1
|
Berve K, Michel J, Tietz S, Blatti C, Ivan D, Enzmann G, Lyck R, Deutsch U, Locatelli G, Engelhardt B. Junctional adhesion molecule-A deficient mice are protected from severe experimental autoimmune encephalomyelitis. Eur J Immunol 2024; 54:e2350761. [PMID: 38566526 DOI: 10.1002/eji.202350761] [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: 09/07/2023] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 04/04/2024]
Abstract
In multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE), early pathological features include immune cell infiltration into the central nervous system (CNS) and blood-brain barrier (BBB) disruption. We investigated the role of junctional adhesion molecule-A (JAM-A), a tight junction protein, in active EAE (aEAE) pathogenesis. Our study confirms JAM-A expression at the blood-brain barrier and its luminal redistribution during aEAE. JAM-A deficient (JAM-A-/-) C57BL/6J mice exhibited milder aEAE, unrelated to myelin oligodendrocyte glycoprotein-specific CD4+ T-cell priming. While JAM-A absence influenced macrophage behavior on primary mouse brain microvascular endothelial cells (pMBMECs) under flow in vitro, it did not impact T-cell extravasation across primary mouse brain microvascular endothelial cells. At aEAE onset, we observed reduced lymphocyte and CCR2+ macrophage infiltration into the spinal cord of JAM-A-/- mice compared to control littermates. This correlated with increased CD3+ T-cell accumulation in spinal cord perivascular spaces and brain leptomeninges, suggesting JAM-A absence leads to T-cell trapping in central nervous system border compartments. In summary, JAM-A plays a role in immune cell infiltration and clinical disease progression in aEAE.
Collapse
Affiliation(s)
- Kristina Berve
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Julia Michel
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Silvia Tietz
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Claudia Blatti
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Daniela Ivan
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Gaby Enzmann
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Ruth Lyck
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Urban Deutsch
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | | | |
Collapse
|
2
|
CZUBAK-PROWIZOR KAMILA, SWIATKOWSKA MARIA. Junctional adhesion molecule-A (JAM-A) in gynecological cancers: Current state of knowledge. BIOCELL 2023. [DOI: 10.32604/biocell.2023.025677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
3
|
Kiss M, Lebegge E, Murgaski A, Van Damme H, Kancheva D, Brughmans J, Scheyltjens I, Talebi A, Awad RM, Elkrim Y, Bardet PMR, Arnouk SM, Goyvaerts C, Swinnen J, Nana FA, Van Ginderachter JA, Laoui D. Junctional adhesion molecule-A is dispensable for myeloid cell recruitment and diversification in the tumor microenvironment. Front Immunol 2022; 13:1003975. [PMID: 36531986 PMCID: PMC9751033 DOI: 10.3389/fimmu.2022.1003975] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/15/2022] [Indexed: 12/04/2022] Open
Abstract
Junctional adhesion molecule-A (JAM-A), expressed on the surface of myeloid cells, is required for extravasation at sites of inflammation and may also modulate myeloid cell activation. Infiltration of myeloid cells is a common feature of tumors that drives disease progression, but the function of JAM-A in this phenomenon and its impact on tumor-infiltrating myeloid cells is little understood. Here we show that systemic cancer-associated inflammation in mice enhanced JAM-A expression selectively on circulating monocytes in an IL1β-dependent manner. Using myeloid-specific JAM-A-deficient mice, we found that JAM-A was dispensable for recruitment of monocytes and other myeloid cells to tumors, in contrast to its reported role in inflammation. Single-cell RNA sequencing revealed that loss of JAM-A did not influence the transcriptional reprogramming of myeloid cells in the tumor microenvironment. Overall, our results support the notion that cancer-associated inflammation can modulate the phenotype of circulating immune cells, and we demonstrate that tumors can bypass the requirement of JAM-A for myeloid cell recruitment and reprogramming.
Collapse
Affiliation(s)
- Máté Kiss
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium,Laboratory of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium,*Correspondence: Máté Kiss, ; Damya Laoui,
| | - Els Lebegge
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Aleksandar Murgaski
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium,Laboratory of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
| | - Helena Van Damme
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Daliya Kancheva
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium,Laboratory of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
| | - Jan Brughmans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium,Laboratory of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
| | - Isabelle Scheyltjens
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ali Talebi
- Laboratory of Lipid Metabolism and Cancer, KU Leuven, Leuven, Belgium
| | - Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yvon Elkrim
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Pauline M. R. Bardet
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium,Laboratory of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
| | - Sana M. Arnouk
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cleo Goyvaerts
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Johan Swinnen
- Laboratory of Lipid Metabolism and Cancer, KU Leuven, Leuven, Belgium
| | - Frank Aboubakar Nana
- Division of Pneumology, CHU UCL Namur (Godinne Site), UCLouvain, Yvoir, Belgium,Division of Pneumology, Cliniques Universitaires St-Luc, UCLouvain, Brussels, Belgium
| | - Jo A. Van Ginderachter
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Damya Laoui
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium,Laboratory of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium,*Correspondence: Máté Kiss, ; Damya Laoui,
| |
Collapse
|
4
|
Choi S, Ferrari G, Moyle LA, Mackinlay K, Naouar N, Jalal S, Benedetti S, Wells C, Muntoni F, Tedesco FS. Assessing and enhancing migration of human myogenic progenitors using directed iPS cell differentiation and advanced tissue modelling. EMBO Mol Med 2022; 14:e14526. [PMID: 36161772 PMCID: PMC9549733 DOI: 10.15252/emmm.202114526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 08/19/2022] [Accepted: 08/19/2022] [Indexed: 02/05/2023] Open
Abstract
Muscle satellite stem cells (MuSCs) are responsible for skeletal muscle growth and regeneration. Despite their differentiation potential, human MuSCs have limited in vitro expansion and in vivo migration capacity, limiting their use in cell therapies for diseases affecting multiple skeletal muscles. Several protocols have been developed to derive MuSC-like progenitors from human induced pluripotent stem (iPS) cells (hiPSCs) to establish a source of myogenic cells with controllable proliferation and differentiation. However, current hiPSC myogenic derivatives also suffer from limitations of cell migration, ultimately delaying their clinical translation. Here we use a multi-disciplinary approach including bioinformatics and tissue engineering to show that DLL4 and PDGF-BB improve migration of hiPSC-derived myogenic progenitors. Transcriptomic analyses demonstrate that this property is conserved across species and multiple hiPSC lines, consistent with results from single cell motility profiling. Treated cells showed enhanced trans-endothelial migration in transwell assays. Finally, increased motility was detected in a novel humanised assay to study cell migration using 3D artificial muscles, harnessing advanced tissue modelling to move hiPSCs closer to future muscle gene and cell therapies.
Collapse
Affiliation(s)
- SungWoo Choi
- The Francis Crick InstituteLondonUK
- Department of Cell and Developmental BiologyUniversity College LondonLondonUK
| | - Giulia Ferrari
- Department of Cell and Developmental BiologyUniversity College LondonLondonUK
| | - Louise A Moyle
- Department of Cell and Developmental BiologyUniversity College LondonLondonUK
- Present address:
Institute of Biomedical EngineeringUniversity of TorontoTorontoONCanada
| | - Kirsty Mackinlay
- Department of Cell and Developmental BiologyUniversity College LondonLondonUK
- Present address:
Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Naira Naouar
- Institut de Biologie Paris Seine FR3631, Plateforme de Bioinformatique ARTbioSorbonne UniversitéParisFrance
| | - Salma Jalal
- The Francis Crick InstituteLondonUK
- Department of Cell and Developmental BiologyUniversity College LondonLondonUK
| | - Sara Benedetti
- UCL Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
- National Institute for Health Research Great Ormond Street Hospital Biomedical Research CentreLondonUK
| | - Christine Wells
- Centre for Stem Cell SystemsThe University of MelbourneMelbourneVICAustralia
| | - Francesco Muntoni
- National Institute for Health Research Great Ormond Street Hospital Biomedical Research CentreLondonUK
- Dubowitz Neuromuscular CentreUCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital for ChildrenLondonUK
| | - Francesco Saverio Tedesco
- The Francis Crick InstituteLondonUK
- Department of Cell and Developmental BiologyUniversity College LondonLondonUK
- Dubowitz Neuromuscular CentreUCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital for ChildrenLondonUK
| |
Collapse
|
5
|
Klingensmith NJ, Fay KT, Swift DA, Bazzano JM, Lyons JD, Chen CW, Meng M, Ramonell KM, Liang Z, Burd EM, Parkos CA, Ford ML, Coopersmith CM. Junctional adhesion molecule-A deletion increases phagocytosis and improves survival in a murine model of sepsis. JCI Insight 2022; 7:156255. [PMID: 35819838 PMCID: PMC9462501 DOI: 10.1172/jci.insight.156255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 07/07/2022] [Indexed: 11/28/2022] Open
Abstract
Expression of the tight junction–associated protein junctional adhesion molecule-A (JAM-A) is increased in sepsis, although the significance of this is unknown. Here, we show that septic JAM-A –/– mice have increased gut permeability, yet paradoxically have decreased bacteremia and systemic TNF and IL-1β expression. Survival is improved in JAM-A–/– mice. However, intestine-specific JAM-A–/– deletion does not alter mortality, suggesting that the mortality benefit conferred in mice lacking JAM-A is independent of the intestine. Septic JAM-A–/– mice have increased numbers of splenic CD44hiCD4+ T cells, decreased frequency of TNF+CD4+ cells, and elevated frequency of IL-2+CD4+ cells. Septic JAM-A–/– mice have increased numbers of B cells in mesenteric lymph nodes with elevated serum IgA and intraepithelial lymphocyte IgA production. JAM-A–/– × RAG–/– mice have improved survival compared with RAG–/– mice and identical mortality as WT mice. Gut neutrophil infiltration and neutrophil phagocytosis are increased in JAM-A–/– mice, while septic JAM-A–/– mice depleted of neutrophils lose their survival advantage. Therefore, increased bacterial clearance via neutrophils and an altered systemic inflammatory response with increased opsonizing IgA produced through the adaptive immune system results in improved survival in septic JAM-A–/– mice. JAM-A may be a therapeutic target in sepsis via immune mechanisms not related to its role in permeability.
Collapse
Affiliation(s)
- Nathan J Klingensmith
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, United States of America
| | - Katherine T Fay
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, United States of America
| | - David A Swift
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, United States of America
| | - Julia Mr Bazzano
- Department of Surgery and Emory Critical Care Center, Emory Univerisity School of Medicine, Atlanta, United States of America
| | - John D Lyons
- Department of Surgery and Emory Critical Care Center, Emory Univerisity School of Medicine, Atlanta, United States of America
| | - Ching-Wen Chen
- Department of Surgery and Emory Critical Care Center, Emory Univerisity School of Medicine, Atlanta, United States of America
| | - Mei Meng
- Department of Surgery and Emory Critical Care Center, Emory Univerisity School of Medicine, Atlanta, United States of America
| | - Kimberly M Ramonell
- Department of Surgery and Emory Critical Care Center, Emory Univerisity School of Medicine, Atlanta, United States of America
| | - Zhe Liang
- Department of Surgery and Emory Critical Care Center, Emory Univerisity School of Medicine, Atlanta, United States of America
| | - Eileen M Burd
- Department of Pathology and Laboratory Medicine, Emory Univerisity School of Medicine, Atlanta, United States of America
| | - Charles A Parkos
- Department of Pathology, University of Michigan, Ann Arbor, United States of America
| | - Mandy L Ford
- Department of Surgery and Emory Critical Care Center, Emory Univerisity School of Medicine, Atlanta, United States of America
| | - Craig M Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory Univerisity School of Medicine, Atlanta, United States of America
| |
Collapse
|
6
|
Wang J, Chen X. Junctional Adhesion Molecules: Potential Proteins in Atherosclerosis. Front Cardiovasc Med 2022; 9:888818. [PMID: 35872908 PMCID: PMC9302484 DOI: 10.3389/fcvm.2022.888818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
Junctional adhesion molecules (JAMs) are cell-cell adhesion molecules of the immunoglobulin superfamily and are involved in the regulation of diverse atherosclerosis-related processes such as endothelial barrier maintenance, leucocytes transendothelial migration, and angiogenesis. To combine and further broaden related results, this review concluded the recent progress in the roles of JAMs and predicted future studies of JAMs in the development of atherosclerosis.
Collapse
Affiliation(s)
- Junqi Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Xiaoping Chen,
| |
Collapse
|
7
|
Brozat JF, Brandt EF, Stark M, Fischer P, Wirtz TH, Flaßhove A, Rodenhausen AN, Vajen T, Heinzmann ACA, Schmitz SMT, Abu Jhaisha S, Röth AA, Koenen RR, Sahin H, Trautwein C, Berres ML. JAM-A is a multifaceted regulator in hepatic fibrogenesis, supporting LSEC integrity and stellate cell quiescence. Liver Int 2022; 42:1185-1203. [PMID: 35129269 DOI: 10.1111/liv.15187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 02/13/2023]
Abstract
BACKGROUND AND AIMS Leukocyte infiltration is a hallmark of hepatic inflammation. The Junctional Adhesion Molecule A (JAM-A) is a crucial regulator of leukocyte extravasation and is upregulated in human viral fibrosis. Reduced shear stress within hepatic sinusoids and the specific phenotype of liver sinusoidal endothelial cells (LSEC) cumulate in differing adhesion characteristics during liver fibrosis. The aim of this study was to define the functional role of cell-specific adhesion molecule JAM-A during hepatic fibrogenesis. METHODS Complete, conditional (intestinal epithelial; endothelial) and bone marrow chimeric Jam-a knockout animals and corresponding C57Bl/6 wild-type animals were treated with carbon tetrachloride (CCl4 , 6 weeks). For functional analyses of JAM-A, comprehensive in vivo studies, co-culture models and flow-based adhesion assays were performed. RESULTS Complete and bone marrow-derived Jam-a-/- animals showed aggravated fibrosis with increased non-sinusoidal, perivascular accumulation of CD11b+ F4/80+ monocyte-derived macrophages in contrast to wild-type mice. Despite being associated with disturbed epithelial barrier function, an intestinal epithelial Jam-a knockout did not affect fibrogenesis. In endothelial-specific Jam-a-/- animals, liver fibrosis was aggravated alongside sinusoid capillarization and hepatic stellate cell (HSC) activation. HSC activation is induced via Jam-a-/- LSEC-derived secretion of soluble factors. Sinusoid CD31 expression and hedgehog gene signalling were increased, but leukocyte infiltration and adhesion to LSECs remained unaffected. CONCLUSIONS Our models decipher cell-specific JAM-A to exert crucial functions during hepatic fibrogenesis. JAM-A on bone marrow-derived cells regulates non-sinusoidal vascular immune cell recruitment, while endothelial JAM-A controls liver sinusoid capillarization and HSC quiescence.
Collapse
Affiliation(s)
- Jonathan F Brozat
- Department of Internal Medicine III, RWTH Aachen University Hospital, RWTH Aachen, Aachen, Germany
| | - Elisa F Brandt
- Department of Internal Medicine III, RWTH Aachen University Hospital, RWTH Aachen, Aachen, Germany
| | - Myriam Stark
- Department of Internal Medicine III, RWTH Aachen University Hospital, RWTH Aachen, Aachen, Germany
| | - Petra Fischer
- Department of Internal Medicine III, RWTH Aachen University Hospital, RWTH Aachen, Aachen, Germany
| | - Theresa H Wirtz
- Department of Internal Medicine III, RWTH Aachen University Hospital, RWTH Aachen, Aachen, Germany
| | - Alexander Flaßhove
- Department of Internal Medicine III, RWTH Aachen University Hospital, RWTH Aachen, Aachen, Germany
| | - Aaron N Rodenhausen
- Department of Internal Medicine III, RWTH Aachen University Hospital, RWTH Aachen, Aachen, Germany
| | - Tanja Vajen
- Cardiovascular Research Laboratory, Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Alexandra C A Heinzmann
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Sophia M-T Schmitz
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, RWTH Aachen, Aachen, Germany
| | - Samira Abu Jhaisha
- Department of Internal Medicine III, RWTH Aachen University Hospital, RWTH Aachen, Aachen, Germany
| | - Anjali A Röth
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, RWTH Aachen, Aachen, Germany
| | - Rory R Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Hacer Sahin
- Department of Internal Medicine III, RWTH Aachen University Hospital, RWTH Aachen, Aachen, Germany
| | - Christian Trautwein
- Department of Internal Medicine III, RWTH Aachen University Hospital, RWTH Aachen, Aachen, Germany
| | - Marie-Luise Berres
- Department of Internal Medicine III, RWTH Aachen University Hospital, RWTH Aachen, Aachen, Germany
| |
Collapse
|
8
|
Gastrointestinal cancer-associated fibroblasts expressing Junctional Adhesion Molecule-A are amenable to infection by oncolytic reovirus. Cancer Gene Ther 2022; 29:1918-1929. [PMID: 35869278 PMCID: PMC9750869 DOI: 10.1038/s41417-022-00507-9] [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: 03/18/2022] [Revised: 06/13/2022] [Accepted: 07/06/2022] [Indexed: 01/25/2023]
Abstract
Gastrointestinal (GI) cancers are characterized by extensive tumor stroma that both promotes tumor progression and acts as a physical barrier for adjacent tumor cells, limiting the effect of current treatment modalities. Oncolytic virotherapy is currently investigated in clinical trials as a novel therapeutic agent for different malignancies of the GI tract, but it is largely unknown whether these viruses can also target the tumor stroma. Here, we investigated the tropism of two commonly studied OVs, adenovirus and reovirus, towards primary GI fibroblasts from human oesophageal, gastric, duodenal and pancreatic carcinomas (N = 36). GI fibroblasts were susceptible to type 3 Dearing (T3D) strain R124 and bioselected mutant reovirus (jin-3) infection but not oncolytic adenovirus (Ad5-Δ24). Efficient infection and apoptosis of human and mouse GI cancer-derived fibroblasts by these reoviruses was partially dependent on the expression of the reovirus entry receptor, Junctional Adhesion Molecule-A (JAM-A). Moreover, human GI cancer organoid-fibroblast co-cultures showed higher overall infectivity when containing JAM-A expressing fibroblasts as compared to JAM-A negative fibroblasts, indicating a potential role of JAM-A expressing fibroblasts for viral dissemination. We further show that JAM-A is not only necessary for efficient reovirus infection of fibroblasts but also partially mediates reovirus-induced apoptosis, dependent on signaling through the C-terminal PDZ-domain of JAM-A. Altogether, our data show the presence of JAM-A expressing fibroblasts in both human and murine GI cancers that are amenable to infection and induction of apoptosis by reovirus, extending the potential anti-cancer actions of reovirus with stromal targeting.
Collapse
|
9
|
Czubak-Prowizor K, Babinska A, Swiatkowska M. The F11 Receptor (F11R)/Junctional Adhesion Molecule-A (JAM-A) (F11R/JAM-A) in cancer progression. Mol Cell Biochem 2021; 477:79-98. [PMID: 34533648 PMCID: PMC8755661 DOI: 10.1007/s11010-021-04259-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 09/08/2021] [Indexed: 12/27/2022]
Abstract
The F11 Receptor (F11R), also called Junctional Adhesion Molecule-A (JAM-A) (F11R/JAM-A), is a transmembrane glycoprotein of the immunoglobulin superfamily, which is mainly located in epithelial and endothelial cell tight junctions and also expressed on circulating platelets and leukocytes. It participates in the regulation of various biological processes, as diverse as paracellular permeability, tight junction formation and maintenance, leukocyte transendothelial migration, epithelial-to-mesenchymal transition, angiogenesis, reovirus binding, and platelet activation. Dysregulation of F11R/JAM-A may result in pathological consequences and disorders in normal cell function. A growing body of evidence points to its role in carcinogenesis and invasiveness, but its tissue-specific pro- or anti-tumorigenic role remains a debated issue. The following review focuses on the F11R/JAM-A tissue-dependent manner in tumorigenesis and metastasis and also discusses the correlation between poor patient clinical outcomes and its aberrant expression. In the future, it will be required to clarify the signaling pathways that are activated or suppressed via the F11R/JAM-A protein in various cancer types to understand its multiple roles in cancer progression and further use it as a novel direct target for cancer treatment.
Collapse
Affiliation(s)
- Kamila Czubak-Prowizor
- Department of Cytobiology and Proteomics, Medical University of Lodz, 6/8 Mazowiecka St., 92-215, Lodz, Poland.
| | - Anna Babinska
- Department of Medicine, State University of New York Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY, 11203, USA
| | - Maria Swiatkowska
- Department of Cytobiology and Proteomics, Medical University of Lodz, 6/8 Mazowiecka St., 92-215, Lodz, Poland
| |
Collapse
|
10
|
Inflammation and tumor progression: signaling pathways and targeted intervention. Signal Transduct Target Ther 2021; 6:263. [PMID: 34248142 PMCID: PMC8273155 DOI: 10.1038/s41392-021-00658-5] [Citation(s) in RCA: 881] [Impact Index Per Article: 293.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 05/11/2021] [Accepted: 05/23/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer development and its response to therapy are regulated by inflammation, which either promotes or suppresses tumor progression, potentially displaying opposing effects on therapeutic outcomes. Chronic inflammation facilitates tumor progression and treatment resistance, whereas induction of acute inflammatory reactions often stimulates the maturation of dendritic cells (DCs) and antigen presentation, leading to anti-tumor immune responses. In addition, multiple signaling pathways, such as nuclear factor kappa B (NF-kB), Janus kinase/signal transducers and activators of transcription (JAK-STAT), toll-like receptor (TLR) pathways, cGAS/STING, and mitogen-activated protein kinase (MAPK); inflammatory factors, including cytokines (e.g., interleukin (IL), interferon (IFN), and tumor necrosis factor (TNF)-α), chemokines (e.g., C-C motif chemokine ligands (CCLs) and C-X-C motif chemokine ligands (CXCLs)), growth factors (e.g., vascular endothelial growth factor (VEGF), transforming growth factor (TGF)-β), and inflammasome; as well as inflammatory metabolites including prostaglandins, leukotrienes, thromboxane, and specialized proresolving mediators (SPM), have been identified as pivotal regulators of the initiation and resolution of inflammation. Nowadays, local irradiation, recombinant cytokines, neutralizing antibodies, small-molecule inhibitors, DC vaccines, oncolytic viruses, TLR agonists, and SPM have been developed to specifically modulate inflammation in cancer therapy, with some of these factors already undergoing clinical trials. Herein, we discuss the initiation and resolution of inflammation, the crosstalk between tumor development and inflammatory processes. We also highlight potential targets for harnessing inflammation in the treatment of cancer.
Collapse
|
11
|
Anti-CD321 antibody immunotherapy protects liver against ischemia and reperfusion-induced injury. Sci Rep 2021; 11:6312. [PMID: 33737554 PMCID: PMC7973783 DOI: 10.1038/s41598-021-85001-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 02/23/2021] [Indexed: 12/18/2022] Open
Abstract
The prognosis of the liver transplant patients was frequently deteriorated by ischemia and reperfusion injury (IRI) in the liver. Infiltration of inflammatory cells is reported to play critical roles in the pathogenesis of hepatic IRI. Although T lymphocytes, neutrophils and monocytes infiltrated into the liver underwent IRI, we found that neutrophil depletion significantly attenuated the injury and serum liver enzyme levels in a murine model. Interestingly, the expression of CD321/JAM-A/F11R, one of essential molecules for transmigration of circulating leukocytes into inflammatory tissues, was significantly augmented on hepatic sinusoid endothelium at 1 h after ischemia and maintained until 45 min after reperfusion. The intraportal administration of anti-CD321 monoclonal antibody (90G4) significantly inhibited the leukocytes infiltration after reperfusion and diminished the damage responses by hepatic IRI (serum liver enzymes, inflammatory cytokines and hepatocyte cell death). Taken together, presented results demonstrated that blockade of CD321 by 90G4 antibody significantly attenuated hepatic IRI accompanied with substantial inhibition of leukocytes infiltration, particularly inhibition of neutrophil infiltration in the early phase of reperfusion. Thus, our work offers a potent therapeutic target, CD321, for preventing liver IRI.
Collapse
|
12
|
Lin WC, Fessler MB. Regulatory mechanisms of neutrophil migration from the circulation to the airspace. Cell Mol Life Sci 2021; 78:4095-4124. [PMID: 33544156 PMCID: PMC7863617 DOI: 10.1007/s00018-021-03768-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/22/2020] [Accepted: 01/16/2021] [Indexed: 02/07/2023]
Abstract
The neutrophil, a short-lived effector leukocyte of the innate immune system best known for its proteases and other degradative cargo, has unique, reciprocal physiological interactions with the lung. During health, large numbers of ‘marginated’ neutrophils reside within the pulmonary vasculature, where they patrol the endothelial surface for pathogens and complete their life cycle. Upon respiratory infection, rapid and sustained recruitment of neutrophils through the endothelial barrier, across the extravascular pulmonary interstitium, and again through the respiratory epithelium into the airspace lumen, is required for pathogen killing. Overexuberant neutrophil trafficking to the lung, however, causes bystander tissue injury and underlies several acute and chronic lung diseases. Due in part to the unique architecture of the lung’s capillary network, the neutrophil follows a microanatomic passage into the distal airspace unlike that observed in other end-organs that it infiltrates. Several of the regulatory mechanisms underlying the stepwise recruitment of circulating neutrophils to the infected lung have been defined over the past few decades; however, fundamental questions remain. In this article, we provide an updated review and perspective on emerging roles for the neutrophil in lung biology, on the molecular mechanisms that control the trafficking of neutrophils to the lung, and on past and ongoing efforts to design therapeutics to intervene upon pulmonary neutrophilia in lung disease.
Collapse
Affiliation(s)
- Wan-Chi Lin
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, P.O. Box 12233, MD D2-01, Research Triangle Park, NC, 27709, USA
| | - Michael B Fessler
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, P.O. Box 12233, MD D2-01, Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
13
|
Bonilha CS, Benson RA, Brewer JM, Garside P. Targeting Opposing Immunological Roles of the Junctional Adhesion Molecule-A in Autoimmunity and Cancer. Front Immunol 2020; 11:602094. [PMID: 33324419 PMCID: PMC7723963 DOI: 10.3389/fimmu.2020.602094] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/02/2020] [Indexed: 01/04/2023] Open
Abstract
The junctional adhesion molecule-A (JAM-A) is a cell surface adhesion molecule expressed on platelets, epithelial cells, endothelial cells and leukocytes (e. g. monocytes and dendritic cells). JAM-A plays a relevant role in leukocyte trafficking and its therapeutic potential has been studied in several pathological conditions due to its capacity to induce leukocyte migration out of inflamed sites or infiltration into tumor sites. However, disruption of JAM-A pathways may worsen clinical pathology in some cases. As such, the effects of JAM-A manipulation on modulating immune responses in the context of different diseases must be better understood. In this mini-review, we discuss the potential of JAM-A as a therapeutic target, summarizing findings from studies manipulating JAM-A in the context of inflammatory diseases (e.g. autoimmune diseases) and cancer and highlighting described mechanisms.
Collapse
Affiliation(s)
- Caio S. Bonilha
- College of Medical, Veterinary and Life Sciences, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Robert A. Benson
- College of Medical, Veterinary and Life Sciences, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
- Research and Development Department, Antibody Analytics Ltd., Newhouse, Lanarkshire, United Kingdom
| | - James M. Brewer
- College of Medical, Veterinary and Life Sciences, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Paul Garside
- College of Medical, Veterinary and Life Sciences, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
14
|
Li X, Li L, Liu X, Wu J, Sun X, Li Z, Geng YJ, Liu F, Zhou Y. Attenuation of Cardiac Ischaemia-reperfusion Injury by Treatment with Hydrogen-rich Water. Curr Mol Med 2020; 19:294-302. [PMID: 30907314 PMCID: PMC7061975 DOI: 10.2174/1566524019666190321113544] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 02/18/2019] [Accepted: 03/19/2019] [Indexed: 02/07/2023]
Abstract
Background: Hydrogen has been shown to exert a bioactive effect on the myocardium. This study examined the signalling pathways for hydrogen attenuating ischaemia-reperfusion injury. Methods: In total, 20 male Wistar rats were evaluated for the effects of hydrogen-rich water on ischaemia-reperfusion in hearts. Left ventricular tissue was taken for screening and analysis of active protein factors by protein chip technology. The enrichment of the KEGG pathway was obtained by using the Gene Ontology (GO) enrichment principle. The expression of JAK2, STAT1, STAT3, p-STAT1, p-JAK2, p-STAT3 in rat myocardium was detected by Western blot analysis and immunohistochemistry. The apoptosis rates of the control and hydrogen-rich water groups were detected by TUNEL staining. Results: The expression levels of 25 proteins, including five transduction pathways, were downregulated in the hydrogen-rich water group. The expression levels of p-JAK2/JAK2, p-STAT3/STAT3 were upregulated in the hydrogen-rich water group compared with the control group, and p-STAT1/STAT1 was downregulated in the hydrogen-rich water group compared with the control group. Furthermore, the apoptosis rate was significantly decreased in the hydrogen-rich water group, as well. Conclusion: Hydrogen-rich water may inhibit the apoptosis of cardiomyocytes after ischaemia-reperfusion by upregulating the expression of the JAK2-STAT3 signalling pathway, which reduces ischaemia-reperfusion injury.
Collapse
Affiliation(s)
- Xiangzi Li
- School of Medicine, Hebei University, Baoding 071000, China
| | - Liangtong Li
- School of Medicine, Hebei University, Baoding 071000, China
| | - Xuanchen Liu
- School of Medicine, Hebei University, Baoding 071000, China
| | - Jiawen Wu
- School of Medicine, Hebei University, Baoding 071000, China
| | - Xiaoyu Sun
- School of Medicine, Hebei University, Baoding 071000, China
| | - Zhilin Li
- School of Chemistry, Hebei University, Baoding 071000, China
| | - Yong-Jian Geng
- Centre for Cardiovascular Biology and Atherosclerosis Research, Division of Cardiovascular Medicine, Department of Internal Medicine, University of Texas Medical School at Houston, Houston, TX 77030, United States
| | - Fulin Liu
- School of Medicine, Hebei University, Baoding 071000, China.,Department of Cardiac Surgery, Affiliated Hospital of Hebei University, Baoding 071000, China
| | - Yujuan Zhou
- School of Medicine, Hebei University, Baoding 071000, China
| |
Collapse
|
15
|
Kakogiannos N, Ferrari L, Giampietro C, Scalise AA, Maderna C, Ravà M, Taddei A, Lampugnani MG, Pisati F, Malinverno M, Martini E, Costa I, Lupia M, Cavallaro U, Beznoussenko GV, Mironov AA, Fernandes B, Rudini N, Dejana E, Giannotta M. JAM-A Acts via C/EBP-α to Promote Claudin-5 Expression and Enhance Endothelial Barrier Function. Circ Res 2020; 127:1056-1073. [PMID: 32673519 PMCID: PMC7508279 DOI: 10.1161/circresaha.120.316742] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
RATIONALE Intercellular tight junctions are crucial for correct regulation of the endothelial barrier. Their composition and integrity are affected in pathological contexts, such as inflammation and tumor growth. JAM-A (junctional adhesion molecule A) is a transmembrane component of tight junctions with a role in maintenance of endothelial barrier function, although how this is accomplished remains elusive. OBJECTIVE We aimed to understand the molecular mechanisms through which JAM-A expression regulates tight junction organization to control endothelial permeability, with potential implications under pathological conditions. METHODS AND RESULTS Genetic deletion of JAM-A in mice significantly increased vascular permeability. This was associated with significantly decreased expression of claudin-5 in the vasculature of various tissues, including brain and lung. We observed that C/EBP-α (CCAAT/enhancer-binding protein-α) can act as a transcription factor to trigger the expression of claudin-5 downstream of JAM-A, to thus enhance vascular barrier function. Accordingly, gain-of-function for C/EBP-α increased claudin-5 expression and decreased endothelial permeability, as measured by the passage of fluorescein isothiocyanate (FITC)-dextran through endothelial monolayers. Conversely, C/EBP-α loss-of-function showed the opposite effects of decreased claudin-5 levels and increased endothelial permeability. Mechanistically, JAM-A promoted C/EBP-α expression through suppression of β-catenin transcriptional activity, and also through activation of EPAC (exchange protein directly activated by cAMP). C/EBP-α then directly binds the promoter of claudin-5 to thereby promote its transcription. Finally, JAM-A-C/EBP-α-mediated regulation of claudin-5 was lost in blood vessels from tissue biopsies from patients with glioblastoma and ovarian cancer. CONCLUSIONS We describe here a novel role for the transcription factor C/EBP-α that is positively modulated by JAM-A, a component of tight junctions that acts through EPAC to up-regulate the expression of claudin-5, to thus decrease endothelial permeability. Overall, these data unravel a regulatory molecular pathway through which tight junctions limit vascular permeability. This will help in the identification of further therapeutic targets for diseases associated with endothelial barrier dysfunction. Graphic Abstract: An graphic abstract is available for this article.
Collapse
Affiliation(s)
- Nikolaos Kakogiannos
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.)
| | - Laura Ferrari
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.)
| | - Costanza Giampietro
- EMPA, Swiss Federal Laboratories for Material Science and Technologies, Experimental Continuum Mechanics, Dübendorf, Switzerland (C.G.)
| | - Anna Agata Scalise
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.)
| | - Claudio Maderna
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.)
| | - Micol Ravà
- Experimental Oncology (M.R.), European Institute of Oncology IRCSS, Milan
| | | | - Maria Grazia Lampugnani
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.).,Mario Negri Institute for Pharmacological Research, Milan (M.G.L.)
| | | | - Matteo Malinverno
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.)
| | - Emanuele Martini
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.)
| | - Ilaria Costa
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.)
| | - Michela Lupia
- Unit of Gynaecological Oncology Research (M.L., U.C.), European Institute of Oncology IRCSS, Milan
| | - Ugo Cavallaro
- Unit of Gynaecological Oncology Research (M.L., U.C.), European Institute of Oncology IRCSS, Milan
| | - Galina V Beznoussenko
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.)
| | - Alexander A Mironov
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.)
| | - Bethania Fernandes
- Pathology Unit, Humanitas Clinical and Research Centre, Rozzano, Milan (B.F., N.R.)
| | - Noemi Rudini
- Pathology Unit, Humanitas Clinical and Research Centre, Rozzano, Milan (B.F., N.R.)
| | - Elisabetta Dejana
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.).,Oncology and Haemato-Oncology, School of Medicine, University of Milan (E.D.).,Immunology, Genetics and Pathology, Uppsala University, Sweden (E.D.)
| | - Monica Giannotta
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.)
| |
Collapse
|
16
|
Czabanka M, Petrilli LL, Elvers-Hornung S, Bieback K, Albert Imhof B, Vajkoczy P, Vinci M. Junctional Adhesion Molecule-C Mediates the Recruitment of Embryonic-Endothelial Progenitor Cells to the Perivascular Niche during Tumor Angiogenesis. Int J Mol Sci 2020; 21:ijms21041209. [PMID: 32054130 PMCID: PMC7072851 DOI: 10.3390/ijms21041209] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 02/07/2020] [Indexed: 11/29/2022] Open
Abstract
The homing of Endothelial Progenitor Cells (EPCs) to tumor angiogenic sites has been described as a multistep process, involving adhesion, migration, incorporation and sprouting, for which the underlying molecular and cellular mechanisms are yet to be fully defined. Here, we studied the expression of Junctional Adhesion Molecule-C (JAM-C) by EPCs and its role in EPC homing to tumor angiogenic vessels. For this, we used mouse embryonic-Endothelial Progenitor Cells (e-EPCs), intravital multi-fluorescence microscopy techniques and the dorsal skin-fold chamber model. JAM-C was found to be expressed by e-EPCs and endothelial cells. Blocking JAM-C did not affect adhesion of e-EPCs to endothelial monolayers in vitro but, interestingly, it did reduce their adhesion to tumor endothelium in vivo. The most striking effect of JAM-C blocking was on tube formation on matrigel in vitro and the incorporation and sprouting of e-EPCs to tumor endothelium in vivo. Our results demonstrate that JAM-C mediates e-EPC recruitment to tumor angiogenic sites, i.e., coordinated homing of EPCs to the perivascular niche, where they cluster and interact with tumor blood vessels. This suggests that JAM-C plays a critical role in the process of vascular assembly and may represent a potential therapeutic target to control tumor angiogenesis.
Collapse
Affiliation(s)
- Marcus Czabanka
- Department of Neurosurgery, Universitätsmedizin Charitè, 10117 Berlin, Germany;
- Department of Neurosurgery Medical Faculty of the University of Heidelberg, 68167 Mannheim, Germany;
| | - Lucia Lisa Petrilli
- Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital – IRCCS, 00146 Rome, Italy;
| | - Susanne Elvers-Hornung
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Fred Cross Blood Donor Service Baden-Württemberg – Hessen, 68167 Mannheim, Germany (K.B.)
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Fred Cross Blood Donor Service Baden-Württemberg – Hessen, 68167 Mannheim, Germany (K.B.)
| | - Beat Albert Imhof
- Department of Pathology and Immunology, Medical Faculty, Centre Medical Universitaire (CMU), University of Geneva, 1206 Geneva, Switzerland;
| | - Peter Vajkoczy
- Department of Neurosurgery, Universitätsmedizin Charitè, 10117 Berlin, Germany;
- Department of Neurosurgery Medical Faculty of the University of Heidelberg, 68167 Mannheim, Germany;
- Correspondence: ; Tel.: +49-30450560-002
| | - Maria Vinci
- Department of Neurosurgery Medical Faculty of the University of Heidelberg, 68167 Mannheim, Germany;
- Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital – IRCCS, 00146 Rome, Italy;
| |
Collapse
|
17
|
Luissint AC, Williams HC, Kim W, Flemming S, Azcutia V, Hilgarth RS, Leary MNO, Denning TL, Nusrat A, Parkos CA. Macrophage-dependent neutrophil recruitment is impaired under conditions of increased intestinal permeability in JAM-A-deficient mice. Mucosal Immunol 2019; 12:668-678. [PMID: 30745566 PMCID: PMC6543824 DOI: 10.1038/s41385-019-0143-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 02/07/2023]
Abstract
Junctional adhesion molecule-A (JAM-A) is a transmembrane glycoprotein expressed on leukocytes, endothelia, and epithelia that regulates biological processes including barrier function and immune responses. While JAM-A has been reported to facilitate tissue infiltration of leukocytes under inflammatory conditions, the contributions of leukocyte-expressed JAM-A in vivo remain unresolved. We investigated the role of leukocyte-expressed JAM-A in acute peritonitis induced by zymosan, lipopolysaccharide (LPS), or TNFα using mice with selective loss of JAM-A in myelomonocytic cells (LysM-Cre;Jam-afl/fl). Surprisingly, in LysM-Cre;Jam-afl/fl mice, loss of JAM-A did not affect neutrophil (PMN) recruitment into the peritoneum in response to zymosan, LPS, or TNFα although it was significantly reduced in Jam-aKO mice. In parallel, Jam-aKO peritoneal macrophages exhibited diminished CXCL1 chemokine production and decreased activation of NF-kB, whereas those from LysM-Cre;Jam-afl/fl mice were unaffected. Using Villin-Cre;Jam-afl/fl mice, targeted loss of JAM-A on intestinal epithelial cells resulted in increased intestinal permeability along with reduced peritoneal PMN migration as well as lower levels of CXCL1 and active NF-kB similar to that observed in Jam-aKO animals. Interestingly, in germ-free Villin-Cre;Jam-afl/fl mice, PMN recruitment was unaffected suggesting dependence on gut microbiota. Such observations highlight the functional link between a leaky gut and regulation of innate immune responses.
Collapse
Affiliation(s)
| | - Holly C Williams
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, USA
| | - Wooki Kim
- Department of Food Science and Biotechnology, Kyung Hee University, Yongin, Korea
| | - Sven Flemming
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Veronica Azcutia
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Timothy L Denning
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Asma Nusrat
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Charles A Parkos
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
18
|
Shi J, Barakat M, Chen D, Chen L. Bicellular Tight Junctions and Wound Healing. Int J Mol Sci 2018; 19:ijms19123862. [PMID: 30518037 PMCID: PMC6321209 DOI: 10.3390/ijms19123862] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 12/15/2022] Open
Abstract
Bicellular tight junctions (TJs) are intercellular junctions comprised of a variety of transmembrane proteins including occludin, claudins, and junctional adhesion molecules (JAMs) as well as intracellular scaffold proteins such as zonula occludens (ZOs). TJs are functional, intercellular structures that form a barrier between adjacent cells, which constantly seals and unseals to control the paracellular passage of molecules. They are primarily present in the epithelial and endothelial cells of all tissues and organs. In addition to their well-recognized roles in maintaining cell polarity and barrier functions, TJs are important regulators of signal transduction, which modulates cell proliferation, migration, and differentiation, as well as some components of the immune response and homeostasis. A vast breadth of research data is available on TJs, but little has been done to decipher their specific roles in wound healing, despite their primary distribution in epithelial and endothelial cells, which are essential contributors to the wound healing process. Some data exists to indicate that a better understanding of the functions and significance of TJs in healing wounds may prove crucial for future improvements in wound healing research and therapy. Specifically, recent studies demonstrate that occludin and claudin-1, which are two TJ component proteins, are present in migrating epithelial cells at the wound edge but are absent in chronic wounds. This indicates that functional TJs may be critical for effective wound healing. A tremendous amount of work is needed to investigate their roles in barrier function, re-epithelialization, angiogenesis, scar formation, and in the interactions between epithelial cells, endothelial cells, and immune cells both in the acute wound healing process and in non-healing wounds. A more thorough understanding of TJs in wound healing may shed new light on potential research targets and reveal novel strategies to enhance tissue regeneration and improve wound repair.
Collapse
Affiliation(s)
- Junhe Shi
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, 801 S. Paulina Street, Chicago, IL 60612, USA.
| | - May Barakat
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, 801 S. Paulina Street, Chicago, IL 60612, USA.
| | - Dandan Chen
- Colgate-Palmolive Company, Piscataway, NJ 08855, USA.
| | - Lin Chen
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, 801 S. Paulina Street, Chicago, IL 60612, USA.
| |
Collapse
|
19
|
Flemming S, Luissint AC, Nusrat A, Parkos CA. Analysis of leukocyte transepithelial migration using an in vivo murine colonic loop model. JCI Insight 2018; 3:99722. [PMID: 30333307 DOI: 10.1172/jci.insight.99722] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 09/11/2018] [Indexed: 12/30/2022] Open
Abstract
Molecular mechanisms that control leukocyte migration across the vascular endothelium (transendothelial migration; TEndoM) have been extensively characterized in vivo, but details of leukocyte transepithelial migration (TEpM) and its dysregulation (a pathologic feature of many mucosal diseases) are missing due to the lack of suitable animal models. Here, we describe a murine model that utilizes a vascularized proximal colonic segment (pcLoop) and enables quantitative studies of leukocyte trafficking across colonic epithelium. Consistent with previous in vitro studies, intraluminal injection of antibodies against integrin CD11b/CD18 reduced recruitment of polymorphonuclear neutrophils (PMN) into the lumen of pcLoops, and it increased subepithelial accumulation of PMN. We extended studies using the pcLoop to determine contributions of Junctional Adhesion Molecule-A (JAM-A, or F11R) in PMN TEpM and confirmed that mice with total loss of JAM-A or mice with intestinal epithelial selective loss of JAM-A had increased colonic permeability. Furthermore, there was reduced PMN migration into the colonic lumen that paralleled subepithelial accumulation of PMN in global-KO mice, as well as in intestinal epithelial-targeted JAM-A-deficient mice. These findings highlight a potentially novel role for JAM-A in regulating PMN TEpM in vivo and demonstrate utility of this model for identifying receptors that may be targeted in vivo to reduce pathologic intestinal inflammation.
Collapse
|
20
|
Steinbacher T, Kummer D, Ebnet K. Junctional adhesion molecule-A: functional diversity through molecular promiscuity. Cell Mol Life Sci 2018; 75:1393-1409. [PMID: 29238845 PMCID: PMC11105642 DOI: 10.1007/s00018-017-2729-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/04/2017] [Accepted: 12/11/2017] [Indexed: 12/27/2022]
Abstract
Cell adhesion molecules (CAMs) of the immunoglobulin superfamily (IgSF) regulate important processes such as cell proliferation, differentiation and morphogenesis. This activity is primarily due to their ability to initiate intracellular signaling cascades at cell-cell contact sites. Junctional adhesion molecule-A (JAM-A) is an IgSF-CAM with a short cytoplasmic tail that has no catalytic activity. Nevertheless, JAM-A is involved in a variety of biological processes. The functional diversity of JAM-A resides to a large part in a C-terminal PDZ domain binding motif which directly interacts with nine different PDZ domain-containing proteins. The molecular promiscuity of its PDZ domain motif allows JAM-A to recruit protein scaffolds to specific sites of cell-cell adhesion and to assemble signaling complexes at those sites. Here, we review the molecular characteristics of JAM-A, including its dimerization, its interaction with scaffolding proteins, and the phosphorylation of its cytoplasmic domain, and we describe how these characteristics translate into diverse biological activities.
Collapse
Affiliation(s)
- Tim Steinbacher
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Von-Esmarch-Str. 56, 48149, Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Daniel Kummer
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Von-Esmarch-Str. 56, 48149, Münster, Germany
- Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany
| | - Klaus Ebnet
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Von-Esmarch-Str. 56, 48149, Münster, Germany.
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany.
- Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany.
| |
Collapse
|
21
|
A novel immunotoxin reveals a new role for CD321 in endothelial cells. PLoS One 2017; 12:e0181502. [PMID: 29028806 PMCID: PMC5640210 DOI: 10.1371/journal.pone.0181502] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 07/03/2017] [Indexed: 11/19/2022] Open
Abstract
There are currently several antibody therapies that directly target tumors, and antibody-drug conjugates represent a novel moiety as next generation therapeutics. Here, we used a unique screening probe, DT3C, to identify functional antibodies that recognized surface molecules and functional epitopes, and which provided toxin delivery capability. Accordingly, we generated the 90G4 antibody, which induced DT3C-dependent cytotoxicity in endothelial cells. Molecular analysis revealed that 90G4 recognized CD321, a protein localized at tight junctions. Although CD321 plays a pivotal role in inflammation and lymphocyte trans-endothelial migration, little is known about its mechanism of action in endothelial cells. Targeting of CD321 by the 90G4 immunotoxin induced cell death. Moreover, 90G4 immunotoxin caused cytotoxicity primarily in migratory endothelial cells, but not in those forming sheets, suggesting a critical role for CD321 in tumor angiogenesis. We also found that hypoxia triggered redistribution of CD321 to a punctate localization on the basal side of cells, resulting in functional impairment of tight junctions and increased motility. Thus, our findings raise the intriguing possibility that endothelial CD321 presented cellular localization in tight junction as well as multifunctional dynamics in several conditions, leading to illuminate the importance of widely-expressed CD321 as a potential target for antitumor therapy.
Collapse
|
22
|
Ebnet K. Junctional Adhesion Molecules (JAMs): Cell Adhesion Receptors With Pleiotropic Functions in Cell Physiology and Development. Physiol Rev 2017; 97:1529-1554. [PMID: 28931565 DOI: 10.1152/physrev.00004.2017] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 05/04/2017] [Accepted: 05/11/2017] [Indexed: 02/06/2023] Open
Abstract
Junctional adhesion molecules (JAM)-A, -B and -C are cell-cell adhesion molecules of the immunoglobulin superfamily which are expressed by a variety of tissues, both during development and in the adult organism. Through their extracellular domains, they interact with other adhesion receptors on opposing cells. Through their cytoplasmic domains, they interact with PDZ domain-containing scaffolding and signaling proteins. In combination, these two properties regulate the assembly of signaling complexes at specific sites of cell-cell adhesion. The multitude of molecular interactions has enabled JAMs to adopt distinct cellular functions such as the regulation of cell-cell contact formation, cell migration, or mitotic spindle orientation. Not surprisingly, JAMs regulate diverse processes such as epithelial and endothelial barrier formation, hemostasis, angiogenesis, hematopoiesis, germ cell development, and the development of the central and peripheral nervous system. This review summarizes the recent progress in the understanding of JAMs, including their characteristic structural features, their molecular interactions, their cellular functions, and their contribution to a multitude of processes during vertebrate development and homeostasis.
Collapse
Affiliation(s)
- Klaus Ebnet
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity", Institute of Medical Biochemistry, ZMBE, Cells-In-Motion Cluster of Excellence (EXC1003-CiM), and Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany
| |
Collapse
|
23
|
Briasoulis A, Androulakis E, Christophides T, Tousoulis D. The role of inflammation and cell death in the pathogenesis, progression and treatment of heart failure. Heart Fail Rev 2016; 21:169-76. [PMID: 26872673 DOI: 10.1007/s10741-016-9533-z] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chronic inflammation underlies a variety of seemingly unrelated conditions including coronary artery disease. The interest in exploring the role of inflammation in heart failure (CHF) arises from earlier observations that circulating pro-inflammatory biomarker levels are elevated in patients with both ischaemic and non-ischaemic cardiomyopathies and correlate with severity of disease and prognosis (McMurray et al. in Eur Heart J 33:1787-1847, 2012; Mosterd and Hoes in Heart 93:1137-1146, 2007; Owan et al. in New Engl J Med 355:251-259, 2006). In acute decompensated HF, pro-inflammatory biomarker levels have been associated with mortality and readmission rates (Cowie et al. in Heart 83:505-510, 2000). Similar to neurohormonal activation and inflammation, production of pro-inflammatory cytokines is a response to stress in an attempt to restore cellular function. However, sustained expression and exposure to cytokines can lead to left ventricular dysfunction, negative inotropic effects, altered cardiac metabolism, myocardial remodelling and HF progression. However, it is unclear whether elevated levels of pro-inflammatory biomarkers, such as high-sensitivity C-reactive protein, signify an ongoing inflammatory process that leads to HF progression, or are merely markers of advanced disease. Beta-blockers, renin-angiotensin-aldosterone axis antagonists, statins and immunosuppressants have been found to decrease the levels of cytokines in small clinical studies of patients with HF (Hobbs et al. in Heart J 28:1128-1134, 2007). However, 'immunomodulatory' approaches applied in the RECOVER, RENAISSANCE, ATTACH, IMAC and ACCLAIM double-blind, placebo-controlled studies had neutral or negative effects on outcomes of patients with HF. In the present review, we focus on the role of inflammation in pathogenesis and progression of the HF, the value of pro-inflammatory cytokines as biomarkers and the potential therapeutic applications of immunomodulation in HF patients.
Collapse
Affiliation(s)
- Alexandros Briasoulis
- Division of Cardiology, Detroit Medical Center, Wayne State University, Detroit, MI, USA.
| | - Emmanuel Androulakis
- Division of Cardiology, Detroit Medical Center, Wayne State University, Detroit, MI, USA.,1st Cardiology Department, Athens University Medical School, Athens, Greece
| | - Theodoros Christophides
- Division of Cardiology, Detroit Medical Center, Wayne State University, Detroit, MI, USA.,1st Cardiology Department, Athens University Medical School, Athens, Greece
| | - Dimitris Tousoulis
- Division of Cardiology, Detroit Medical Center, Wayne State University, Detroit, MI, USA.,1st Cardiology Department, Athens University Medical School, Athens, Greece
| |
Collapse
|
24
|
Bonaventura A, Montecucco F, Dallegri F. Cellular recruitment in myocardial ischaemia/reperfusion injury. Eur J Clin Invest 2016; 46:590-601. [PMID: 27090739 DOI: 10.1111/eci.12633] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 04/17/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND Myocardial infarction (MI) is strictly linked to atherosclerosis. Beyond the mechanical narrowing of coronary vessels lumen, during MI a great burden of inflammation is carried out. One of the crucial events is represented by the ischaemia/reperfusion injury, a complex event involving inflammatory cells (such as neutrophils, platelets, monocytes/macrophages, lymphocytes and mast cells) and key activating signals (such as cytokines, chemokines and growth factors). Cardiac repair following myocardial infarction is dependent on a finely regulated response involving a sequential recruitment and the clearance of different subsets of inflammatory cells. MATERIALS AND METHODS This narrative review was based on the works detected on PubMed and MEDLINE up to November 2015. RESULTS Infarct healing classically follows three overlapping phases: the inflammatory phase, in which the innate immune pathways are activated and inflammatory leucocytes are recruited in order to clear the wound from dead cells; the proliferative phase, characterized by the suppression of pro-inflammatory signalling and infiltration of 'repairing' cells secreting matrix proteins in the injured area; and the maturation phase, which is associated with the quiescence and the elimination of the reparative cells together with cross-linking of the matrix. All these phases are timely regulated by the production of soluble mediators, such as cytokines, chemokines and growth factors. CONCLUSION Targeting inflammatory cell recruitment early during reperfusion and healing might be promising to selectively inhibit injury and favour repair. This approach might substantially improve adverse postischaemic left ventricle remodelling, characterized by dilation, hypertrophy of viable segments and progressive dysfunction.
Collapse
Affiliation(s)
- Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.,IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.,IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.,IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| |
Collapse
|
25
|
Schmidt EP, Kuebler WM, Lee WL, Downey GP. Adhesion Molecules: Master Controllers of the Circulatory System. Compr Physiol 2016; 6:945-73. [PMID: 27065171 DOI: 10.1002/cphy.c150020] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This manuscript will review our current understanding of cellular adhesion molecules (CAMs) relevant to the circulatory system, their physiological role in control of vascular homeostasis, innate and adaptive immune responses, and their importance in pathophysiological (disease) processes such as acute lung injury, atherosclerosis, and pulmonary hypertension. This is a complex and rapidly changing area of research that is incompletely understood. By design, we will begin with a brief overview of the structure and classification of the major groups of adhesion molecules and their physiological functions including cellular adhesion and signaling. The role of specific CAMs in the process of platelet aggregation and hemostasis and leukocyte adhesion and transendothelial migration will be reviewed as examples of the complex and cooperative interplay between CAMs during physiological and pathophysiological processes. The role of the endothelial glycocalyx and the glycobiology of this complex system related to inflammatory states such as sepsis will be reviewed. We will then focus on the role of adhesion molecules in the pathogenesis of specific disease processes involving the lungs and cardiovascular system. The potential of targeting adhesion molecules in the treatment of immune and inflammatory diseases will be highlighted in the relevant sections throughout the manuscript.
Collapse
Affiliation(s)
- Eric P Schmidt
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Wolfgang M Kuebler
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Departments of Surgery and Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Warren L Lee
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Division of Respirology and the Interdepartmental Division of Critical Care Medicine, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Gregory P Downey
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Departments of Medicine, Pediatrics, and Biomedical Research, National Jewish Health, Denver, Colorado, USA
- Departments of Medicine, and Immunology and Microbiology, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
26
|
Abstract
Immune responses depend on the ability of leukocytes to move from the circulation into tissue. This is enabled by mechanisms that guide leukocytes to the right exit sites and allow them to cross the barrier of the blood vessel wall. This process is regulated by a concerted action between endothelial cells and leukocytes, whereby endothelial cells activate leukocytes and direct them to extravasation sites, and leukocytes in turn instruct endothelial cells to open a path for transmigration. This Review focuses on recently described mechanisms that control and open exit routes for leukocytes through the endothelial barrier.
Collapse
|
27
|
Ahmadsei M, Lievens D, Weber C, von Hundelshausen P, Gerdes N. Immune-mediated and lipid-mediated platelet function in atherosclerosis. Curr Opin Lipidol 2015; 26:438-48. [PMID: 26270811 DOI: 10.1097/mol.0000000000000212] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Cardiovascular disease (CVD) is the leading cause of death and morbidity worldwide. Detailed knowledge of the mechanisms of atherosclerosis, the main underlying disease of CVD, will enable improved preventive and therapeutic options, thus potentially limiting the burden of vascular disease in aging societies. A large body of evidence illustrates the contribution of platelets to processes beyond their traditionally recognized role as mediators in thrombosis and hemostasis. Recent advances in molecular biology help to understand the complexity of atherosclerosis. RECENT FINDINGS This article outlines the role of platelets as modulators of immune responses in the context of atherosclerosis. It provides a short overview of interactions between platelets and endothelial cells or immune cells via direct cell contact or soluble factors during atherogenesis. By means of some well examined, exemplary pathways (e.g. CD40/CD40L dyad), this article will discuss recent discoveries in immune-related function of platelets. We also focus on the relationship between platelets and the lipid metabolism highlighting potential consequences to atherosclerosis and dyslipidemia. SUMMARY A better understanding of the molecular mechanisms of platelet-related immune activity allows their utilization as powerful diagnostic tools or targets of therapeutic intervention. Those findings might help to develop new classes of drugs which may supplement or replace classical anticoagulants and help clinicians to tackle CVD more efficiently.
Collapse
Affiliation(s)
- Maiwand Ahmadsei
- aInstitute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany bDZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | | | | | | | | |
Collapse
|
28
|
Abstract
Myocardial infarction is defined as sudden ischemic death of myocardial tissue. In the clinical context, myocardial infarction is usually due to thrombotic occlusion of a coronary vessel caused by rupture of a vulnerable plaque. Ischemia induces profound metabolic and ionic perturbations in the affected myocardium and causes rapid depression of systolic function. Prolonged myocardial ischemia activates a "wavefront" of cardiomyocyte death that extends from the subendocardium to the subepicardium. Mitochondrial alterations are prominently involved in apoptosis and necrosis of cardiomyocytes in the infarcted heart. The adult mammalian heart has negligible regenerative capacity, thus the infarcted myocardium heals through formation of a scar. Infarct healing is dependent on an inflammatory cascade, triggered by alarmins released by dying cells. Clearance of dead cells and matrix debris by infiltrating phagocytes activates anti-inflammatory pathways leading to suppression of cytokine and chemokine signaling. Activation of the renin-angiotensin-aldosterone system and release of transforming growth factor-β induce conversion of fibroblasts into myofibroblasts, promoting deposition of extracellular matrix proteins. Infarct healing is intertwined with geometric remodeling of the chamber, characterized by dilation, hypertrophy of viable segments, and progressive dysfunction. This review manuscript describes the molecular signals and cellular effectors implicated in injury, repair, and remodeling of the infarcted heart, the mechanistic basis of the most common complications associated with myocardial infarction, and the pathophysiologic effects of established treatment strategies. Moreover, we discuss the implications of pathophysiological insights in design and implementation of new promising therapeutic approaches for patients with myocardial infarction.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
29
|
Karshovska E, Zhao Z, Blanchet X, Schmitt MMN, Bidzhekov K, Soehnlein O, von Hundelshausen P, Mattheij NJ, Cosemans JMEM, Megens RTA, Koeppel TA, Schober A, Hackeng TM, Weber C, Koenen RR. Hyperreactivity of junctional adhesion molecule A-deficient platelets accelerates atherosclerosis in hyperlipidemic mice. Circ Res 2014; 116:587-99. [PMID: 25472975 DOI: 10.1161/circresaha.116.304035] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
RATIONALE Besides their essential role in hemostasis, platelets also have functions in inflammation. In platelets, junctional adhesion molecule (JAM)-A was previously identified as an inhibitor of integrin αIIbβ3-mediated outside-in signaling and its genetic knockdown resulted in hyperreactivity. OBJECTIVE This gain-of-function was specifically exploited to investigate the role of platelet hyperreactivity in plaque development. METHODS AND RESULTS JAM-A-deficient platelets showed increased aggregation and cellular and sarcoma tyrosine-protein kinase activation. On αIIbβ3 ligation, JAM-A was shown to be dephosphorylated, which could be prevented by protein tyrosine phosphatase nonreceptor type 1 inhibition. Mice with or without platelet-specific (tr)JAM-A-deficiency in an apolipoprotein e (apoe(-/-)) background were fed a high-fat diet. After ≤12 weeks of diet, trJAM-A(-/-)apoe-/- mice showed increased aortic plaque formation when compared with trJAM-A(+/+) apoe(-/-) controls, and these differences were most evident at early time points. At 2 weeks, the plaques of the trJAM-A(-/-) apoe(-/-) animals revealed increased macrophage, T cell, and smooth muscle cell content. Interestingly, plasma levels of chemokines CC chemokine ligand 5 and CXC-chemokine ligand 4 were increased in the trJAM-A(-/-) apoe(-/-)mice, and JAM-A-deficient platelets showed increased binding to monocytes and neutrophils. Whole-blood perfusion experiments and intravital microscopy revealed increased recruitment of platelets and monocytes to the inflamed endothelium in blood of trJAM-A(-/-) apoe(-/-)mice. Notably, these proinflammatory effects of JAM-A-deficient platelets could be abolished by the inhibition of αIIbβ3 signaling in vitro. CONCLUSIONS Deletion of JAM-A causes a gain-of-function in platelets, with lower activation thresholds and increased inflammatory activities. This leads to an increase of plaque formation, particularly in early stages of the disease.
Collapse
Affiliation(s)
- Ela Karshovska
- From the Institute for Cardiovascular Prevention (IPEK) (E.K., Z.Z., X.B., M.M.N.S., K.B., O.S., P.v.H., R.T.A.M., A.S., C.W., R.R.K.) and Division of Vascular and Endovascular Surgery (Z.Z., T.A.K.), Ludwig-Maximilians-University Munich, Munich, Germany; Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands (O.S.); German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (O.S., P.v.H., A.S., C.W.); and Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (N.J.M., J.M.E.M.C., R.T.A.M., T.M.H., C.W., R.R.K.)
| | - Zhen Zhao
- From the Institute for Cardiovascular Prevention (IPEK) (E.K., Z.Z., X.B., M.M.N.S., K.B., O.S., P.v.H., R.T.A.M., A.S., C.W., R.R.K.) and Division of Vascular and Endovascular Surgery (Z.Z., T.A.K.), Ludwig-Maximilians-University Munich, Munich, Germany; Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands (O.S.); German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (O.S., P.v.H., A.S., C.W.); and Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (N.J.M., J.M.E.M.C., R.T.A.M., T.M.H., C.W., R.R.K.)
| | - Xavier Blanchet
- From the Institute for Cardiovascular Prevention (IPEK) (E.K., Z.Z., X.B., M.M.N.S., K.B., O.S., P.v.H., R.T.A.M., A.S., C.W., R.R.K.) and Division of Vascular and Endovascular Surgery (Z.Z., T.A.K.), Ludwig-Maximilians-University Munich, Munich, Germany; Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands (O.S.); German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (O.S., P.v.H., A.S., C.W.); and Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (N.J.M., J.M.E.M.C., R.T.A.M., T.M.H., C.W., R.R.K.)
| | - Martin M N Schmitt
- From the Institute for Cardiovascular Prevention (IPEK) (E.K., Z.Z., X.B., M.M.N.S., K.B., O.S., P.v.H., R.T.A.M., A.S., C.W., R.R.K.) and Division of Vascular and Endovascular Surgery (Z.Z., T.A.K.), Ludwig-Maximilians-University Munich, Munich, Germany; Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands (O.S.); German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (O.S., P.v.H., A.S., C.W.); and Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (N.J.M., J.M.E.M.C., R.T.A.M., T.M.H., C.W., R.R.K.)
| | - Kiril Bidzhekov
- From the Institute for Cardiovascular Prevention (IPEK) (E.K., Z.Z., X.B., M.M.N.S., K.B., O.S., P.v.H., R.T.A.M., A.S., C.W., R.R.K.) and Division of Vascular and Endovascular Surgery (Z.Z., T.A.K.), Ludwig-Maximilians-University Munich, Munich, Germany; Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands (O.S.); German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (O.S., P.v.H., A.S., C.W.); and Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (N.J.M., J.M.E.M.C., R.T.A.M., T.M.H., C.W., R.R.K.)
| | - Oliver Soehnlein
- From the Institute for Cardiovascular Prevention (IPEK) (E.K., Z.Z., X.B., M.M.N.S., K.B., O.S., P.v.H., R.T.A.M., A.S., C.W., R.R.K.) and Division of Vascular and Endovascular Surgery (Z.Z., T.A.K.), Ludwig-Maximilians-University Munich, Munich, Germany; Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands (O.S.); German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (O.S., P.v.H., A.S., C.W.); and Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (N.J.M., J.M.E.M.C., R.T.A.M., T.M.H., C.W., R.R.K.)
| | - Philipp von Hundelshausen
- From the Institute for Cardiovascular Prevention (IPEK) (E.K., Z.Z., X.B., M.M.N.S., K.B., O.S., P.v.H., R.T.A.M., A.S., C.W., R.R.K.) and Division of Vascular and Endovascular Surgery (Z.Z., T.A.K.), Ludwig-Maximilians-University Munich, Munich, Germany; Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands (O.S.); German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (O.S., P.v.H., A.S., C.W.); and Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (N.J.M., J.M.E.M.C., R.T.A.M., T.M.H., C.W., R.R.K.)
| | - Nadine J Mattheij
- From the Institute for Cardiovascular Prevention (IPEK) (E.K., Z.Z., X.B., M.M.N.S., K.B., O.S., P.v.H., R.T.A.M., A.S., C.W., R.R.K.) and Division of Vascular and Endovascular Surgery (Z.Z., T.A.K.), Ludwig-Maximilians-University Munich, Munich, Germany; Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands (O.S.); German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (O.S., P.v.H., A.S., C.W.); and Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (N.J.M., J.M.E.M.C., R.T.A.M., T.M.H., C.W., R.R.K.)
| | - Judith M E M Cosemans
- From the Institute for Cardiovascular Prevention (IPEK) (E.K., Z.Z., X.B., M.M.N.S., K.B., O.S., P.v.H., R.T.A.M., A.S., C.W., R.R.K.) and Division of Vascular and Endovascular Surgery (Z.Z., T.A.K.), Ludwig-Maximilians-University Munich, Munich, Germany; Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands (O.S.); German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (O.S., P.v.H., A.S., C.W.); and Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (N.J.M., J.M.E.M.C., R.T.A.M., T.M.H., C.W., R.R.K.)
| | - Remco T A Megens
- From the Institute for Cardiovascular Prevention (IPEK) (E.K., Z.Z., X.B., M.M.N.S., K.B., O.S., P.v.H., R.T.A.M., A.S., C.W., R.R.K.) and Division of Vascular and Endovascular Surgery (Z.Z., T.A.K.), Ludwig-Maximilians-University Munich, Munich, Germany; Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands (O.S.); German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (O.S., P.v.H., A.S., C.W.); and Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (N.J.M., J.M.E.M.C., R.T.A.M., T.M.H., C.W., R.R.K.)
| | - Thomas A Koeppel
- From the Institute for Cardiovascular Prevention (IPEK) (E.K., Z.Z., X.B., M.M.N.S., K.B., O.S., P.v.H., R.T.A.M., A.S., C.W., R.R.K.) and Division of Vascular and Endovascular Surgery (Z.Z., T.A.K.), Ludwig-Maximilians-University Munich, Munich, Germany; Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands (O.S.); German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (O.S., P.v.H., A.S., C.W.); and Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (N.J.M., J.M.E.M.C., R.T.A.M., T.M.H., C.W., R.R.K.)
| | - Andreas Schober
- From the Institute for Cardiovascular Prevention (IPEK) (E.K., Z.Z., X.B., M.M.N.S., K.B., O.S., P.v.H., R.T.A.M., A.S., C.W., R.R.K.) and Division of Vascular and Endovascular Surgery (Z.Z., T.A.K.), Ludwig-Maximilians-University Munich, Munich, Germany; Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands (O.S.); German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (O.S., P.v.H., A.S., C.W.); and Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (N.J.M., J.M.E.M.C., R.T.A.M., T.M.H., C.W., R.R.K.)
| | - Tilman M Hackeng
- From the Institute for Cardiovascular Prevention (IPEK) (E.K., Z.Z., X.B., M.M.N.S., K.B., O.S., P.v.H., R.T.A.M., A.S., C.W., R.R.K.) and Division of Vascular and Endovascular Surgery (Z.Z., T.A.K.), Ludwig-Maximilians-University Munich, Munich, Germany; Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands (O.S.); German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (O.S., P.v.H., A.S., C.W.); and Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (N.J.M., J.M.E.M.C., R.T.A.M., T.M.H., C.W., R.R.K.)
| | - Christian Weber
- From the Institute for Cardiovascular Prevention (IPEK) (E.K., Z.Z., X.B., M.M.N.S., K.B., O.S., P.v.H., R.T.A.M., A.S., C.W., R.R.K.) and Division of Vascular and Endovascular Surgery (Z.Z., T.A.K.), Ludwig-Maximilians-University Munich, Munich, Germany; Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands (O.S.); German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (O.S., P.v.H., A.S., C.W.); and Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (N.J.M., J.M.E.M.C., R.T.A.M., T.M.H., C.W., R.R.K.)
| | - Rory R Koenen
- From the Institute for Cardiovascular Prevention (IPEK) (E.K., Z.Z., X.B., M.M.N.S., K.B., O.S., P.v.H., R.T.A.M., A.S., C.W., R.R.K.) and Division of Vascular and Endovascular Surgery (Z.Z., T.A.K.), Ludwig-Maximilians-University Munich, Munich, Germany; Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands (O.S.); German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (O.S., P.v.H., A.S., C.W.); and Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (N.J.M., J.M.E.M.C., R.T.A.M., T.M.H., C.W., R.R.K.).
| |
Collapse
|
30
|
Liu ML, Nagai T, Tokunaga M, Iwanaga K, Matsuura K, Takahashi T, Kanda M, Kondo N, Naito AT, Komuro I, Kobayashi Y. Anti-inflammatory peptides from cardiac progenitors ameliorate dysfunction after myocardial infarction. J Am Heart Assoc 2014; 3:e001101. [PMID: 25468657 PMCID: PMC4338698 DOI: 10.1161/jaha.114.001101] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Cardiac cell therapy has been proposed as one of the new strategies against myocardial infarction. Although several reports showed improvement of the function of ischemic heart, the effects of cell therapy vary among the studies and the mechanisms of the beneficial effects are still unknown. Previously, we reported that clonal stem cell antigen‐1–positive cardiac progenitor cells exerted a therapeutic effect when transplanted into the ischemic heart. Our aims were to identify the cardiac progenitor‐specific paracrine factor and to elucidate the mechanism of its beneficial effect. Methods and Results By using an antibody array, we found that soluble junctional adhesion molecule‐A (JAM‐A) was abundantly secreted from cardiac progenitor cells. Pretreatment of neutrophils with conditioned medium from cultured cardiac progenitor cells or soluble JAM‐A inhibited transendothelial migration and reduced motility of neutrophils. These inhibitory effects were attenuated by anti–JAM‐A neutralizing antibody. Injection of cardiac progenitor cells into infarct heart attenuated neutrophil infiltration and expression of inflammatory cytokines. Injection of soluble JAM‐A–expressing, but not of JAM‐A siRNA–expressing, cardiac progenitor cells into the infarct heart prevented cardiac remodeling and reduced fibrosis area. Conclusions Soluble JAM‐A secreted from cardiac progenitor cells reduces infiltration of neutrophils after myocardial infarction and ameliorates tissue damage through prevention of excess inflammation. Our finding may lead to a new therapy for cardiovascular disease by using the anti‐inflammatory effect of JAM‐A.
Collapse
Affiliation(s)
- Mei-Lan Liu
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan (M.L.L., T.N., M.T., K.I., T.T., M.K., N.K., Y.K.)
| | - Toshio Nagai
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan (M.L.L., T.N., M.T., K.I., T.T., M.K., N.K., Y.K.)
| | - Masakuni Tokunaga
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan (M.L.L., T.N., M.T., K.I., T.T., M.K., N.K., Y.K.)
| | - Koji Iwanaga
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan (M.L.L., T.N., M.T., K.I., T.T., M.K., N.K., Y.K.)
| | - Katsuhisa Matsuura
- Department of Cardiology and Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan (K.M.)
| | - Toshinao Takahashi
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan (M.L.L., T.N., M.T., K.I., T.T., M.K., N.K., Y.K.)
| | - Masato Kanda
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan (M.L.L., T.N., M.T., K.I., T.T., M.K., N.K., Y.K.)
| | - Naomichi Kondo
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan (M.L.L., T.N., M.T., K.I., T.T., M.K., N.K., Y.K.)
| | - Atsuhiko T Naito
- Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Tokyo, Japan (A.T.N., I.K.)
| | - Issei Komuro
- Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Tokyo, Japan (A.T.N., I.K.)
| | - Yoshio Kobayashi
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan (M.L.L., T.N., M.T., K.I., T.T., M.K., N.K., Y.K.)
| |
Collapse
|
31
|
The immune system and the remodeling infarcted heart: cell biological insights and therapeutic opportunities. J Cardiovasc Pharmacol 2014; 63:185-95. [PMID: 24072174 DOI: 10.1097/fjc.0000000000000003] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Extensive necrosis of ischemic cardiomyocytes in the infarcted myocardium activates the innate immune response triggering an intense inflammatory reaction. Release of danger signals from dying cells and damaged matrix activates the complement cascade and stimulates Toll-like receptor/interleukin-1 signaling, resulting in the activation of the nuclear factor-κB system and induction of chemokines, cytokines, and adhesion molecules. Subsequent infiltration of the infarct with neutrophils and mononuclear cells serves to clear the wound from dead cells and matrix debris, while stimulating reparative pathways. In addition to its role in repair of the infarcted heart and formation of a scar, the immune system is also involved in adverse remodeling of the infarcted ventricle. Overactive immune responses and defects in suppression, containment, and resolution of the postinfarction inflammatory reaction accentuate dilative remodeling in experimental models and may be associated with chamber dilation, systolic dysfunction, and heart failure in patients surviving a myocardial infarction. Interventions targeting the inflammatory response to attenuate adverse remodeling may hold promise in patients with myocardial infarction that exhibit accentuated, prolonged, or dysregulated immune responses to the acute injury.
Collapse
|
32
|
Zhang J, Zhang M, Sun L. Junctional adhesion molecule A of red drum (Sciaenops ocellatus): a possible immunomodulator and a target for bacterial immune evasion. Vet Immunol Immunopathol 2014; 161:99-107. [PMID: 25108665 DOI: 10.1016/j.vetimm.2014.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 07/18/2014] [Indexed: 12/01/2022]
Abstract
Junctional adhesion molecules (JAMs) are a family of type I cell surface receptors with two immunoglobulin (Ig) domains in the extracellular region. The family contains three classical members, i.e., JAM-A, -B, and -C. To date very little is known about the function of JAMs in teleost. In this work, we identified a JAM-A homologue (named SoJAMa) from red drum (Sciaenops ocellatus) and examined its expression and biological property. SoJAMa is composed of 347 amino acid residues and was predicted to be a transmembrane protein with a large extracellular region that contains two Ig domains. SoJAMa expression occurred in multiple tissues, in particular immune relevant organs. SoJAMa expression was downregulated by experimental challenge with an extracellular pathogen but upregulated by challenge with an intracellular pathogen that is known to be capable of immune evasion. Likewise, cellular study showed that infection of peripheral blood leukocytes (PBL) with intracellular pathogen induced significantly higher expression of SoJAMa. Immunofluorescence microscopy showed that SoJAMa was localized on the surface of PBL and recognized by antibodies against recombinant SoJAMa. Blockage of the SoJAMa on PBL with antibodies resulted in augmented respiratory burst activity. Consistently, antibody-treated PBL exhibited enhanced resistance against bacterial infection. Taken together, these results suggest for the first time that a teleost JAM-A likely possesses immunoregulatory property in a negative manner, and that this property may be taken advantage of by intracellular pathogens as an invasion strategy.
Collapse
Affiliation(s)
- Jian Zhang
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Graduate University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Min Zhang
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; College of Marine Science and Engineering, Qingdao Agricultural University, Chengyang, Qingdao 266109, China
| | - Li Sun
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China.
| |
Collapse
|
33
|
Luissint AC, Nusrat A, Parkos CA. JAM-related proteins in mucosal homeostasis and inflammation. Semin Immunopathol 2014; 36:211-26. [PMID: 24667924 DOI: 10.1007/s00281-014-0421-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 02/25/2014] [Indexed: 02/06/2023]
Abstract
Mucosal surfaces are lined by epithelial cells that form a physical barrier protecting the body against external noxious substances and pathogens. At a molecular level, the mucosal barrier is regulated by tight junctions (TJs) that seal the paracellular space between adjacent epithelial cells. Transmembrane proteins within TJs include junctional adhesion molecules (JAMs) that belong to the cortical thymocyte marker for Xenopus family of proteins. JAM family encompasses three classical members (JAM-A, JAM-B, and JAM-C) and related molecules including JAM4, JAM-like protein, Coxsackie and adenovirus receptor (CAR), CAR-like membrane protein and endothelial cell-selective adhesion molecule. JAMs have multiple functions that include regulation of endothelial and epithelial paracellular permeability, leukocyte recruitment during inflammation, angiogenesis, cell migration, and proliferation. In this review, we summarize the current knowledge regarding the roles of the JAM family members in the regulation of mucosal homeostasis and leukocyte trafficking with a particular emphasis on barrier function and its perturbation during pathological inflammation.
Collapse
Affiliation(s)
- Anny-Claude Luissint
- Epithelial pathobiology and mucosal inflammation research unit, Department of Pathology and Laboratory Medicine, Emory University, 615 Michael Street, 30306, Atlanta, GA, USA
| | | | | |
Collapse
|
34
|
Sladojevic N, Stamatovic SM, Keep RF, Grailer JJ, Sarma JV, Ward PA, Andjelkovic AV. Inhibition of junctional adhesion molecule-A/LFA interaction attenuates leukocyte trafficking and inflammation in brain ischemia/reperfusion injury. Neurobiol Dis 2014; 67:57-70. [PMID: 24657919 DOI: 10.1016/j.nbd.2014.03.010] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 02/27/2014] [Accepted: 03/13/2014] [Indexed: 11/15/2022] Open
Abstract
Proinflammatory mediators trigger intensive postischemic inflammatory remodeling of the blood-brain barrier (BBB) including extensive brain endothelial cell surface and junctional complex changes. Junctional adhesion molecule-A (JAM-A) is a component of the brain endothelial junctional complex with dual roles: paracellular route occlusion and regulating leukocyte docking and migration. The current study examined the contribution of JAM-A to the regulation of leukocyte (neutrophils and monocytes/macrophages) infiltration and the postischemic inflammatory response in brain ischemia/reperfusion (I/R injury). Brain I/R injury was induced by transient middle cerebral artery occlusion (MCAO) for 30min in mice followed by reperfusion for 0-5days, during which time JAM-A antagonist peptide (JAM-Ap) was administered. The peptide, which inhibits JAM-A/leukocyte interaction by blocking the interaction of the C2 domain of JAM-A with LFA on neutrophils and monocytes/macrophages, attenuated I/R-induced neutrophil and monocyte infiltration into brain parenchyma. Consequently, mice treated with JAM-A peptide during reperfusion had reduced expression (~3-fold) of inflammatory mediators in the ischemic penumbra, reduced infarct size (94±39 vs 211±38mm3) and significantly improved neurological score. BBB hyperpermeability was also reduced. Collectively, these results indicate that JAM-A has a prominent role in regulating leukocyte infiltration after brain I/R injury and could be a new target in limiting post-ischemic inflammation.
Collapse
Affiliation(s)
- Nikola Sladojevic
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Svetlana M Stamatovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jamison J Grailer
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - J Vidya Sarma
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Peter A Ward
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Anuska V Andjelkovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
35
|
Giannotta M, Benedetti S, Tedesco FS, Corada M, Trani M, D'Antuono R, Millet Q, Orsenigo F, Gálvez BG, Cossu G, Dejana E. Targeting endothelial junctional adhesion molecule-A/ EPAC/ Rap-1 axis as a novel strategy to increase stem cell engraftment in dystrophic muscles. EMBO Mol Med 2013; 6:239-58. [PMID: 24378569 PMCID: PMC3927958 DOI: 10.1002/emmm.201302520] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Muscular dystrophies are severe genetic diseases for which no efficacious therapies exist. Experimental clinical treatments include intra-arterial administration of vessel-associated stem cells, called mesoangioblasts (MABs). However, one of the limitations of this approach is the relatively low number of cells that engraft the diseased tissue, due, at least in part, to the sub-optimal efficiency of extravasation, whose mechanisms for MAB are unknown. Leukocytes emigrate into the inflamed tissues by crossing endothelial cell-to-cell junctions and junctional proteins direct and control leukocyte diapedesis. Here, we identify the endothelial junctional protein JAM-A as a key regulator of MAB extravasation. We show that JAM-A gene inactivation and JAM-A blocking antibodies strongly enhance MAB engraftment in dystrophic muscle. In the absence of JAM-A, the exchange factors EPAC-1 and 2 are down-regulated, which prevents the activation of the small GTPase Rap-1. As a consequence, junction tightening is reduced, allowing MAB diapedesis. Notably, pharmacological inhibition of Rap-1 increases MAB engraftment in dystrophic muscle, which results into a significant improvement of muscle function offering a novel strategy for stem cell-based therapies.
Collapse
Affiliation(s)
- Monica Giannotta
- FIRC Institute of Molecular Oncology Foundation (IFOM), Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Many viruses cause disease within an infected host after spread from an initial portal of entry to sites of secondary replication. Viruses can disseminate via the bloodstream or through nerves. Mammalian orthoreoviruses (reoviruses) are neurotropic viruses that use both bloodborne and neural pathways to spread systemically within their hosts to cause disease. Using a robust mouse model and a dynamic reverse genetics system, we have identified a viral receptor and a viral nonstructural protein that are essential for hematogenous reovirus dissemination. Junctional adhesion molecule-A (JAM-A) is a member of the immunoglobulin superfamily expressed in tight junctions and on hematopoietic cells that serves as a receptor for all reovirus serotypes. Expression of JAM-A is required for infection of endothelial cells and development of viremia in mice, suggesting that release of virus into the bloodstream from infected endothelial cells requires JAM-A. Nonstructural protein σ1s is implicated in cell cycle arrest and apoptosis in reovirus-infected cells but is completely dispensable for reovirus replication in cultured cells. Surprisingly, a recombinant σ1s-null reovirus strain fails to spread hematogenously in infected mice, suggesting that σ1s facilitates apoptosis of reovirus-infected intestinal epithelial cells. It is possible that apoptotic bodies formed as a consequence of σ1s expression lead to reovirus uptake by dendritic cells for subsequent delivery to the mesenteric lymph node and the blood. Thus, both host and viral factors are required for efficient hematogenous dissemination of reovirus. Understanding mechanisms of reovirus bloodborne spread may shed light on how microbial pathogens invade the bloodstream to disseminate and cause disease in infected hosts.
Collapse
Affiliation(s)
- Karl W Boehme
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | | |
Collapse
|
37
|
Schmitt MMN, Megens RTA, Zernecke A, Bidzhekov K, van den Akker NM, Rademakers T, van Zandvoort MA, Hackeng TM, Koenen RR, Weber C. Endothelial junctional adhesion molecule-a guides monocytes into flow-dependent predilection sites of atherosclerosis. Circulation 2013; 129:66-76. [PMID: 24065611 DOI: 10.1161/circulationaha.113.004149] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Junctional adhesion molecule (JAM)-A expressed in endothelial, epithelial, and blood cells can regulate permeability and leukocyte extravasation. Atherosclerosis develops at sites of disturbed flow in large arteries, but the mechanisms guiding inflammatory cells into these predilection sites remain unknown. METHODS AND RESULTS To characterize cell-specific functions of JAM-A in atherosclerosis, we used apolipoprotein E-deficient mice with a somatic or endothelium-specific deficiency in JAM-A and bone marrow chimeras with JAM-A-deficient leukocytes. We show that impaired JAM-A expression in endothelial cells reduced mononuclear cell recruitment into the arterial wall and limited atherosclerotic lesion formation in hyperlipidemic mice. In contrast, JAM-A deficiency in bone marrow cells impeded monocyte de-adhesion, thereby increasing vascular permeability and lesion formation, whereas somatic JAM-A deletion revealed no significant effects. Regions with disturbed flow displayed a focal enrichment and luminal redistribution of endothelial JAM-A and were preferentially protected by its deficiency. The functional expression and redistribution of endothelial JAM-A was increased by oxidized low-density lipoprotein, but confined by atheroprotective laminar flow through an upregulation of microRNA (miR)-145, which repressed JAM-A. CONCLUSIONS Our data identify endothelial JAM-A as an important effector molecule integrating atherogenic conditions to direct inflammatory cell entry at predilection sites of atherosclerosis.
Collapse
Affiliation(s)
- Martin M N Schmitt
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (M.M.N.S., R.T.A.M., K.B., R.R.K., C.W.); the Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany (M.M.N.S., M.A.v.Z.); the Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (M.M.N.S., R.T.A.M., N.M.v.d.A., T.R., M.A.v.Z., T.M.H., R.R.K., C.W.); the Division of Vascular Biology, Department of Vascular Surgery, Klinikum rechts der Isar, Technical University Munich, Germany (A.Z.); and the German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany (A.Z., C.W.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Christia P, Frangogiannis NG. Targeting inflammatory pathways in myocardial infarction. Eur J Clin Invest 2013; 43:986-95. [PMID: 23772948 PMCID: PMC3745791 DOI: 10.1111/eci.12118] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 05/21/2013] [Indexed: 12/14/2022]
Abstract
Acute cardiomyocyte necrosis in the infarcted heart generates damage-associated molecular patterns (DAMPs), activating complement and Toll-Like Receptor (TLR)/Interleukin (IL)-1 signalling and triggering an intense inflammatory reaction. Infiltrating leucocytes clear the infarct from dead cells, while activating reparative pathways that lead to formation of a scar. As the infarct heals the ventricle remodels, the geometric, functional and molecular alterations associated with postinfarction remodelling are driven by the inflammatory cascade and are involved in the development of heart failure. Because unrestrained inflammation in the infarcted heart induces matrix degradation and cardiomyocyte apoptosis, timely suppression of the postinfarction inflammatory reaction may be crucial to protect the myocardium from dilative remodelling and progressive dysfunction. Inhibition and resolution of postinfarction inflammation involve mobilization of inhibitory mononuclear cell subsets and require activation of endogenous STOP signals. Our manuscript discusses the basic cellular and molecular events involved in initiation, activation and resolution of the postinfarction inflammatory response, focusing on identification of therapeutic targets. The failure of anti-integrin approaches in patients with myocardial infarction and a growing body of experimental evidence suggest that inflammation may not increase ischaemic cardiomyocyte death, but accentuates matrix degradation causing dilative remodelling. Given the pathophysiologic complexity of postinfarction remodelling, personalized biomarker-based approaches are needed to target patient subpopulations with dysregulated inflammatory and reparative responses. Inhibition of pro-inflammatory signals (such as IL-1 and monocyte chemoattractant protein-1) may be effective in patients with defective resolution of postinfarction inflammation who exhibit progressive dilative remodelling. In contrast, patients with predominant hypertrophic/fibrotic responses may benefit from anti-TGF strategies.
Collapse
Affiliation(s)
- Panagiota Christia
- Department of Medicine, Albert Einstein College of Medicine, The Wilf Family Cardiovascular Research Institute, Bronx, NY, USA
| | | |
Collapse
|
39
|
Raccosta L, Fontana R, Maggioni D, Lanterna C, Villablanca EJ, Paniccia A, Musumeci A, Chiricozzi E, Trincavelli ML, Daniele S, Martini C, Gustafsson JA, Doglioni C, Feo SG, Leiva A, Ciampa MG, Mauri L, Sensi C, Prinetti A, Eberini I, Mora JR, Bordignon C, Steffensen KR, Sonnino S, Sozzani S, Traversari C, Russo V. The oxysterol-CXCR2 axis plays a key role in the recruitment of tumor-promoting neutrophils. J Exp Med 2013; 210:1711-28. [PMID: 23897983 PMCID: PMC3754872 DOI: 10.1084/jem.20130440] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 06/21/2013] [Indexed: 12/19/2022] Open
Abstract
Tumor-infiltrating immune cells can be conditioned by molecules released within the microenvironment to thwart antitumor immune responses, thereby facilitating tumor growth. Among immune cells, neutrophils play an important protumorigenic role by favoring neoangiogenesis and/or by suppressing antitumor immune responses. Tumor-derived oxysterols have recently been shown to favor tumor growth by inhibiting dendritic cell migration toward lymphoid organs. We report that tumor-derived oxysterols recruit protumor neutrophils in a liver X receptor (LXR)-independent, CXCR2-dependent manner, thus favoring tumor growth by promoting neoangiogenesis and immunosuppression. We demonstrate that interfering with the oxysterol-CXCR2 axis delays tumor growth and prolongs the overall survival of tumor-bearing mice. These results identify an unanticipated protumor function of the oxysterol-CXCR2 axis and a possible target for cancer therapy.
Collapse
Affiliation(s)
- Laura Raccosta
- Cancer Gene Therapy Unit, Program of Immunology and Bio Immuno Gene Therapy of Cancer, Division of Molecular Oncology, and Department of Pathology, Scientific Institute San Raffaele, 20132 Milan, Italy
| | - Raffaella Fontana
- Cancer Gene Therapy Unit, Program of Immunology and Bio Immuno Gene Therapy of Cancer, Division of Molecular Oncology, and Department of Pathology, Scientific Institute San Raffaele, 20132 Milan, Italy
| | - Daniela Maggioni
- Cancer Gene Therapy Unit, Program of Immunology and Bio Immuno Gene Therapy of Cancer, Division of Molecular Oncology, and Department of Pathology, Scientific Institute San Raffaele, 20132 Milan, Italy
| | - Claudia Lanterna
- Cancer Gene Therapy Unit, Program of Immunology and Bio Immuno Gene Therapy of Cancer, Division of Molecular Oncology, and Department of Pathology, Scientific Institute San Raffaele, 20132 Milan, Italy
| | | | - Aida Paniccia
- Cancer Gene Therapy Unit, Program of Immunology and Bio Immuno Gene Therapy of Cancer, Division of Molecular Oncology, and Department of Pathology, Scientific Institute San Raffaele, 20132 Milan, Italy
| | - Andrea Musumeci
- Cancer Gene Therapy Unit, Program of Immunology and Bio Immuno Gene Therapy of Cancer, Division of Molecular Oncology, and Department of Pathology, Scientific Institute San Raffaele, 20132 Milan, Italy
| | - Elena Chiricozzi
- Department of Medical Chemistry, Biochemistry and Biotechnology, Center of Excellence on Neurodegenerative Diseases, University of Milan, 20090 Segrate, Italy
| | | | - Simona Daniele
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | - Jan-Ake Gustafsson
- Department of Biosciences and Nutrition, Karolinska Institute, S-14183 Huddinge, Sweden
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX 77204
| | - Claudio Doglioni
- Cancer Gene Therapy Unit, Program of Immunology and Bio Immuno Gene Therapy of Cancer, Division of Molecular Oncology, and Department of Pathology, Scientific Institute San Raffaele, 20132 Milan, Italy
- Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | | | - Andrea Leiva
- Cancer Gene Therapy Unit, Program of Immunology and Bio Immuno Gene Therapy of Cancer, Division of Molecular Oncology, and Department of Pathology, Scientific Institute San Raffaele, 20132 Milan, Italy
| | - Maria Grazia Ciampa
- Department of Medical Chemistry, Biochemistry and Biotechnology, Center of Excellence on Neurodegenerative Diseases, University of Milan, 20090 Segrate, Italy
| | - Laura Mauri
- Department of Medical Chemistry, Biochemistry and Biotechnology, Center of Excellence on Neurodegenerative Diseases, University of Milan, 20090 Segrate, Italy
| | - Cristina Sensi
- Proteomics and Protein Structure Study Group, Department of Pharmacological Sciences, University of Milan, 20133 Milan, Italy
| | - Alessandro Prinetti
- Department of Medical Chemistry, Biochemistry and Biotechnology, Center of Excellence on Neurodegenerative Diseases, University of Milan, 20090 Segrate, Italy
| | - Ivano Eberini
- Proteomics and Protein Structure Study Group, Department of Pharmacological Sciences, University of Milan, 20133 Milan, Italy
| | - J. Rodrigo Mora
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA 02114
| | - Claudio Bordignon
- Università Vita-Salute San Raffaele, 20132 Milan, Italy
- MolMed S.p.A., 20132 Milan, Italy
| | - Knut R. Steffensen
- Department of Biosciences and Nutrition, Karolinska Institute, S-14183 Huddinge, Sweden
| | - Sandro Sonnino
- Department of Medical Chemistry, Biochemistry and Biotechnology, Center of Excellence on Neurodegenerative Diseases, University of Milan, 20090 Segrate, Italy
| | - Silvano Sozzani
- Humanitas Clinical and Research Center, 20089 Rozzano, Italy
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | | | - Vincenzo Russo
- Cancer Gene Therapy Unit, Program of Immunology and Bio Immuno Gene Therapy of Cancer, Division of Molecular Oncology, and Department of Pathology, Scientific Institute San Raffaele, 20132 Milan, Italy
| |
Collapse
|
40
|
Oldenburger A, Maarsingh H, Schmidt M. Multiple facets of cAMP signalling and physiological impact: cAMP compartmentalization in the lung. Pharmaceuticals (Basel) 2012; 5:1291-331. [PMID: 24281338 PMCID: PMC3816672 DOI: 10.3390/ph5121291] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 11/15/2012] [Accepted: 11/20/2012] [Indexed: 12/20/2022] Open
Abstract
Therapies involving elevation of the endogenous suppressor cyclic AMP (cAMP) are currently used in the treatment of several chronic inflammatory disorders, including chronic obstructive pulmonary disease (COPD). Characteristics of COPD are airway obstruction, airway inflammation and airway remodelling, processes encompassed by increased airway smooth muscle mass, epithelial changes, goblet cell and submucosal gland hyperplasia. In addition to inflammatory cells, airway smooth muscle cells and (myo)fibroblasts, epithelial cells underpin a variety of key responses in the airways such as inflammatory cytokine release, airway remodelling, mucus hypersecretion and airway barrier function. Cigarette smoke, being next to environmental pollution the main cause of COPD, is believed to cause epithelial hyperpermeability by disrupting the barrier function. Here we will focus on the most recent progress on compartmentalized signalling by cAMP. In addition to G protein-coupled receptors, adenylyl cyclases, cAMP-specific phospho-diesterases (PDEs) maintain compartmentalized cAMP signalling. Intriguingly, spatially discrete cAMP-sensing signalling complexes seem also to involve distinct members of the A-kinase anchoring (AKAP) superfamily and IQ motif containing GTPase activating protein (IQGAPs). In this review, we will highlight the interaction between cAMP and the epithelial barrier to retain proper lung function and to alleviate COPD symptoms and focus on the possible molecular mechanisms involved in this process. Future studies should include the development of cAMP-sensing multiprotein complex specific disruptors and/or stabilizers to orchestrate cellular functions. Compartmentalized cAMP signalling regulates important cellular processes in the lung and may serve as a therapeutic target.
Collapse
Affiliation(s)
- Anouk Oldenburger
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, 9713 AV, Groningen, The Netherlands.
| | | | | |
Collapse
|
41
|
Khounlotham M, Kim W, Peatman E, Nava P, Medina-Contreras O, Addis C, Koch S, Fournier B, Nusrat A, Denning TL, Parkos CA. Compromised intestinal epithelial barrier induces adaptive immune compensation that protects from colitis. Immunity 2012; 37:563-73. [PMID: 22981539 DOI: 10.1016/j.immuni.2012.06.017] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 04/13/2012] [Accepted: 06/07/2012] [Indexed: 11/24/2022]
Abstract
Mice lacking junctional adhesion molecule A (JAM-A, encoded by F11r) exhibit enhanced intestinal epithelial permeability, bacterial translocation, and elevated colonic lymphocyte numbers, yet do not develop colitis. To investigate the contribution of adaptive immune compensation in response to increased intestinal epithelial permeability, we examined the susceptibility of F11r(-/-)Rag1(-/-) mice to acute colitis. Although negligible contributions of adaptive immunity in F11r(+/+)Rag1(-/-) mice were observed, F11r(-/-)Rag1(-/-) mice exhibited increased microflora-dependent colitis. Elimination of T cell subsets and cytokine analyses revealed a protective role for TGF-β-producing CD4(+) T cells in F11r(-/-) mice. Additionally, loss of JAM-A resulted in elevated mucosal and serum IgA that was dependent upon CD4(+) T cells and TGF-β. Absence of IgA in F11r(+/+)Igha(-/-) mice did not affect disease, whereas F11r(-/-)Igha(-/-) mice displayed markedly increased susceptibility to acute injury-induced colitis. These data establish a role for adaptive immune-mediated protection from acute colitis under conditions of intestinal epithelial barrier compromise.
Collapse
Affiliation(s)
- Manirath Khounlotham
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Function of junctional adhesion molecules (JAMs) in leukocyte migration and homeostasis. Arch Immunol Ther Exp (Warsz) 2012; 61:15-23. [PMID: 22940878 DOI: 10.1007/s00005-012-0199-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 07/23/2012] [Indexed: 01/09/2023]
Abstract
Homeostasis is a word widely used in the scientific community to refer to the property of a system to maintain its uniformity and functionality. In living organisms, the word refers to the concept enunciated 150 years ago by C. Bernard by which external variations must be compensated for in order to maintain internal conditions compatible with life. This is especially true in the case of highly dynamic system such as the hematopoietic system that requires the coordinated control of cell proliferation and death within specialized microenvironments that are anatomically distinct. As a consequence, hematopoietic cell adhesion and migration must be tightly controlled in order for hematopoietic cells to reach and to be maintained in appropriate microenvironments. The junctional adhesion molecules (JAMs) are adhesion molecules that belong to the immunoglobulin superfamily (IgSf) and that have been initially identified as important players controlling vascular permeability and leukocyte transendothelial migration. This involves the regulated localization of the JAMs at lateral endothelial cell/cell borders and their interaction with leukocyte integrins. More recently, some of the JAM family members have also been found to be expressed by stromal cells and to regulate chemokine secretion within lymphoid organs, acting not only on leukocyte transendothelial migration, but also on hematopoietic cell retention within specialized microenvironments. This review summarizes recent progress in understanding the role of the JAMs in leukocyte adhesion and migration to tentatively draw an integrated view of the homeostatic function of the JAMs within the hematopoietic system.
Collapse
|
43
|
Lakshmi SP, Reddy AT, Naik MU, Naik UP, Reddy RC. Effects of JAM-A deficiency or blocking antibodies on neutrophil migration and lung injury in a murine model of ALI. Am J Physiol Lung Cell Mol Physiol 2012; 303:L758-66. [PMID: 22904169 DOI: 10.1152/ajplung.00107.2012] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Transmigration of neutrophils (PMNs) from the vasculature into inflamed tissues, mediated by interactions between PMNs and adhesion molecules on endothelial cells, is an essential aspect of inflammation. The crucial adhesion molecules include junctional adhesion molecule (JAM)-A. Investigation of the role of this molecule in models of inflammatory disease has been limited, however, and results in different disease models have varied. No previous study has addressed JAM-A in lung disease or effects on oxidant stress and proinflammatory cytokines. We use JAM-A knockout mice and blocking antibodies to investigate the role of JAM-A in a murine model of acute lung injury (ALI). With either experimental system, we find that absence of JAM-A activity significantly reduces migration of PMNs into the alveolar space, with a resulting decrease in oxidative stress. However, there is no reduction in whole lung activity of PMN-associated myeloperoxidase, presumably reflecting the histologically observed retention of PMNs in lung tissue. Activity of these retained PMNs may account for our failure to find significant change in markers of lung oxidative stress or cytokine and chemokine levels in plasma, lung, and bronchoalveolar lavage fluid. We likewise see no JAM-A-related changes in markers of capillary permeability or lung injury. A similar lack of congruence between effects on PMN migration and tissue injury has been reported in other disease models and for other adhesion molecules in models of ALI. Our results thus confirm the crucial role of JAM-A in PMN transmigration but demonstrate that transmigration is not essential for other aspects of inflammation or for lung injury in ALI.
Collapse
Affiliation(s)
- Sowmya P Lakshmi
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, Georgia, USA
| | | | | | | | | |
Collapse
|
44
|
Relocalization of junctional adhesion molecule A during inflammatory stimulation of brain endothelial cells. Mol Cell Biol 2012; 32:3414-27. [PMID: 22733993 DOI: 10.1128/mcb.06678-11] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Junctional adhesion molecule A (JAM-A) is a unique tight junction (TJ) transmembrane protein that under basal conditions maintains endothelial cell-cell interactions but under inflammatory conditions acts as a leukocyte adhesion molecule. This study investigates the fate of JAM-A during inflammatory TJ complex remodeling and paracellular route formation in brain endothelial cells. The chemokine (C-C motif) ligand 2 (CCL2) induced JAM-A redistribution from the interendothelial cell area to the apical surface, where JAM-A played a role as a leukocyte adhesion molecule participating in transendothelial cell migration of neutrophils and monocytes. JAM-A redistribution was associated with internalization via macropinocytosis during paracellular route opening. A tracer study with dextran-Texas Red indicated that internalization occurred within a short time period (~10 min) by dextran-positive vesicles and then became sorted to dextran-positive/Rab34-positive/Rab5-positive vesicles and then Rab4-positive endosomes. By ~20 min, most internalized JAM-A moved to the brain endothelial cell apical membrane. Treatment with a macropinocytosis inhibitor, 5-(N-ethyl-N-isopropyl)amiloride, or Rab5/Rab4 depletion with small interfering RNA oligonucleotides prevented JAM-A relocalization, suggesting that macropinocytosis and recycling to the membrane surface occur during JAM-A redistribution. Analysis of the signaling pathways indicated involvement of RhoA and Rho kinase in JAM-A relocalization. These data provide new insights into the molecular and cellular mechanisms involved in blood-brain barrier remodeling during inflammation.
Collapse
|
45
|
Abstract
BACKGROUND The process of extravasation of leucocytes from the vasculature into an infected, inflamed or injured tissue, designated the leucocyte adhesion cascade, is a major process in innate and adaptive immunity. In every immune process, both agonists and inhibitors, that is, positive and negative regulators, exist. MATERIALS AND METHODS It was only recently that endogenous inhibitors of the leucocyte adhesion cascade were identified, whereas many selectin, integrin and immunoglobulin superfamily adhesion receptors as well as chemokines and chemokine receptors promoting leucocyte recruitment have been described over the last three decades. Endogenous negative regulators include for instance pentraxin-3 (PTX-3) that blocks selectin-dependent leucocyte rolling, or the endothelium-derived developmental endothelial locus-1 (Del-1) that antagonizes beta2-integrin-mediated firm adhesion of leucocytes to the endothelium. CONCLUSIONS As leucocyte infiltration is a major therapeutic target in inflammatory and autoimmune disease, it becomes obvious that such endogenous anti-adhesive and anti-inflammatory agents may represent an attractive novel therapeutic platform for inflammatory and immune disorders. This review focuses on these novel endogenous inhibitors of leucocyte recruitment.
Collapse
Affiliation(s)
- Triantafyllos Chavakis
- Division of Vascular Inflammation, Diabetes and Kidney, Department of Medicine and Institute of Physiology, University Dresden, Dresden, Germany.
| |
Collapse
|
46
|
Iwaya H, Maeta K, Hara H, Ishizuka S. Mucosal permeability is an intrinsic factor in susceptibility to dextran sulfate sodium-induced colitis in rats. Exp Biol Med (Maywood) 2012; 237:451-60. [PMID: 22522346 DOI: 10.1258/ebm.2011.011269] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
We investigated differences in the pathogenesis of dextran sulfate sodium (DSS)-induced colitis between two inbred rat strains, Wistar King A Hokkaido (WKAH) and Dark Agouti (DA) rats, to determine the intrinsic factors responsible for the development of colitis. DSS exposure exacerbated the clinical symptoms such as body weight loss, stool consistency and rectal bleeding in DA rats rather than that in WKAH rats. Additionally, the average survival was shorter in DA rats than in WKAH rats. The expression levels of tumor necrosis factor-α, interleukin (IL)-12 p35 and IL-23 p19 increased prominently in the DA rats that were administered DSS, accompanied by severe infiltration of leukocytes into the colon. We also found that colonic permeability was greater in the DA rats than in the WKAH rats. In Ussing chambers, exposure of the isolated colon tissue to DSS enhanced the colonic permeability of both strains. Immunoblot analysis revealed that the expression levels of tight junction (TJ) proteins were modulated during DSS administration. Higher expression levels of claudin-4 and junctional adhesion molecule-A proteins were observed in DA rats than in WKAH rats, even in intact conditions. These results indicated that the expression pattern of TJ proteins determines the colonic permeability of the rats. In conclusion, the intrinsic colonic permeability is one of critical factors responsible for the susceptibility of rats to colitis.
Collapse
Affiliation(s)
- Hitoshi Iwaya
- Division of Applied Bioscience, Research Faculty of Agriculture, Hokkaido University, Sapporo 060-8589, Japan
| | | | | | | |
Collapse
|
47
|
Schmidt EP, Lee WL, Zemans RL, Yamashita C, Downey GP. On, around, and through: neutrophil-endothelial interactions in innate immunity. Physiology (Bethesda) 2012; 26:334-47. [PMID: 22013192 DOI: 10.1152/physiol.00011.2011] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
This manuscript will review our current understanding of neutrophilic polymorphonuclear leukocyte (neutrophil) interactions with the endothelium during immune and inflammatory responses, focusing on the molecular mechanisms regulating neutrophil adhesion to and migration through the endothelium in response to infection or tissue injury. This is a complex and dynamic area of research and one that has been the topic of several recent comprehensive reviews to which the interested reader is referred (64, 118, 131). By design, this review will begin with a brief review of some basic aspects of neutrophil biology and endothelial adhesion to provide a foundation. The remainder of the review will focus on selected areas of this complex field, specifically the role of the endothelial glycocalyx in regulating neutrophil adhesion and the mechanisms and consequences of migration of neutrophils between (paracellular) and through (transcellular) endothelial cells during egress from the vasculature.
Collapse
Affiliation(s)
- Eric P Schmidt
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | | | | | | | | |
Collapse
|
48
|
Williams DW, Eugenin EA, Calderon TM, Berman JW. Monocyte maturation, HIV susceptibility, and transmigration across the blood brain barrier are critical in HIV neuropathogenesis. J Leukoc Biol 2012; 91:401-15. [PMID: 22227964 DOI: 10.1189/jlb.0811394] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
HIV continues to be a global health crisis with more than 34 million people infected worldwide (UNAIDS: Report on the Global AIDS Epidemic 2010, Geneva, World Health Organization). HIV enters the CNS within 2 weeks of infection and establishes a spectrum of HAND in a large percentage of infected individuals. These neurologic deficits greatly impact the quality of life of those infected with HIV. The establishment of HAND is largely attributed to monocyte transmigration, particularly that of a mature CD14(+)CD16(+) monocyte population, which is more susceptible to HIV infection, across the BBB into the CNS parenchyma in response to chemotactic signals. To enter the CNS, junctional proteins on the monocytes must participate in homo- and heterotypic interactions with those present on BMVECs of the BBB as they transmigrate across the barrier. This transmigration is responsible for bringing virus into the brain and establishing chronic neuroinflammation. While there is baseline trafficking of monocytes into the CNS, the increased chemotactic signals present during HIV infection of the brain promote exuberant monocyte transmigration into the CNS. This review will discuss the mechanisms of monocyte differentiation/maturation, HIV infectivity, and transmigration into the CNS parenchyma that contribute to the establishment of cognitive impairment in HIV-infected individuals. It will focus on markers of monocyte subpopulations, how differentiation/maturation alters HIV infectivity, and the mechanisms that promote their increased transmigration across the BBB into the CNS.
Collapse
Affiliation(s)
- Dionna W Williams
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY, 10461, USA
| | | | | | | |
Collapse
|
49
|
Abstract
Junctional adhesion molecules are transmembrane proteins that belong to the immunoglobulin superfamily. In addition to their localization in close proximity to the tight junctions in endothelial and epithelial cells, junctional adhesion molecules are also expressed in circulating cells that do not form junctions, such as leukocytes and platelets. As a consequence, these proteins are associated not only with the permeability-regulating barrier function of the tight junctions, but also with other biologic processes, such as inflammatory reactions, responses to vascular injury, and tumor angiogenesis. Furthermore, because of their transmembrane topology, junctional adhesion molecules are poised both for receiving inputs from the cell interior (their expression, localization, and function being regulated in response to inflammatory cytokines and growth factors) and for translating extracellular adhesive events into functional responses. This review focuses on the different roles of junctional adhesion molecules in normal and pathologic conditions, with emphasis on inflammatory reactions and vascular responses to injury.
Collapse
Affiliation(s)
- Gianfranco Bazzoni
- Department of Biochemistry and Molecular Pharmacology Mario Negri Institute of Pharmacological Research, Milano, Italy.
| |
Collapse
|
50
|
Abrogation of junctional adhesion molecule-A expression induces cell apoptosis and reduces breast cancer progression. PLoS One 2011; 6:e21242. [PMID: 21695058 PMCID: PMC3117883 DOI: 10.1371/journal.pone.0021242] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Accepted: 05/24/2011] [Indexed: 01/13/2023] Open
Abstract
Intercellular junctions promote homotypic cell to cell adhesion and transfer intracellular signals which control cell growth and apoptosis. Junctional adhesion molecule-A (JAM-A) is a transmembrane immunoglobulin located at tight junctions of normal epithelial cells of mammary ducts and glands. In the present paper we show that JAM-A acts as a survival factor for mammary carcinoma cells. JAM-A null mice expressing Polyoma Middle T under MMTV promoter develop significantly smaller mammary tumors than JAM-A positive mice. Angiogenesis and inflammatory or immune infiltrate were not statistically modified in absence of JAM-A but tumor cell apoptosis was significantly increased. Tumor cells isolated from JAM-A null mice or 4T1 cells incubated with JAM-A blocking antibodies showed reduced growth and increased apoptosis which paralleled altered junctional architecture and adhesive function. In a breast cancer clinical data set, tissue microarray data show that JAM-A expression correlates with poor prognosis. Gene expression analysis of mouse tumor samples showed a correlation between genes enriched in human G3 tumors and genes over expressed in JAM-A +/+ mammary tumors. Conversely, genes enriched in G1 human tumors correlate with genes overexpressed in JAM-A−/− tumors. We conclude that down regulation of JAM-A reduces tumor aggressive behavior by increasing cell susceptibility to apoptosis. JAM-A may be considered a negative prognostic factor and a potential therapeutic target.
Collapse
|