, Hamburg, 20246 (Germany)
| |
Collapse
|
30
|
Schmid M, Zimara N, Wege AK, Ritter U. Myeloid-derived suppressor cell functionality and interaction withLeishmania majorparasites differ in C57BL/6 and BALB/c mice. Eur J Immunol 2014; 44:3295-306. [DOI: 10.1002/eji.201344335] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 06/27/2014] [Accepted: 08/13/2014] [Indexed: 01/20/2023]
Affiliation(s)
- Maximilian Schmid
- Institute of Immunology; University of Regensburg; Regensburg Germany
| | - Nicole Zimara
- Institute of Immunology; University of Regensburg; Regensburg Germany
| | - Anja Kathrin Wege
- Department of Gynecology and Obstetrics; University Medical Center Regensburg; Regensburg Germany
| | - Uwe Ritter
- Institute of Immunology; University of Regensburg; Regensburg Germany
| |
Collapse
|
31
|
Xie Q, Brackenbury LS, Hill DJ, Williams NA, Qu X, Virji M. Moraxella catarrhalis adhesin UspA1-derived recombinant fragment rD-7 induces monocyte differentiation to CD14+CD206+ phenotype. PLoS One 2014; 9:e90999. [PMID: 24599281 PMCID: PMC3944954 DOI: 10.1371/journal.pone.0090999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 02/06/2014] [Indexed: 12/04/2022] Open
Abstract
Circulating monocytes in the bloodstream typically migrate to other tissues and differentiate into tissue resident macrophages, the process being determined by the constituents of the microenvironments encountered. These may include microbes and their products. In this study, we investigated whether Moraxella catarrhalis Ubiquitous Surface Protein A1 (UspA1), known to bind to a widely expressed human cell surface receptor CEACAM1, influences monocyte differentiation as receptor engagement has been shown to have profound effects on monocytes. We used the recombinant molecules corresponding to the regions of UspA1 which either bind (rD-7; UspA1527–665) or do not bind (r6–8; UspA1659–863) to CEACAM1 and investigated their effects on CD206, CD80 and CD86 expression on freshly isolated human CD14+ monocytes from peripheral blood mononuclear cells (PBMC). Exposure to rD-7, but not r6–8, biased monocyte differentiation towards a CD14+CD206+ phenotype, with reduced CD80 expression. Monocytes treated with rD-7 also secreted high levels of IL-1ra and chemokine IL-8 but not IL-10 or IL-12p70. The effects of rD-7 were independent of any residual endotoxin. Unexpectedly, these effects of rD-7 were also independent of its ability to bind to CEACAM1, as monocyte pre-treatment with the anti-CEACAM antibody A0115 known to inhibit rD-7 binding to the receptor, did not affect rD-7-driven differentiation. Further, another control protein rD-7/D (a mutant form of rD-7, known not to bind to CEACAMs), also behaved as the parent molecule. Our data suggest that specific regions of M. catarrhalis adhesin UspA1 may modulate inflammation during infection through a yet unknown receptor on monocytes.
Collapse
Affiliation(s)
- Qi Xie
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Louise S. Brackenbury
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Darryl J. Hill
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Neil A. Williams
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
- * E-mail: (XQ); (NW)
| | - Xun Qu
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
- * E-mail: (XQ); (NW)
| | - Mumtaz Virji
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
32
|
Han EC, Lee J, Ryu SW, Choi C. Tumor-conditioned Gr-1(+)CD11b(+) myeloid cells induce angiogenesis through the synergistic action of CCL2 and CXCL16 in vitro. Biochem Biophys Res Commun 2014; 443:1218-25. [PMID: 24388986 DOI: 10.1016/j.bbrc.2013.12.117] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 12/22/2013] [Indexed: 12/12/2022]
Abstract
Gr-1(+)CD11b(+) cells can suppress innate and adaptive immunity, and the functional immunosuppressive characteristics of these cells can be modulated by the tumor microenvironment. Since Gr-1(+)CD11(+) cells are also involved in tumor-associated angiogenesis, we hypothesized that the angiogenic nature of Gr-1(+)CD11b(+) cells could be regulated by the tumor milieu. To address this hypothesis, we imitated a tumor microenvironment by exposing Gr-1(+)CD11b(+) cells isolated from spleen of 4T1 mammary carcinoma-bearing mice to tumor-conditioned medium. Supernatants from tumor-conditioned Gr-1(+)CD11b(+) cells significantly induced capillary-like tube formation and migration of human umbilical vein endothelial cells (HUVECs) compared to naive Gr-1(+)CD11b(+) cells. Incubation of Gr-1(+)CD11b(+) cells with tumor-conditioned medium induced production of pro-angiogenic chemokines CCL2 and CXCL16. Pretreatment with an anti-CCL2 antibody, but not an anti-CXCL16 antibody, suppressed the angiogenic effects of tumor-conditioned Gr-1(+)CD11b(+) cells on HUVECs. Simultaneous neutralization of CCL2 and CXCL16 significantly inhibited tube formation and migration of HUVECs compared to the sole neutralization against CCL2. Supernatants from tumor-conditioned Gr-1(+)CD11b(+) cells induced phosphorylation of ERK1/2 in HUVECs, and inhibition of the ERK pathway blocked angiogenic effects. ERK pathway activity was partially abrogated by neutralization of CCL2 and more suppressed by simultaneous neutralization of CCL2 and CXCL16. These results collectively indicate that CCL2 and CXCL16 chemokines produced by tumor-conditioned Gr-1(+)CD11b(+) myeloid cells synergistically induce angiogenesis in vitro by stimulating the ERK1/2 signaling pathway. Thus, regulation of Gr-1(+)CD11b(+) cells in the tumor microenvironment may contribute to angiogenesis through the secretion of pro-angiogenic chemokines.
Collapse
Affiliation(s)
- Eun Chun Han
- Graduate School of Medical Science and Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 305-701, Republic of Korea
| | - Jungwhoi Lee
- Department of Bio and Brain Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 305-701, Republic of Korea
| | - Seung-Wook Ryu
- Department of Bio and Brain Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 305-701, Republic of Korea; KI for the BioCentury, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 305-701, Republic of Korea
| | - Chulhee Choi
- Graduate School of Medical Science and Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 305-701, Republic of Korea; Department of Bio and Brain Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 305-701, Republic of Korea; KI for the BioCentury, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 305-701, Republic of Korea.
| |
Collapse
|
33
|
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: 26.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
34
|
Ludewig P, Sedlacik J, Gelderblom M, Bernreuther C, Korkusuz Y, Wagener C, Gerloff C, Fiehler J, Magnus T, Horst AK. Carcinoembryonic antigen-related cell adhesion molecule 1 inhibits MMP-9-mediated blood-brain-barrier breakdown in a mouse model for ischemic stroke. Circ Res 2013; 113:1013-22. [PMID: 23780386 DOI: 10.1161/circresaha.113.301207] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
RATIONALE Blood-brain-barrier (BBB) breakdown and cerebral edema result from postischemic inflammation and contribute to mortality and morbidity after ischemic stroke. A functional role for the carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) in the regulation of reperfusion injury has not yet been demonstrated. OBJECTIVE We sought to identify and characterize the relevance of CEACAM1-expressing inflammatory cells in BBB breakdown and outcome after ischemic stroke in Ceacam1(-/-) and wild-type mice. METHODS AND RESULTS Focal ischemia was induced by temporary occlusion of the middle cerebral artery with a microfilament. Using MRI and Evans blue permeability assays, we observed increased stroke volumes, BBB breakdown and edema formation, reduction of cerebral perfusion, and brain atrophy in Ceacam1(-/-) mice. This translated into poor performance in neurological scoring and high poststroke-associated mortality. Elevated neutrophil influx, hyperproduction, and release of neutrophil-related matrix metalloproteinase-9 in Ceacam1(-/-) mice were confirmed by immune fluorescence, flow cytometry, zymography, and stimulation of neutrophils. Importantly, neutralization of matrix metalloproteinase-9 activity in Ceacam1(-/-) mice was sufficient to alleviate stroke sizes and improve survival to the level of CEACAM1-competent animals. Immune histochemistry of murine and human poststroke autoptic brains congruently identified abundance of CEACAM1(+)matrix metalloproteinase-9(+) neutrophils in the ischemic hemispheres. CONCLUSIONS CEACAM1 controls matrix metalloproteinase-9 secretion by neutrophils in postischemic inflammation at the BBB after stroke. We propose CEACAM1 as an important inhibitory regulator of neutrophil-mediated tissue damage and BBB breakdown in focal cerebral ischemia.
Collapse
Affiliation(s)
- Peter Ludewig
- From the Institute of Clinical Chemistry, Department of Neurology, Department of Neuropathology, and Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Feduska JM, Garcia PL, Brennan SB, Bu S, Council LN, Yoon KJ. N-glycosylation of ICAM-2 is required for ICAM-2-mediated complete suppression of metastatic potential of SK-N-AS neuroblastoma cells. BMC Cancer 2013; 13:261. [PMID: 23714211 PMCID: PMC3700849 DOI: 10.1186/1471-2407-13-261] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 05/22/2013] [Indexed: 11/12/2022] Open
Abstract
Background Cell adhesion molecules (CAMs) are expressed ubiquitously. Each of the four families of CAMs is comprised of glycosylated, membrane-bound proteins that participate in multiple cellular processes including cell-cell communication, cell motility, inside-out and outside-in signaling, tumorigenesis, angiogenesis and metastasis. Intercellular adhesion molecule-2 (ICAM-2), a member of the immunoglobulin superfamily of CAMs, has six N-linked glycosylation sites at amino acids (asparagines) 47, 82, 105, 153, 178 and 187. Recently, we demonstrated a previously unknown function for ICAM-2 in tumor cells. We showed that ICAM-2 suppressed neuroblastoma cell motility and growth in soft agar, and induced a juxtamembrane distribution of F-actin in vitro. We also showed that ICAM-2 completely suppressed development of disseminated tumors in vivo in a murine model of metastatic NB. These effects of ICAM-2 on NB cell phenotype in vitro and in vivo depended on the interaction of ICAM-2 with the cytoskeletal linker protein α-actinin. Interestingly, ICAM-2 did not suppress subcutaneous growth of tumors in mice, suggesting that ICAM-2 affects the metastatic but not the tumorigenic potential of NB cells. The goal of the study presented here was to determine if the glycosylation status of ICAM-2 influenced its function in neuroblastoma cells. Methods Because it is well documented that glycosylation facilitates essential steps in tumor progression and metastasis, we investigated whether the glycosylation status of ICAM-2 affected the phenotype of NB cells. We used site-directed mutagenesis to express hypo- or non-glycosylated variants of ICAM-2, by substituting alanine for asparagine at glycosylation sites, and compared the impact of each variant on NB cell motility, anchorage-independent growth, interaction with intracellular proteins, effect on F-actin distribution and metastatic potential in vivo. Results The in vitro and in vivo phenotypes of cells expressing glycosylation site variants differed from cells expressing fully-glycosylated ICAM-2 or no ICAM-2. Most striking was the finding that mice injected intravenously with NB cells expressing glycosylation site variants survived longer (P ≤ 0.002) than mice receiving SK-N-AS cells with undetectable ICAM-2. However, unlike fully-glycosylated ICAM-2, glycosylation site variants did not completely suppress disseminated tumor development. Conclusions Reduced glycosylation of ICAM-2 significantly attenuated, but did not abolish, its ability to suppress metastatic properties of NB cells.
Collapse
Affiliation(s)
- Joseph M Feduska
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | | | |
Collapse
|
36
|
Reichmann J, Reddington JP, Best D, Read D, Öllinger R, Meehan RR, Adams IR. The genome-defence gene Tex19.1 suppresses LINE-1 retrotransposons in the placenta and prevents intra-uterine growth retardation in mice. Hum Mol Genet 2013; 22:1791-806. [PMID: 23364048 PMCID: PMC3613164 DOI: 10.1093/hmg/ddt029] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 12/19/2012] [Accepted: 01/24/2013] [Indexed: 12/11/2022] Open
Abstract
DNA methylation plays an important role in suppressing retrotransposon activity in mammalian genomes, yet there are stages of mammalian development where global hypomethylation puts the genome at risk of retrotransposition-mediated genetic instability. Hypomethylated primordial germ cells appear to limit this risk by expressing a cohort of retrotransposon-suppressing genome-defence genes whose silencing depends on promoter DNA methylation. Here, we investigate whether similar mechanisms operate in hypomethylated trophectoderm-derived components of the mammalian placenta to couple expression of genome-defence genes to the potential for retrotransposon activity. We show that the hypomethylated state of the mouse placenta results in activation of only one of the hypomethylation-sensitive germline genome-defence genes: Tex19.1. Tex19.1 appears to play an important role in placenta function as Tex19.1(-/-) mouse embryos exhibit intra-uterine growth retardation and have small placentas due to a reduction in the number of spongiotrophoblast, glycogen trophoblast and sinusoidal trophoblast giant cells. Furthermore, we show that retrotransposon mRNAs are derepressed in Tex19.1(-/-) placentas and that protein encoded by the LINE-1 retrotransposon is upregulated in hypomethylated trophectoderm-derived cells that normally express Tex19.1. This study suggests that post-transcriptional genome-defence mechanisms are operating in the placenta to protect the hypomethylated cells in this tissue from retrotransposons and suggests that imbalances between retrotransposon activity and genome-defence mechanisms could contribute to placenta dysfunction and disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Richard R. Meehan
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Ian R. Adams
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| |
Collapse
|
37
|
Bickert T, Marshall RP, Zhang Z, Ludewig P, Binder M, Klinke A, Rottbauer W, Amling M, Wagener C, Ito WD, Horst AK. Acceleration of Collateral Development by Carcinoembryonic Antigen-Related Cell Adhesion Molecule 1 Expression on CD11b
+
/Gr-1
+
Myeloid Cells—Brief Report. Arterioscler Thromb Vasc Biol 2012; 32:2566-8. [DOI: 10.1161/atvbaha.112.300015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Thomas Bickert
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| | - Robert Percy Marshall
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| | - Ziyang Zhang
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| | - Peter Ludewig
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| | - Mascha Binder
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| | - Anna Klinke
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| | - Wolfgang Rottbauer
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| | - Michael Amling
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| | - Christoph Wagener
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| | - Wulf D. Ito
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| | - Andrea Kristina Horst
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| |
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: 3.1] [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
|
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: 0.8] [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
|
40
|
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: 2.3] [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
|
41
|
Ji RC. Macrophages are important mediators of either tumor- or inflammation-induced lymphangiogenesis. Cell Mol Life Sci 2012; 69:897-914. [PMID: 21984600 PMCID: PMC11114502 DOI: 10.1007/s00018-011-0848-6] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 07/26/2011] [Accepted: 09/22/2011] [Indexed: 01/13/2023]
Abstract
The lymphatic system provides important functions for tissue fluid homeostasis and immune response. Lymphangiogenesis, the formation of new lymphatics, comprises a series of complex cellular events in vitro or in vivo, e.g., proliferation, differentiation, and sprouting. Recent evidence has implied that macrophages act as a direct structural contributor to lymphatic endothelial walls or secret VEGF-C/-D and VEGF-A to initiate lymphangiogenesis in inflamed or tumor tissues. Bone marrow-derived macrophages are versatile cells that express different functional programs in response to exposure to microenvironmental signals, and can be identified by specific expression of a number of proteins, F4/80, CD11b, and CD68. Several causative factors, e.g., NF-κB, IL-1β, TNF-α, SDF-1, M-CSF, especially TonEBP/VEGF-C signaling, may be actively involved in macrophage-induced lymphangiogenesis. Alteration of macrophage phenotype and function has a profound effect on the development and progression of inflammation and malignancy, and macrophage depletion for controlling lymphangiogenesis may provide a novel approach for prevention and treatment of lymphatic-associated diseases.
Collapse
Affiliation(s)
- Rui-Cheng Ji
- Department of Human Anatomy, Oita University Faculty of Medicine, Oita 879-5593, Japan.
| |
Collapse
|
42
|
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: 1.1] [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
|
43
|
Abstract
Leishmania is a genus of protozoan parasites that are transmitted by the bite of phlebotomine sandflies and give rise to a range of diseases (collectively known as leishmaniases) that affect over 150 million people worldwide. Cellular immune mechanisms have a major role in the control of infections with all Leishmania spp. However, as discussed in this Review, recent evidence suggests that each host-pathogen combination evokes different solutions to the problems of parasite establishment, survival and persistence. Understanding the extent of this diversity will be increasingly important in ensuring the development of broadly applicable vaccines, drugs and immunotherapeutic interventions.
Collapse
|
44
|
CEACAM1 creates a pro-angiogenic tumor microenvironment that supports tumor vessel maturation. Oncogene 2011; 30:4275-88. [PMID: 21532628 DOI: 10.1038/onc.2011.146] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We have studied the effects of carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) on tumor angiogenesis in murine ductal mammary adenocarcinomas. We crossed transgenic mice with whey acidic protein promoter-driven large T-antigen expression (WAP-T mice) with oncogene-induced mammary carcinogenesis with CEACAM1null mice, and with Tie2-Ceacam1 transgenics, in which the Tie2 promoter drives endothelial overexpression of CEACAM1 (WAP-T × CEACAM1(endo+) mice), and analyzed tumor vascularization, angiogenesis and vessel maturation in these mice. Using flat-panel volume computed tomography (fpVCT) and histology, we found that WAP-T × CEACAM1(endo+) mice exhibited enhanced tumoral vascularization owing to CEACAM1(+) vessels in the tumor periphery, and increased intratumoral angiogenesis compared with controls. In contrast, vascularization of CEACAM1null/WAP-T-derived tumors was poor, and tumor vessels were dilated, leaky and showed poor pericyte coverage. Consequently, the tumoral vasculature could not be visualized in CEACAM1null/WAP-T mice by fpVCT, and we observed poor organization of the perivascular extracellular matrix (ECM), accompanied by the accumulation of collagen IV-degrading matrix metalloproteinase 9(+) (MMP9(+)) leukocytes and stromal cells. Vascular instability and alterations in ECM structure were accompanied by a significant increase in pulmonary metastases in CEACAM1null/WAP-T mice, whereas only occasional metastases were observed in CEACAM1(+) hosts. In CEACAM1(+) hosts, intratumoral vessels did not express CEACAM1, but they were intact, extensively covered with pericytes and framed by a well-organized perivascular ECM. MMP9(+) accessory cells were largely absent. Orthotopic transplantation of primary WAP-T- and CEACAM1null/WAP-T tumors into all three mouse lines confirmed that a CEACAM1(+) host environment is a prerequisite for productive angiogenic remodeling of the tumor microenvironment. Hence, CEACAM1 expression in the tumor periphery determines the vascular phenotype in a tumor, whereas systemic absence of CEACAM1 interferes with the formation of an organized tumor matrix and intratumoral vessel maturation.
Collapse
|
45
|
Yurdakul P, Dalton J, Beattie L, Brown N, Erguven S, Maroof A, Kaye PM. Compartment-specific remodeling of splenic micro-architecture during experimental visceral leishmaniasis. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:23-9. [PMID: 21703391 PMCID: PMC3123882 DOI: 10.1016/j.ajpath.2011.03.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 03/10/2011] [Accepted: 03/21/2011] [Indexed: 12/14/2022]
Abstract
Progressive splenomegaly is a hallmark of visceral leishmaniasis in humans, canids, and rodents. In experimental murine visceral leishmaniasis, splenomegaly is accompanied by pronounced changes in microarchitecture, including expansion of the red pulp vascular system, neovascularization of the white pulp, and remodeling of the stromal cell populations that define the B-cell and T-cell compartments. Here, we show that Ly6C/G+ (Gr-1+) cells, including neutrophils and inflammatory monocytes, accumulate in the splenic red pulp during infection. Cell depletion using monoclonal antibody against either Ly6C/G+ (Gr-1; RB6) or Ly6G+ (1A8) cells increased parasite burden. In contrast, depletion of Ly6C/G+ cells, but not Ly6G+ cells, halted the progressive remodeling of Meca-32+ and CD31+ red pulp vasculature. Strikingly, neither treatment affected white pulp neovascularization or the remodeling of the fibroblastic reticular cell and follicular dendritic cell networks. These findings demonstrate a previously unrecognized compartment-dependent selectivity to the process of splenic vascular remodeling during experimental murine visceral leishmaniasis, attributable to Ly6C+ inflammatory monocytes.
Collapse
Affiliation(s)
- Pinar Yurdakul
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
46
|
Nouvion AL, Oubaha M, LeBlanc S, Davis EC, Jastrow H, Kammerer R, Breton V, Turbide C, Ergun S, Gratton JP, Beauchemin N. CEACAM1: a key regulator of vascular permeability. J Cell Sci 2010; 123:4221-30. [DOI: 10.1242/jcs.073635] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Carcinoembryonic antigen cell adhesion molecule-1 (CEACAM1) is an immunoglobulin-like cell surface co-receptor expressed on epithelial, hematopoietic and endothelial cells. CEACAM1 functions as an adhesion molecule, mainly binding to itself or other members of the CEA family. We and others have previously shown that CEACAM1 is crucial for in vivo vascular integrity during ischemic neo-vascularization. Here, we have deciphered the roles of CEACAM1 in normal and pathological vascularization. We have found that Ceacam1−/− mice exhibit a significant increase in basal vascular permeability related to increased basal Akt and endothelial nitric oxide synthase (eNOS) activation in primary murine lung endothelial cells (MLECs). Moreover, CEACAM1 deletion in MLECs inhibits VEGF-mediated nitric oxide (NO) production, consistent with defective VEGF-dependent in vivo permeability in Ceacam1−/− mice. In addition, Ceacam1-null mice exhibit increased permeability of tumor vasculature. Finally, we demonstrate that CEACAM1 is tyrosine-phosphorylated upon VEGF treatment in a SHP-1- and Src-dependent manner, and that the key residues of the long cytoplasmic domain of CEACAM1 are crucial for CEACAM1 phosphorylation and NO production. This data represents the first report, to our knowledge, of a functional link between CEACAM1 and the VEGFR2/Akt/eNOS-mediated vascular permeability pathway.
Collapse
Affiliation(s)
- Anne-Laure Nouvion
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Malika Oubaha
- Laboratory of Endothelial Cell Biology, Institut de Recherches Cliniques de Montréal, Université de Montréal, Montréal, QC H2W 2T2, Canada
| | - Sarah LeBlanc
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Elaine C. Davis
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 2B2, Canada
| | - Holger Jastrow
- Institute of Anatomy, University Hospital Essen, Essen 45147, Germany
| | - Robert Kammerer
- Institute of Immunology, Friedrich-Loeffler-Institute, Tuebingen 72076, Germany
| | - Valérie Breton
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Claire Turbide
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Suleyman Ergun
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 2B2, Canada
| | - Jean-Philippe Gratton
- Laboratory of Endothelial Cell Biology, Institut de Recherches Cliniques de Montréal, Université de Montréal, Montréal, QC H2W 2T2, Canada
| | - Nicole Beauchemin
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada
- Departments of Biochemistry, Medicine and Oncology, McGill University, Montreal, QC H3G 1Y6, Canada
| |
Collapse
|
47
|
Watt SM, Athanassopoulos A, Harris AL, Tsaknakis G. Human endothelial stem/progenitor cells, angiogenic factors and vascular repair. J R Soc Interface 2010; 7 Suppl 6:S731-51. [PMID: 20843839 DOI: 10.1098/rsif.2010.0377.focus] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Neovascularization or new blood vessel formation is of utmost importance not only for tissue and organ development and for tissue repair and regeneration, but also for pathological processes, such as tumour development. Despite this, the endothelial lineage, its origin, and the regulation of endothelial development and function either intrinsically from stem cells or extrinsically by proangiogenic supporting cells and other elements within local and specific microenvironmental niches are still not fully understood. There can be no doubt that for most tissues and organs, revascularization represents the holy grail for tissue repair, with autologous endothelial stem/progenitor cells, their proangiogenic counterparts and the products of these cells all being attractive targets for therapeutic intervention. Historically, a great deal of controversy has surrounded the identification and origin of cells and factors that contribute to revascularization, the use of such cells or their products as biomarkers to predict and monitor tissue damage and repair or tumour progression and therapeutic responses, and indeed their efficacy in revascularizing and repairing damaged tissues. Here, we will review the role of endothelial progenitor cells and of supporting proangiogenic cells and their products, principally in humans, as diagnostic and therapeutic agents for wound repair and tissue regeneration.
Collapse
Affiliation(s)
- Suzanne M Watt
- Stem Cell Laboratory and Stem Cells and Immunotherapies, NHS Blood and Transplant, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK.
| | | | | | | |
Collapse
|
48
|
Forrester JV, Xu H, Kuffová L, Dick AD, McMenamin PG. Dendritic cell physiology and function in the eye. Immunol Rev 2010; 234:282-304. [PMID: 20193026 DOI: 10.1111/j.0105-2896.2009.00873.x] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The eye and the brain are immunologically privileged sites, a property previously attributed to the lack of a lymphatic circulation. However, recent tracking studies confirm that these organs have good communication through classical site-specific lymph nodes, as well as direct connection through the blood circulation with the spleen. In addition, like all tissues, they contain resident myeloid cell populations that play important roles in tissue homeostasis and the response to foreign antigens. Most of the macrophage and dendritic cell (DC) populations in the eye are restricted to the supporting connective tissues, including the cornea, while the neural tissue (the retina) contains almost no DCs, occasional macrophages (perivascularly distributed), and a specialized myeloid cell type, the microglial cell. Resident microglial cells are normally programmed for immunological tolerance. The privileged status of the eye, however, is relative, as it is susceptible to immune-mediated inflammatory disease, both infectious and autoimmune. Intraocular inflammation (uveitis and uveoretinitis) and corneal graft rejection constitute two of the more common inflammatory conditions affecting the eye leading to considerable morbidity (blindness). As corneal graft rejection occurs almost exclusively by indirect allorecognition, host DCs play a major role in this process and are likely to be modified in their behavior by the ocular microenvironment. Ocular surface disease, including allergy and atopy, also comprise a significant group of immune-mediated eye disorders in which DCs participate, while infectious disease such as herpes simplex keratitis is thought to be initiated via corneal DCs. Intriguingly, some more common conditions previously thought to be degenerative (e.g. age-related macular degeneration) may have an autoimmune component in which ocular DCs and macrophages are critically involved. Recently, the possibility of harnessing the tolerizing potential of DCs has been applied to experimental models of autoimmune uveoretinitis with good effect. This approach has considerable potential for use in translational clinical therapy to prevent sight-threatening disease caused by ocular inflammation.
Collapse
Affiliation(s)
- John V Forrester
- Section of Immunology and Infection, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK.
| | | | | | | | | |
Collapse
|
49
|
|