1
|
Andretto V, Rosso A, Zilio S, Sidi-Boumedine J, Boschetti G, Sankar S, Buffier M, Miele AE, Denis M, Choffour PA, Briançon S, Nancey S, Kryza D, Lollo G. Peptide-Based Hydrogel for Nanosystems Encapsulation: the Next Generation of Localized Delivery Systems for the Treatment of Intestinal Inflammations. Adv Healthc Mater 2024; 13:e2303280. [PMID: 38445812 DOI: 10.1002/adhm.202303280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/27/2023] [Revised: 02/10/2024] [Indexed: 03/07/2024]
Abstract
Conventional therapies for inflammatory bowel diseases are mainly based on systemic treatments which cause side effects and toxicity over long-term administration. Nanoparticles appear as a valid alternative to allow a preferential accumulation in inflamed tissues following oral administration while reducing systemic drug exposure. To increase their residence time in the inflamed intestine, the nanoparticles are here associated with a hydrogel matrix. A bioadhesive peptide-based hydrogel is mixed with nanoemulsions, creating a hybrid lipid-polymer nanocomposite. Mucopenetrating nanoemulsions of 100 nm are embedded in a scaffold constituted of the self-assembling peptide hydrogel product PuraStat. The nanocomposite is fully characterized to study the impact of lipid particles in the hydrogel structure. Rheological measurements and circular dichroism analyses are performed to investigate the system's microstructure and physical properties. Biodistribution studies demonstrate that the nanocomposite acts as a depot in the stomach and facilitates the slow release of the nanoemulsions in the intestine. Efficacy studies upon oral administration of the drug-loaded system show the improvement of the disease score in a mouse model of intestinal inflammation.
Collapse
Affiliation(s)
- Valentina Andretto
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, Villeurbanne, F-69622, France
| | - Annalisa Rosso
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, Villeurbanne, F-69622, France
| | - Serena Zilio
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, Villeurbanne, F-69622, France
- SATT, Ouest Valorisation, 14C Rue du Patis Tatelin, Renne, 35708, France
| | - Jacqueline Sidi-Boumedine
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, Villeurbanne, F-69622, France
| | - Gilles Boschetti
- Department of Gastroenterology, Lyon Sud Hospital, Hospices Civil de Lyon and CIRI, Lyon, 69495, France
| | - Sharanya Sankar
- 3-D Matrix Europe SAS, Medical Technology, Caluire-et-Cuire, 69300, France
| | - Marie Buffier
- 3-D Matrix Europe SAS, Medical Technology, Caluire-et-Cuire, 69300, France
| | - Adriana Erica Miele
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, ISA UMR 5280, 5 rue de la Doua, Villeurbanne, F-69100, France
- Dept Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, Rome, I-00185, Italy
| | - Morgane Denis
- Univ Lyon, Université Claude Bernard Lyon, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69008, France
- Antineo, R&D Department, Lyon, 69008, France
| | | | - Stéphanie Briançon
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, Villeurbanne, F-69622, France
| | - Stéphane Nancey
- Department of Gastroenterology, Lyon Sud Hospital, Hospices Civil de Lyon and CIRI, Lyon, 69495, France
| | - David Kryza
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, Villeurbanne, F-69622, France
- Hospices Civils de Lyon, Lyon, 69437, France
| | - Giovanna Lollo
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, Villeurbanne, F-69622, France
| |
Collapse
|
2
|
Sumagin R. Phenotypic and Functional Diversity of Neutrophils in Gut Inflammation and Cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:2-12. [PMID: 37918801 PMCID: PMC10768535 DOI: 10.1016/j.ajpath.2023.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 08/21/2023] [Revised: 10/09/2023] [Accepted: 10/18/2023] [Indexed: 11/04/2023]
Abstract
Neutrophils [polymorphonuclear leukocytes (PMNs)] execute important effector functions protecting the host against invading pathogens. However, their activity in tissue can exacerbate inflammation and inflammation-associated tissue injury and tumorigenesis. Until recently, PMNs were considered to be short-lived, terminally differentiated phagocytes. However, this view is rapidly changing with the emerging evidence of increased PMN lifespan in tissues, PMN plasticity, and phenotypic heterogeneity. Specialized PMN subsets have been identified in inflammation and in developing tumors, consistent with both beneficial and detrimental functions of PMNs in these conditions. Because PMN and tumor-associated neutrophil activity and the resulting beneficial/detrimental impacts primarily occur after homing to inflamed tissue/tumors, studying the underlying mechanisms of PMN/tumor-associated neutrophil trafficking is of high interest and clinical relevance. This review summarizes some of the key findings from over a decade of work from my laboratory and others on the regulation of PMN recruitment and identification of phenotypically and functionally diverse PMN subtypes as they pertain to gut inflammation and colon cancer.
Collapse
Affiliation(s)
- Ronen Sumagin
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
3
|
Mraz V, Lohmann RKD, Menzel M, Hawkes A, Vaher H, Funch AB, Jee MH, Gadsbøll ASØ, Weber JF, Yeung K, Ødum N, Woetmann A, McKay D, Witherden D, Geisler C, Bonefeld CM. The junctional adhesion molecule-like protein (JAML) is important for the inflammatory response during contact hypersensitivity. Contact Dermatitis 2023; 89:323-334. [PMID: 37619972 PMCID: PMC11034946 DOI: 10.1111/cod.14409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/14/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND The junctional adhesion molecule-like protein (JAML) plays important roles in wound healing and activation of epidermal γδ T cells in mice. Whether JAML plays a role in contact hypersensitivity (CHS), the animal model of allergic contact dermatitis (ACD), is not known. METHODS To examine the role of JAML in CHS, we used various mouse models of CHS in JAML knockout (KO) and wild-type (WT) mice. Furthermore, the expression of the JAML ligand coxsackievirus and adenovirus receptor (CXADR) on keratinocytes was accessed in vitro and in vivo. RESULTS JAML KO mice had a diminished inflammatory response during both the sensitization and elicitation phase of CHS and had reduced numbers of CD8+ and CD4+ T cells in the epidermis. Furthermore, interferon γ (IFNγ), interleukin 1β (IL-1β) and CXCL10 production were significantly reduced in JAML KO mice during the elicitation phase. We found that CD8+ T cells express JAML and that JAML is essential for rapid flare-up responses to contact allergens. Finally, we show that keratinocytes up-regulate the JAML ligand CXADR following exposure to contact allergens. CONCLUSION Our study is the first to show a central role of JAML in CHS and reveals a potential new target for the treatment of ACD in humans.
Collapse
Affiliation(s)
- Veronika Mraz
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
| | - Rebecca K. D. Lohmann
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
| | - Mandy Menzel
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
- Respiratory Research Unit, Department of Respiratory Medicine, Bispebjerg Hospital, Copenhagen, Denmark
| | - Alana Hawkes
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Helen Vaher
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
| | - Anders B. Funch
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
- Department of Dermatology and Allergy, National Allergy Research Center, Copenhagen University Hospital Gentofte, Hellerup, Denmark
| | - Mia H. Jee
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
| | - Anne-Sofie Ø. Gadsbøll
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
| | - Julie F. Weber
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
| | - Kelvin Yeung
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
- Department of Dermatology and Allergy, National Allergy Research Center, Copenhagen University Hospital Gentofte, Hellerup, Denmark
| | - Niels Ødum
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
| | - Anders Woetmann
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
| | - Dianne McKay
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Deborah Witherden
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Carsten Geisler
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
| | - Charlotte M. Bonefeld
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Mack KL, Talbott HE, Griffin MF, Parker JBL, Guardino NJ, Spielman AF, Davitt MF, Mascharak S, Downer M, Morgan A, Valencia C, Akras D, Berger MJ, Wan DC, Fraser HB, Longaker MT. Allele-specific expression reveals genetic drivers of tissue regeneration in mice. Cell Stem Cell 2023; 30:1368-1381.e6. [PMID: 37714154 PMCID: PMC10592051 DOI: 10.1016/j.stem.2023.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/05/2022] [Revised: 06/16/2023] [Accepted: 08/22/2023] [Indexed: 09/17/2023]
Abstract
In adult mammals, skin wounds typically heal by scarring rather than through regeneration. In contrast, "super-healer" Murphy Roths Large (MRL) mice have the unusual ability to regenerate ear punch wounds; however, the molecular basis for this regeneration remains elusive. Here, in hybrid crosses between MRL and non-regenerating mice, we used allele-specific gene expression to identify cis-regulatory variation associated with ear regeneration. Analyzing three major cell populations (immune, fibroblast, and endothelial), we found that genes with cis-regulatory differences specifically in fibroblasts were associated with wound-healing pathways and also co-localized with quantitative trait loci for ear wound-healing. Ectopic treatment with one of these proteins, complement factor H (CFH), accelerated wound repair and induced regeneration in typically fibrotic wounds. Through single-cell RNA sequencing (RNA-seq), we observed that CFH treatment dramatically reduced immune cell recruitment to wounds, suggesting a potential mechanism for CFH's effect. Overall, our results provide insights into the molecular drivers of regeneration with potential clinical implications.
Collapse
Affiliation(s)
- Katya L Mack
- Stanford University, Department of Biology, Stanford, CA, USA
| | - Heather E Talbott
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA; Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA
| | - Michelle F Griffin
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Jennifer B L Parker
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA; Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA
| | - Nicholas J Guardino
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Amanda F Spielman
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Michael F Davitt
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Shamik Mascharak
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA; Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA
| | - Mauricio Downer
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Annah Morgan
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Caleb Valencia
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Deena Akras
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Mark J Berger
- Stanford University, Department of Computer Science, Stanford, CA 94305, USA
| | - Derrick C Wan
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Hunter B Fraser
- Stanford University, Department of Biology, Stanford, CA, USA.
| | - Michael T Longaker
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA; Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA.
| |
Collapse
|
5
|
Eschweiler S, Wang A, Ramírez-Suástegui C, von Witzleben A, Li Y, Chee SJ, Simon H, Mondal M, Ellis M, Thomas GJ, Chandra V, Ottensmeier CH, Vijayanand P. JAML immunotherapy targets recently activated tumor-infiltrating CD8 + T cells. Cell Rep 2023; 42:112040. [PMID: 36701231 PMCID: PMC10366340 DOI: 10.1016/j.celrep.2023.112040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/04/2022] [Revised: 11/27/2022] [Accepted: 01/12/2023] [Indexed: 01/26/2023] Open
Abstract
Junctional adhesion molecule-like protein (JAML) serves as a co-stimulatory molecule in γδ T cells. While it has recently been described as a cancer immunotherapy target in mice, its potential to cause toxicity, specific mode of action with regard to its cellular targets, and whether it can be targeted in humans remain unknown. Here, we show that JAML is induced by T cell receptor engagement, reveal that this induction is linked to cis-regulatory interactions between the CD3D and JAML gene loci. When compared with other immunotherapy targets plagued by low target specificity and end-organ toxicity, we find JAML to be mostly restricted to and highly expressed by tissue-resident memory CD8+ T cells in multiple cancer types. By delineating the key cellular targets and functional consequences of agonistic anti-JAML therapy in a murine melanoma model, we show its specific mode of action and the reason for its synergistic effects with anti-PD-1.
Collapse
Affiliation(s)
| | - Alice Wang
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Adrian von Witzleben
- Department of Otorhinolaryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Yingcong Li
- La Jolla Institute for Immunology, La Jolla, CA, USA; University of California San Diego, La Jolla, CA, USA
| | - Serena J Chee
- Department of Molecular and Clinical Cancer Medicine and NIHR and CRUK Liverpool Experimental Cancer Medicine Center, University of Liverpool, Liverpool, UK; Department of Respiratory Medicine, Liverpool Heart and Chest Hospital and NHS Foundation Trust, Liverpool, UK
| | - Hayley Simon
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Matthew Ellis
- NIHR and CRUK Southampton Experimental Cancer Medicine Center, Faculty of Medicine, University of Southampton, Southampton, UK; NIHR Southampton Biomedical Research Center, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Gareth J Thomas
- NIHR and CRUK Southampton Experimental Cancer Medicine Center, Faculty of Medicine, University of Southampton, Southampton, UK; NIHR Southampton Biomedical Research Center, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Vivek Chandra
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Christian H Ottensmeier
- La Jolla Institute for Immunology, La Jolla, CA, USA; Department of Molecular and Clinical Cancer Medicine and NIHR and CRUK Liverpool Experimental Cancer Medicine Center, University of Liverpool, Liverpool, UK
| | - Pandurangan Vijayanand
- La Jolla Institute for Immunology, La Jolla, CA, USA; University of California San Diego, La Jolla, CA, USA; Department of Molecular and Clinical Cancer Medicine and NIHR and CRUK Liverpool Experimental Cancer Medicine Center, University of Liverpool, Liverpool, UK.
| |
Collapse
|
6
|
Ortiz-Zapater E, Bagley DC, Hernandez VL, Roberts LB, Maguire TJA, Voss F, Mertins P, Kirchner M, Peset-Martin I, Woszczek G, Rosenblatt J, Gotthardt M, Santis G, Parsons M. Epithelial coxsackievirus adenovirus receptor promotes house dust mite-induced lung inflammation. Nat Commun 2022; 13:6407. [PMID: 36302767 PMCID: PMC9613683 DOI: 10.1038/s41467-022-33882-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/03/2022] [Accepted: 10/06/2022] [Indexed: 12/25/2022] Open
Abstract
Airway inflammation and remodelling are important pathophysiologic features in asthma and other respiratory conditions. An intact epithelial cell layer is crucial to maintain lung homoeostasis, and this depends on intercellular adhesion, whilst damaged respiratory epithelium is the primary instigator of airway inflammation. The Coxsackievirus Adenovirus Receptor (CAR) is highly expressed in the epithelium where it modulates cell-cell adhesion stability and facilitates immune cell transepithelial migration. However, the contribution of CAR to lung inflammation remains unclear. Here we investigate the mechanistic contribution of CAR in mediating responses to the common aeroallergen, House Dust Mite (HDM). We demonstrate that administration of HDM in mice lacking CAR in the respiratory epithelium leads to loss of peri-bronchial inflammatory cell infiltration, fewer goblet-cells and decreased pro-inflammatory cytokine release. In vitro analysis in human lung epithelial cells confirms that loss of CAR leads to reduced HDM-dependent inflammatory cytokine release and neutrophil migration. Epithelial CAR depletion also promoted smooth muscle cell proliferation mediated by GSK3β and TGF-β, basal matrix production and airway hyperresponsiveness. Our data demonstrate that CAR coordinates lung inflammation through a dual function in leucocyte recruitment and tissue remodelling and may represent an important target for future therapeutic development in inflammatory lung diseases.
Collapse
Affiliation(s)
- Elena Ortiz-Zapater
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Science King's College London, London, UK
| | - Dustin C Bagley
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
| | | | - Luke B Roberts
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Thomas J A Maguire
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Felizia Voss
- Max-Delbrück-Centrum für Molekulare Medizin in the Helmholtz Assoziation (MDC), Berlin, Germany
- DZHK Partner site Berlin, Berlin, Germany
| | - Philipp Mertins
- Berlin Institute of Health at Charité, Universitaetsmedizin Berlin, Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Marieluise Kirchner
- Berlin Institute of Health at Charité, Universitaetsmedizin Berlin, Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | | | - Grzegorz Woszczek
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Jody Rosenblatt
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
| | - Michael Gotthardt
- Max-Delbrück-Centrum für Molekulare Medizin in the Helmholtz Assoziation (MDC), Berlin, Germany
- Berlin Institute of Health at Charité, Universitaetsmedizin Berlin, Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Charité Universitätsmedizin Berlin, Berlin, Germany
| | - George Santis
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Science King's College London, London, UK
- Department of Respiratory Medicine, Guy's & St Thomas NHS Trust, London, UK
| | - Maddy Parsons
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK.
| |
Collapse
|
7
|
Neutrophil-Epithelial Crosstalk During Intestinal Inflammation. Cell Mol Gastroenterol Hepatol 2022; 14:1257-1267. [PMID: 36089244 PMCID: PMC9583449 DOI: 10.1016/j.jcmgh.2022.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/10/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 01/31/2023]
Abstract
Neutrophils are the most abundant leukocyte population in the human circulatory system and are rapidly recruited to sites of inflammation. Neutrophils play a multifaceted role in intestinal inflammation, as they contribute to the elimination of invading pathogens. Recently, their role in epithelial restitution has been widely recognized; however, they are also associated with bystander tissue damage. The intestinal epithelium provides a physical barrier to prevent direct contact of luminal contents with subepithelial tissues, which is extremely important for the maintenance of intestinal homeostasis. Numerous studies have demonstrated that transepithelial migration of neutrophils is closely related to disease symptoms and disruption of crypt architecture in inflammatory bowel disease and experimental colitis. There has been growing interest in how neutrophils interact with the epithelium under inflammatory conditions. Most studies focus on the effects of neutrophils on intestinal epithelial cells; however, the effects of intestinal epithelial cells on neutrophils during intestinal inflammation need to be well-established. Based on these data, we have summarized recent articles on the role of neutrophil-epithelial interactions in intestinal inflammation, particularly highlighting the epithelium-derived molecular regulators that mediate neutrophil recruitment, transepithelial migration, and detachment from the epithelium, as well as the functional consequences of their crosstalk. A better understanding of these molecular events may help develop novel therapeutic targets for mitigating the deleterious effects of neutrophils in inflammatory bowel disease.
Collapse
|
8
|
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: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution 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
|
9
|
Wu Q, Li R, Wang QX, Zhang MY, Liu TT, Qu YQ. Junctional adhesion molecule-like protein promotes tumor progression via the Wnt/β-catenin signaling pathway in lung adenocarcinoma. J Transl Med 2022; 20:260. [PMID: 35672776 PMCID: PMC9171988 DOI: 10.1186/s12967-022-03457-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/27/2022] [Accepted: 05/24/2022] [Indexed: 11/18/2022] Open
Abstract
Background Lung adenocarcinoma (LUAD) is a heavy social burden worldwide. Because the mechanisms involved in LUAD remain unclear, the prognosis of LUAD remains poor. Consequently, it is urgent to investigate the potential mechanisms of LUAD. Junctional adhesion molecule-like protein (JAML), is recognized as a tumorigenesis molecule in gastric cancer. However, the role of JAML in LUAD is still unclear. Here we aimed to evaluate the role of JAML in LUAD. Methods qRT-PCR, Western blotting and immunohistochemistry were conducted to investigate the expression of JAML in LUAD tissues. JAML was knocked down and overexpressed in LUAD cells using transient transfection by siRNA and plasmids or stable transfection by lentivirus. Proliferation potential of LUAD cells were detected by Cell Counting Kit-8, EdU incorporation and Colony formation assay. Migration and invasion abilities of LUAD cells were determined by wound healing, transwell migration and invasion assays. Cell cycle and cell apoptosis were detected by flow cytometry. The effects of JAML in vivo were studied in xenograft tumor models. Western blotting was used to explore the molecular mechanisms of JAML function. In addition, rescue experiments were performed to verify the possible mechanisms. Results JAML expression was elevated in LUAD tissues compared with peritumor tissues, and this upregulation was positively related to pT and pTNM. Furthermore, both in vitro and in vivo, JAML silencing markedly repressed malignant behaviors of LUAD cells and vice versa. Knockdown of JAML also mediated cell cycle arrest at G0/G1 phase and promoted apoptosis in LUAD cells. Mechanistically, silencing JAML repressed the process of epithelial-mesenchymal transition by inactivating the Wnt/β-catenin pathway in LUAD cells. Effects of JAML can be rescued by Wnt/β-catenin pathway activator in A549 cells. Conclusions Our data reveal the oncogenic role of JAML in LUAD, indicating that JAML may be a predictive biomarker and novel therapeutic target for LUAD. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03457-w.
Collapse
|
10
|
Wang J, Liu H. The Roles of Junctional Adhesion Molecules (JAMs) in Cell Migration. Front Cell Dev Biol 2022; 10:843671. [PMID: 35356274 PMCID: PMC8959349 DOI: 10.3389/fcell.2022.843671] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/26/2021] [Accepted: 02/10/2022] [Indexed: 01/15/2023] Open
Abstract
The review briefly summarizes the role of the family of adhesion molecules, JAMs (junctional adhesion molecules), in various cell migration, covering germ cells, epithelial cells, endothelial cells, several leukocytes, and different cancer cells. These functions affect multiple diseases, including reproductive diseases, inflammation-related diseases, cardiovascular diseases, and cancers. JAMs bind to both similar and dissimilar proteins and take both similar and dissimilar effects on different cells. Concluding relevant results provides a reference to further research.
Collapse
Affiliation(s)
- Junqi Wang
- Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Han Liu
- Department of Pharmacy, People’s Hospital of Longhua, Shenzhen, China
- *Correspondence: Han Liu,
| |
Collapse
|
11
|
Parkos CA. LEUKOCYTE-EPITHELIAL INTERACTIONS: A DOUBLE-EDGED SWORD THAT PROTECTS AND INJURES DURING HEALTH AND DISEASE. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2022; 132:22-33. [PMID: 36196189 PMCID: PMC9480551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Academic Contribution Register] [Indexed: 06/16/2023]
Abstract
Neutrophils (PMNs) play a critical role in innate immunity, yet many pathologic conditions are associated with dysregulated infiltration of PMNs into tissues. In the gut, robust PMN accumulation and migration across the intestinal epithelium closely correlates with clinical symptoms in conditions such as ulcerative colitis. While much is known about how PMNs migrate out of blood vessels, far less is understood about how PMNs traverse epithelial barriers. Until fairly recently, in vitro models of PMN transepithelial migration (TEpM) across cultured intestinal epithelial cell lines provided many of the insights into the molecular basis of TEpM. However, innovative animal models have provided new avenues for investigating in vivo mechanisms regulating PMN TEpM. This report will highlight molecular insights gained from studies on PMN TEpM and provide a rationale for developing tissue targeted strategies directed at reducing pathologic consequences of dysregulated PMN trafficking in the gut.
Collapse
|
12
|
Sumioka T, Iwanishi H, Okada Y, Miyajima M, Ichikawa K, Reinach PS, Matsumoto KI, Saika S. Impairment of corneal epithelial wound healing is association with increased neutrophil infiltration and reactive oxygen species activation in tenascin X-deficient mice. J Transl Med 2021; 101:690-700. [PMID: 33782532 PMCID: PMC8137452 DOI: 10.1038/s41374-021-00576-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/01/2020] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 01/14/2023] Open
Abstract
The purpose of the study was to uncover the role of tenascin X in modulation of healing in mouse corneas subjected to epithelium debridement. Healing in corneas with an epithelial defect was evaluated at the levels of gene and protein expression. Wound healing-related mediators and inflammatory cell infiltration were detected by histology, immunohistochemistry and real-time RT-PCR. Tenascin X protein was upregulated in the wounded wild-type (WT) corneal epithelium. The lack of tenascin X impaired closure of an epithelial defect and accelerated infiltration of neutrophils into the wound periphery as compared to the response in WT tissue. Expression of wound healing-related proinflammatory and reparative components, i.e., interleukin-6, transforming growth factor β, matrix metalloproteinases, were unaffected by the loss of tenascin X expression. Marked accumulation of malondialdehyde (a lipid peroxidation-derived product) was observed in KO healing epithelia as compared with its WT counterpart. Neutropenia induced by systemic administration of a specific antibody rescued the impairment of epithelial healing in KO corneas, with reduction of malondialdehyde levels in the epithelial cells. Finally, we showed that a chemical scavenging reactive oxygen species reversed the impairment of attenuation of epithelial repair with a reduction of tissue levels of malondialdehyde. In conclusion, loss of tenascin X prolonged corneal epithelial wound healing and increased neutrophilic inflammatory response to debridement in mice. Tenascin X contributes to the control of neutrophil infiltration needed to support the regenerative response to injury and prevent the oxidative stress mediators from rising to cytotoxic levels.
Collapse
Affiliation(s)
- Takayoshi Sumioka
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan.
| | - Hiroki Iwanishi
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Yuka Okada
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Masayasu Miyajima
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Kana Ichikawa
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Peter S Reinach
- Department of Ophthalmology, Wenzhou Medical University, Wenzhou, P. R. China
| | - Ken-Ichi Matsumoto
- Department of Biosignaling and Radioisotope Experiment, Interdisciplinary Center for Science Research, Organization for Research and Academic Information, Shimane University, Izumo, Enya-cho, Japan
| | - Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| |
Collapse
|
13
|
Fang Y, Yang J, Zu G, Cong C, Liu S, Xue F, Ma S, Liu J, Sun Y, Sun M. Junctional Adhesion Molecule-Like Protein Promotes Tumor Progression and Metastasis via p38 Signaling Pathway in Gastric Cancer. Front Oncol 2021; 11:565676. [PMID: 33777731 PMCID: PMC7991718 DOI: 10.3389/fonc.2021.565676] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/25/2020] [Accepted: 02/01/2021] [Indexed: 12/29/2022] Open
Abstract
Junctional adhesion molecule-like protein (JAML), a newly discovered junctional adhesion molecule (JAM), mediates the adhesion and migration processes of various immune cells and endothelial/epithelial cells, ultimately regulating inflammation reaction. However, its role in tumors remains to be determined. The expression of JAML was examined in gastric cancer (GC) and peritumoral tissues from 63 patients. The relationship between JAML expression and clinical characteristics was also observed. In vitro, GC cell migration and proliferation were assessed by wound healing assay, transwell migration assay and EdU incorporation assay. Immunohistochemical staining results showed that JAML expression level was higher in GC tissues than in peritumoral tissues. High expression of JAML in cancer tissues was associated with worse cell differentiation, local lymph node involvement, deep infiltration, and advanced stage. In vitro, we found that JAML silencing inhibited GC cell migration and proliferation, while JAML overexpression promoted GC cell migration and proliferation, partially via p38 signaling. Taken together, our study revealed a critical role for JAML to promote GC cell migration and proliferation. JAML might be a novel diagnostic biomarker and therapeutic target for GC.
Collapse
Affiliation(s)
- Yuying Fang
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jianmin Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guohong Zu
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Changsheng Cong
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shuai Liu
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Fei Xue
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shuzhen Ma
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jie Liu
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yuping Sun
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Meili Sun
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China.,Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
14
|
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: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution 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
|
15
|
Alshetaiwi H, Pervolarakis N, McIntyre LL, Ma D, Nguyen Q, Rath JA, Nee K, Hernandez G, Evans K, Torosian L, Silva A, Walsh C, Kessenbrock K. Defining the emergence of myeloid-derived suppressor cells in breast cancer using single-cell transcriptomics. Sci Immunol 2020; 5:5/44/eaay6017. [PMID: 32086381 PMCID: PMC7219211 DOI: 10.1126/sciimmunol.aay6017] [Citation(s) in RCA: 305] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/02/2019] [Accepted: 01/23/2020] [Indexed: 12/26/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) are innate immune cells that acquire the capacity to suppress adaptive immune responses during cancer. It remains elusive how MDSCs differ from their normal myeloid counterparts, which limits our ability to specifically detect and therapeutically target MDSCs during cancer. Here, we sought to determine the molecular features of breast cancer-associated MDSCs using the widely studied mouse model based on the mouse mammary tumor virus (MMTV) promoter-driven expression of the polyomavirus middle T oncoprotein (MMTV-PyMT). To identify MDSCs in an unbiased manner, we used single-cell RNA sequencing to compare MDSC-containing splenic myeloid cells from breast tumor-bearing mice with wild-type controls. Our computational analysis of 14,646 single-cell transcriptomes revealed that MDSCs emerge through an aberrant neutrophil maturation trajectory in the spleen that confers them an immunosuppressive cell state. We establish the MDSC-specific gene signature and identify CD84 as a surface marker for improved detection and enrichment of MDSCs in breast cancers.
Collapse
Affiliation(s)
- Hamad Alshetaiwi
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA.,Department of Pathology, University of Hail, Hail 2440, Saudi Arabia
| | - Nicholas Pervolarakis
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Laura Lynn McIntyre
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Dennis Ma
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Quy Nguyen
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Jan Akara Rath
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges 1066, Switzerland
| | - Kevin Nee
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Grace Hernandez
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Katrina Evans
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Leona Torosian
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Anushka Silva
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Craig Walsh
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Kai Kessenbrock
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
16
|
Fu Y, Sun Y, Wang M, Hou Y, Huang W, Zhou D, Wang Z, Yang S, Tang W, Zhen J, Li Y, Wang X, Liu M, Zhang Y, Wang B, Liu G, Yu X, Sun J, Zhang C, Yi F. Elevation of JAML Promotes Diabetic Kidney Disease by Modulating Podocyte Lipid Metabolism. Cell Metab 2020; 32:1052-1062.e8. [PMID: 33186558 DOI: 10.1016/j.cmet.2020.10.019] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 12/22/2019] [Revised: 08/18/2020] [Accepted: 10/19/2020] [Indexed: 02/08/2023]
Abstract
Lipid accumulation in podocytes is a major determinant of diabetic kidney disease (DKD) and identification of potential therapeutic targets by mediating podocyte lipid metabolism has clinical importance. This study was to elucidate the role of JAML (junctional adhesion molecule-like protein) in the pathogenesis of DKD. We first confirmed the expression of JAML in podocytes and found that podocyte-specific deletion of Jaml ameliorated podocyte injury and proteinuria in two different models of diabetic mice. We further demonstrated a novel role of JAML in regulating podocyte lipid metabolism through SIRT1-mediated SREBP1 signaling. Similar results were also found in mice with adriamycin-induced nephropathy. Importantly, we observed a higher expression of JAML in glomeruli from subjects with DKD and other types of proteinuric kidney diseases, and the level of JAML was correlated with lipid accumulation and glomerular filtration rate, suggesting that JAML may be an attractive therapeutic target for proteinuric kidney disease.
Collapse
Affiliation(s)
- Yi Fu
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Yu Sun
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Mei Wang
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Yunfeng Hou
- Intensive Care Unit, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan 250014, China
| | - Wei Huang
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Di Zhou
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Ziying Wang
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Shuting Yang
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Wei Tang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Junhui Zhen
- Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Yujia Li
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Xiaojie Wang
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Min Liu
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Yan Zhang
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Baobao Wang
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan 250014, China
| | - Guangyi Liu
- Department of Nephrology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Xiao Yu
- Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan 250012 China
| | - Jinpeng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Fan Yi
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China.
| |
Collapse
|
17
|
Excoffon KJDA. The coxsackievirus and adenovirus receptor: virological and biological beauty. FEBS Lett 2020; 594:1828-1837. [PMID: 32298477 DOI: 10.1002/1873-3468.13794] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/03/2019] [Revised: 03/31/2020] [Accepted: 04/04/2020] [Indexed: 12/17/2022]
Abstract
The coxsackievirus and adenovirus receptor (CAR) is an essential multifunctional cellular protein that is only beginning to be understood. CAR serves as a receptor for many adenoviruses, human group B coxsackieviruses, swine vesicular disease virus, and possibly other viruses. While named for its function as a viral receptor, CAR is also involved in cell adhesion, immune cell activation, synaptic transmission, and signaling. Knockout mouse models were first to identify some of these biological functions; however, tissue-specific model systems have shed light on the complexity of different CAR isoforms and their specific activities. Many of these functions are mediated by the large number of interacting proteins described so far, and several new putative interactions have recently been discovered. As antiviral and gene therapy strategies that target CAR continue to emerge, future work poised to understand the biological implications of manipulating CAR in vivo is critical.
Collapse
Affiliation(s)
- Katherine J D A Excoffon
- Biological Sciences, Wright State University, Dayton, OH, USA.,Spirovant Sciences, Inc, Philadelphia, PA, USA
| |
Collapse
|
18
|
Wiesolek HL, Bui TM, Lee JJ, Dalal P, Finkielsztein A, Batra A, Thorp EB, Sumagin R. Intercellular Adhesion Molecule 1 Functions as an Efferocytosis Receptor in Inflammatory Macrophages. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:874-885. [PMID: 32035057 PMCID: PMC7180595 DOI: 10.1016/j.ajpath.2019.12.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 06/27/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 12/26/2022]
Abstract
Intercellular adhesion molecule-1 (ICAM-1) is up-regulated during inflammation by several cell types. ICAM-1 is best known for its role in mediating leukocyte adhesion to endothelial cells and guiding leukocytes across the vascular wall. Recently, macrophages have been shown to express ICAM-1, however, their role in macrophage function is unclear. We found that ICAM-1 expression was induced during inflammatory macrophage polarization and high numbers of ICAM-1-expressing macrophages were noted in inflamed colon tissue in a murine colitis model and in human inflammatory bowel disease. Because tissue macrophages play a critical role in removing apoptotic/necrotic cells in inflammation and injury, a process termed efferocytosis, it was examined whether ICAM-1 contributes to this process. Genetic deletion (ICAM-1 knockout mice) or siRNA-mediated knockdown of ICAM-1 in isolated murine and human macrophages significantly impaired apoptotic cell (AC) engulfment. Impairment in the engulfment of Jurkat T cells, neutrophils, and epithelial cells was confirmed ex vivo by inflammatory macrophages and in vivo by thioglycolate-recruited peritoneal macrophages. Decreased efferocytosis was also seen in vitro and in vivo with inhibition of ICAM-1 adhesive interactions, using a function blocking anti-ICAM-1 antibody. Mechanistically, it was found that ICAM-1 actively redistributes to cluster around engulfed ACs to facilitate macrophage-AC binding. Our findings define a new role for ICAM-1 in promoting macrophage efferocytosis, a critical process in the resolution of inflammation and restoration of tissue homeostasis.
Collapse
Affiliation(s)
- Hannah L Wiesolek
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Triet M Bui
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Joseph J Lee
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Prarthana Dalal
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ariel Finkielsztein
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ayush Batra
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Edward B Thorp
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ronen Sumagin
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
19
|
Bui TM, Sumagin R. Progressing from Recurring Tissue Injury to Genomic Instability: A New Mechanism of Neutrophil Pathogenesis. DNA Cell Biol 2019; 38:747-753. [PMID: 31188020 PMCID: PMC7643757 DOI: 10.1089/dna.2019.4842] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/14/2019] [Accepted: 05/14/2019] [Indexed: 12/11/2022] Open
Abstract
Aberrant neutrophil (PMN) infiltration of the intestinal mucosa is a hallmark of inflammatory bowel diseases, including Crohn's disease and ulcerative colitis. While the genotoxic function of PMNs and its implications in carcinogenesis have been primarily associated with oxidative stress, recent work by Butin-Israeli and colleagues has defined a novel mechanism where PMN-derived microparticles through the delivery and activity of specific miRNAs promoted formation of double-strand breaks (DSBs), and in parallel, suppressed DSB repair through the downregulation of lamin B1 and Rad51. Respective downregulation of these two proteins compromised the nuclear envelope and high-fidelity repair by homologous recombination, increasing DSB accumulation and aneuploidy. This discovery defined a novel mode of action where PMN-mediated suppression of DSB repair leading to genomic instability in the injured mucosa may facilitate progression toward colorectal cancer.
Collapse
Affiliation(s)
- Triet M. Bui
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Ronen Sumagin
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
20
|
Un-JAMming atherosclerotic arteries: JAM-L as a target to attenuate plaque development. Clin Sci (Lond) 2019; 133:1581-1585. [PMID: 31331991 DOI: 10.1042/cs20190541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/20/2019] [Revised: 06/20/2019] [Accepted: 07/02/2019] [Indexed: 12/18/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease and a major driver of heart attack and stroke. Atherosclerosis development is driven by the infiltration of leukocytes, including monocytes and neutrophils, among other inflammatory cells into the artery wall, monocyte differentiation to macrophages and uptake of oxidized low density lipoprotein. Macrophage activation and inflammatory cytokine production are major factors which drive ongoing inflammation and plaque development. Identification of novel pathways driving this on-going inflammatory process may provide new opportunities for therapeutic intervention. In their article published in Clinical Science (2019) (vol 133, 1215-1228), Sun and colleagues demonstrate a novel role for the junction adhesion molecule-like (JAML) protein in driving on-going atherosclerotic plaque inflammation and plaque development. They report that JAML is expressed in macrophages and other cells in atherosclerotic plaques in both humans and mice, and that silencing JAML expression attenuates atherosclerotic plaque progression in mouse models of early and late stage plaque development. They demonstrate that JAML is required for oxidized-low density lipoprotein (OxLDL)-induced up-regulation of inflammatory cytokine production by macrophages, pointing to it as a potential therapeutic target for reducing ongoing plaque inflammation.
Collapse
|
21
|
Silencing of junctional adhesion molecule-like protein attenuates atherogenesis and enhances plaque stability in ApoE -/- mice. Clin Sci (Lond) 2019; 133:1215-1228. [PMID: 31101724 DOI: 10.1042/cs20180561] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/25/2018] [Revised: 04/06/2019] [Accepted: 05/17/2019] [Indexed: 02/06/2023]
Abstract
Background: Although junctional adhesion molecule-like protein (JAML) has recently been implicated in leukocyte recruitment during inflammation and wound repair, its role in atherosclerosis remains to be elucidated. Methods and results: First, we showed that JAML was strongly expressed in atherosclerotic plaques of cardiovascular patients. Similar results were obtained with atherosclerotic plaques of ApoE-/- mice. Co-immunofluorescence staining showed that JAML was mainly expressed in macrophages. Enhanced expression of JAML in cultured macrophages was observed following exposure of the cells to oxLDL. The functional role of JAML in atherosclerosis and macrophages function was assessed by interference of JAML with shRNA in vivo and siRNA in vitro Silencing of JAML in mice significantly attenuated atherosclerotic lesion formation, reduced necrotic core area, increased plaque fibrous cap thickness, decreased macrophages content and inflammation. In addition, histological staining showed that JAML deficiency promoted plaques to stable phenotype. In vitro, JAML siRNA treatment lowered the expression of inflammatory cytokines in macrophages treated with oxLDL. The mechanism by which JAML mediated the inflammatory responses may be related to the ERK/NF-κB activation. Conclusions: Our results demonstrated that therapeutic drugs which antagonize the function of JAML may be a potentially effective approach to attenuate atherogenesis and enhance plaque stability.
Collapse
|
22
|
Phillipson M, Kubes P. The Healing Power of Neutrophils. Trends Immunol 2019; 40:635-647. [PMID: 31160208 DOI: 10.1016/j.it.2019.05.001] [Citation(s) in RCA: 289] [Impact Index Per Article: 48.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/20/2019] [Revised: 05/01/2019] [Accepted: 05/01/2019] [Indexed: 11/28/2022]
Abstract
Neutrophils promptly accumulate in large numbers at sites of tissue injury. Injuries to the skin or mucosae disrupt barriers against the external environment, and the bactericidal actions of neutrophils are important in preventing microbial invasion. Neutrophils have also been associated with exacerbated inflammation, for example in non-healing wounds or in conditions such as inflammatory bowel disease (IBD). However, additional neutrophil functions important for angiogenesis and tissue restoration have been uncovered in models of sterile and ischemic injury, as well as in tumors. These functions are also relevant in healing skin and mucosal wounds, and can be impaired in conditions associated with non-healing wounds, such as diabetes. Here, we discuss our current understanding of neutrophil contributions to healing, and how the latter can be compromised in disease.
Collapse
Affiliation(s)
- Mia Phillipson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Paul Kubes
- Snyder Institute of Infection, Immunity, and Inflammation, University of Calgary, Calgary, Alberta, T2N 4N1, Canada; Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, T2N 4N1, Canada.
| |
Collapse
|
23
|
Zhang Z, Long C, Li X, Xie Q, Song M, Zhang Y. CEACAM-1 promotes myocardial injury following coxsackievirus infection by regulating the coxsackievirus-adenovirus receptor. Medicine (Baltimore) 2019; 98:e15629. [PMID: 31083261 PMCID: PMC6531060 DOI: 10.1097/md.0000000000015629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE To determine the effects and mechanism of carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM-1, CC1)-mediated regulation of the Coxsackie and Adenovirus Receptor (CAR) after Coxsackievirus B3 (CVB3) infection. METHODS A mouse CC1 overexpression recombinant virus was constructed, followed by insertion of a pLVX-CEACAM 1-zsgreen-puro (rLV-CEACAM 1) plasmid into the recombinant retrovirus. Cardiac myocytes were assigned into different groups according to various treatments. The apoptosis rate and cell activity in each group were observed. Further, CAR expression and SYK, IL-1β, and p-SYK levels were measured. RESULTS The recombinant retrovirus titer was measured as 1.5 × 10 TUs/ml. The apoptosis rate of cardiac myocytes in the CC1 overexpression plus CVB3 group was significantly elevated, and the relative expression of the CAR gene was the highest in the CC1 overexpression plus CVB3 group. TNF-α and IL-1β levels increased due to CC1 overexpression and further increased after CVB3 infection. CAR protein expression also changed along with the levels of CC1, SYK, and TNF-α after infection. CONCLUSION CC1 may promote CAR expression after CVB3 infection and regulate CAR protein expression by activating the CC1-SYK-TNF-α signaling axis during the infection process.
Collapse
Affiliation(s)
- Zaiyong Zhang
- Department of Cardiology, Panyu Central Hospital
- Cardiovascular Institute of Panyu District
- School of Life Sciences, South China Normal University
| | - Cheng Long
- School of Life Sciences, South China Normal University
| | - Xinzhong Li
- Department of Cardiology, Nanfang Hospital, Southern Medical University
| | - Qiang Xie
- Department of Cardiology, Panyu Central Hospital
- Cardiovascular Institute of Panyu District
| | - Mingcai Song
- Department of Cardiology, Panyu Central Hospital
- Cardiovascular Institute of Panyu District
| | - Yulan Zhang
- Department of Ultrasound, Guangdong Women and Children Hospital, Guangzhou, China
| |
Collapse
|
24
|
Roh SE, Jeong Y, Kang MH, Bae YS. Junctional adhesion molecules mediate transendothelial migration of dendritic cell vaccine in cancer immunotherapy. Cancer Lett 2018; 434:196-205. [PMID: 30055289 DOI: 10.1016/j.canlet.2018.07.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/26/2018] [Revised: 06/21/2018] [Accepted: 07/21/2018] [Indexed: 12/14/2022]
Abstract
In vitro generated dendritic cells (DCs) have been studied in cancer immunotherapy for decades. However, the detailed molecular mechanism underlying transendothelial migration (TEM) of DC vaccine across the endothelial barrier to regional lymph nodes (LNs) remains largely unknown. Here, we found that junctional adhesion molecule (JAM)-Like (JAML) is involved in the TEM of mouse bone marrow-derived DCs (BMDCs). Treatment with an anti-JAML antibody or JAML knock-down significantly reduced the TEM activity of BMDCs, leading to impairment of DC-based cancer immunotherapy. We found that the interaction of JAML of BMDCs with the coxsackie and adenovirus receptor of endothelial cells plays a crucial role in the TEM of BMDCs. On the other hand, human monocyte-derived DCs (MoDCs) did not express the JAML protein but still showed normal TEM activity. We found that MoDCs express only JAM1 and that the homophilic interaction of JAM1 is essential for MoDC TEM across a HUVEC monolayer. Our findings suggest that specific JAM family members play an important role in the TEM of in vitro-generated mouse and human DCs from the inoculation site to regional LNs in DC-based cancer immunotherapy.
Collapse
Affiliation(s)
- Seung-Eon Roh
- Department of Biological Sciences, Science Research Center (SRC) for Immune Research on Non-lymphoid Organ (CIRNO), Sungkyunkwan University, Jangan-gu, Suwon, Gyeonggi-do, 16419, South Korea
| | - Yideul Jeong
- Department of Biological Sciences, Science Research Center (SRC) for Immune Research on Non-lymphoid Organ (CIRNO), Sungkyunkwan University, Jangan-gu, Suwon, Gyeonggi-do, 16419, South Korea
| | - Myeong-Ho Kang
- Department of Biological Sciences, Science Research Center (SRC) for Immune Research on Non-lymphoid Organ (CIRNO), Sungkyunkwan University, Jangan-gu, Suwon, Gyeonggi-do, 16419, South Korea
| | - Yong-Soo Bae
- Department of Biological Sciences, Science Research Center (SRC) for Immune Research on Non-lymphoid Organ (CIRNO), Sungkyunkwan University, Jangan-gu, Suwon, Gyeonggi-do, 16419, South Korea.
| |
Collapse
|
25
|
Esnault S, Hebert AS, Jarjour NN, Coon JJ, Mosher DF. Proteomic and Phosphoproteomic Changes Induced by Prolonged Activation of Human Eosinophils with IL-3. J Proteome Res 2018; 17:2102-2111. [PMID: 29706072 DOI: 10.1021/acs.jproteome.8b00057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/17/2022]
Abstract
Purified human eosinophils treated for 18-24 h with IL-3 adopt a unique activated phenotype marked by increased reactivity to aggregated immunoglobulin-G (IgG). To characterize this phenotype, we quantified protein abundance and phosphorylation by multiplexed isobaric labeling combined with high-resolution mass spectrometry. Purified blood eosinophils of five individuals were treated with IL-3 or no cytokine for 20 h, and comparative data were obtained on abundance of 5385 proteins and phosphorylation at 7330 sites. The 1150 proteins that were significantly up-regulated ( q < 0.05, pairwise t test with Benjamini-Hochberg correction) by IL-3 included the IL3RA and CSF2RB subunits of the IL-3 receptor, the low-affinity receptor for IgG (FCGR2B), 96 proteins involved in protein translation, and 55 proteins involved in cytoskeleton organization. Among the 703 proteins that decreased were 78 mitochondrial proteins. Dynamic regulation of protein phosphorylation was detected at 4218 sites. These included multiple serines in CSF2RB; Y694 of STAT5, a key site of activating phosphorylation downstream of IL3RA/CSF2RB; and multiple sites in RPS6KA1, RPS6, and EIF4B, which are responsible for translational initiation. We conclude that IL-3 up-regulates overall protein synthesis and targets specific proteins for up-regulation, including its own receptor.
Collapse
Affiliation(s)
- Stephane Esnault
- Department of Medicine , University of Wisconsin , Madison , Wisconsin 53792 , United States
| | - Alexander S Hebert
- Department of Chemistry , University of Wisconsin , Madison , Wisconsin 53706 , United States
| | - Nizar N Jarjour
- Department of Medicine , University of Wisconsin , Madison , Wisconsin 53792 , United States
| | - Joshua J Coon
- Department of Chemistry , University of Wisconsin , Madison , Wisconsin 53706 , United States.,Department of Biomolecular Chemistry , University of Wisconsin , Madison , Wisconsin 53706 , United States.,Morgridge Institute for Research , Madison , Wisconsin 53715 , United States.,Genome Center of Wisconsin , Madison , Wisconsin 53706 , United States
| | - Deane F Mosher
- Department of Medicine , University of Wisconsin , Madison , Wisconsin 53792 , United States.,Department of Biomolecular Chemistry , University of Wisconsin , Madison , Wisconsin 53706 , United States
| |
Collapse
|
26
|
Konjar Š, Ferreira C, Blankenhaus B, Veldhoen M. Intestinal Barrier Interactions with Specialized CD8 T Cells. Front Immunol 2017; 8:1281. [PMID: 29075263 PMCID: PMC5641586 DOI: 10.3389/fimmu.2017.01281] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/29/2017] [Accepted: 09/25/2017] [Indexed: 01/09/2023] Open
Abstract
The trillions of microorganisms that reside in the gastrointestinal tract, essential for nutrient absorption, are kept under control by a single cell barrier and large amounts of immune cells. Intestinal epithelial cells (IECs) are critical in establishing an environment supporting microbial colonization and immunological tolerance. A large population of CD8+ T cells is in direct and constant contact with the IECs and the intraepithelial lymphocytes (IELs). Due to their location, at the interphase of the intestinal lumen and external environment and the host tissues, they seem ideally positioned to balance immune tolerance and protection to preserve the fragile intestinal barrier from invasion as well as immunopathology. IELs are a heterogeneous population, with a large innate-like contribution of unknown specificity, intercalated with antigen-specific tissue-resident memory T cells. In this review, we provide a comprehensive overview of IEL physiology and how they interact with the IECs and contribute to immune surveillance to preserve intestinal homeostasis and host-microbial relationships.
Collapse
Affiliation(s)
- Špela Konjar
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Cristina Ferreira
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Birte Blankenhaus
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Marc Veldhoen
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
27
|
Interleukin-17A Aggravates Middle Ear Injury Induced by Streptococcus pneumoniae through the p38 Mitogen-Activated Protein Kinase Signaling Pathway. Infect Immun 2017; 85:IAI.00438-17. [PMID: 28739823 DOI: 10.1128/iai.00438-17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/19/2017] [Accepted: 07/11/2017] [Indexed: 12/26/2022] Open
Abstract
Acute otitis media (AOM) is one of the most common bacterial infectious diseases in children aged 2 to 7 years worldwide. We previously demonstrated that interleukin-17A (IL-17A) promotes an acute inflammatory response characterized by the influx of neutrophils into the middle ear cavity during Streptococcus pneumoniae-induced AOM. In general, the inflammatory response is viewed as an effector that frequently causes local tissue damage. However, little is known about the pathogenic effects of IL-17A in AOM. Here, we investigated the pathogenic effects of IL-17A by using wild-type (WT) and IL-17A knockout (KO) mouse models. The results showed that the pathogenic effects of AOM, including weight loss, histopathological changes, and proinflammatory cytokine production, were more severe in WT mice than in IL-17A KO mice, suggesting that IL-17A aggravates tissue damage in AOM. Furthermore, these pathogenic effects were found to be dependent on p38 mitogen-activated protein kinase (MAPK) and could be reversed in the presence of a p38 MAPK-specific inhibitor. It was also demonstrated that IL-17A promoted the production of neutrophil myeloperoxidase (MPO) through the p38 MAPK signaling pathway, which was responsible for the middle ear tissue injury. These data support the conclusion that IL-17A contributes to middle ear injury through the p38 MAPK signaling pathway.
Collapse
|
28
|
Abstract
Polymorphonuclear neutrophils (PMNs) are innate immune system cells that play an essential role in eradicating invading pathogens. PMN migration to sites of infection/inflammation requires exiting the microcirculation and subsequent crossing of epithelial barriers in mucosa-lined organs such as the lungs and intestines. Although these processes usually occur without significant damage to surrounding host tissues, dysregulated/excessive PMN transmigration and resultant bystander-tissue damage are characteristic of numerous mucosal inflammatory disorders. Mechanisms controlling PMN extravasation have been well characterized, but the molecular details regarding regulation of PMN migration across mucosal epithelia are poorly understood. Given that PMN migration across mucosal epithelia is strongly correlated with disease symptoms in many inflammatory mucosal disorders, enhanced understanding of the mechanisms regulating PMN transepithelial migration should provide insights into clinically relevant tissue-targeted therapies aimed at ameliorating PMN-mediated bystander-tissue damage. This review will highlight current understanding of the molecular interactions between PMNs and mucosal epithelia and the associated functional consequences.
Collapse
Affiliation(s)
- Jennifer C Brazil
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Charles A Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
29
|
Slater TW, Finkielsztein A, Mascarenhas LA, Mehl LC, Butin-Israeli V, Sumagin R. Neutrophil Microparticles Deliver Active Myeloperoxidase to Injured Mucosa To Inhibit Epithelial Wound Healing. THE JOURNAL OF IMMUNOLOGY 2017; 198:2886-2897. [PMID: 28242649 DOI: 10.4049/jimmunol.1601810] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 10/24/2016] [Accepted: 01/29/2017] [Indexed: 12/19/2022]
Abstract
Neutrophil (PMN) infiltration of the intestinal mucosa often leads to severe epithelial injury; however, how this process occurs is unclear. This article describes a novel mechanism whereby membrane-derived microparticles released by tissue infiltrating PMNs (PMN-MPs) serve as shuttles to protect and deliver active mediators to locally modulate cellular function during inflammation. Specifically, myeloperoxidase (MPO), which is abundantly expressed in PMN azurophilic granules and is used for microbial killing, was found to be mobilized to the PMN surface and subsequently released in association with PMN-MPs upon PMN activation and binding to intestinal epithelial cells (IECs). The enzymatic activity of PMN-MP-associated MPO was enhanced compared with soluble protein, leading to potent inhibition of wound closure following PMN-MP binding to IECs. Importantly, localized microinjection of PMN-MPs into wounded colonic mucosa was sufficient to impair epithelial wound healing in vivo. PMN-MP/MPO-dependent inhibition of IEC wound healing was due to impaired IEC migration and proliferation, resulting from impeded actin dynamics, cell spreading, and cell cycle arrest. Thus, our findings provide new insight into mechanisms governing PMN-induced tissue injury and implicate PMN-MPs and MPO as important regulators of cellular function.
Collapse
Affiliation(s)
- Thomas W Slater
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Ariel Finkielsztein
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Lorraine A Mascarenhas
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Lindsey C Mehl
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Veronika Butin-Israeli
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Ronen Sumagin
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| |
Collapse
|
30
|
Sharma P, Martis PC, Excoffon KJDA. Adenovirus transduction: More complicated than receptor expression. Virology 2016; 502:144-151. [PMID: 28049062 DOI: 10.1016/j.virol.2016.12.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/30/2016] [Revised: 12/02/2016] [Accepted: 12/19/2016] [Indexed: 02/06/2023]
Abstract
The abundance and accessibility of a primary virus receptor are critical factors that impact the susceptibility of a host cell to virus infection. The Coxsackievirus and adenovirus receptor (CAR) has two transmembrane isoforms that occur due to alternative splicing and differ in localization and function in polarized epithelia. To determine the relevance of isoform-specific expression across cell types, the abundance and localization of both isoforms were determined in ten common cell lines, and correlated with susceptibility to adenovirus transduction relative to polarized primary human airway epithelia. Data show that the gene and protein expression for each isoform of CAR varies significantly between cell lines and polarization, as indicated by high transepithelial resistance, is inversely related to adenovirus transduction. In summary, the variability of polarity and isoform-specific expression among model cells are critical parameters that must be considered when evaluating the clinical relevance of potential adenovirus-mediated gene therapy and anti-adenovirus strategies.
Collapse
Affiliation(s)
- Priyanka Sharma
- Department of Biological Sciences, Wright State University, Dayton, OH, USA
| | - Prithy C Martis
- Biomedical Sciences PhD Program, Wright State University, Dayton, OH 45435, USA
| | - Katherine J D A Excoffon
- Department of Biological Sciences, Wright State University, Dayton, OH, USA; Biomedical Sciences PhD Program, Wright State University, Dayton, OH 45435, USA.
| |
Collapse
|
31
|
Luissint AC, Parkos CA, Nusrat A. Inflammation and the Intestinal Barrier: Leukocyte-Epithelial Cell Interactions, Cell Junction Remodeling, and Mucosal Repair. Gastroenterology 2016; 151:616-32. [PMID: 27436072 PMCID: PMC5317033 DOI: 10.1053/j.gastro.2016.07.008] [Citation(s) in RCA: 374] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 05/02/2016] [Revised: 06/13/2016] [Accepted: 07/06/2016] [Indexed: 02/06/2023]
Abstract
The intestinal tract is lined by a single layer of columnar epithelial cells that forms a dynamic, permeable barrier allowing for selective absorption of nutrients, while restricting access to pathogens and food-borne antigens. Precise regulation of epithelial barrier function is therefore required for maintaining mucosal homeostasis and depends, in part, on barrier-forming elements within the epithelium and a balance between pro- and anti-inflammatory factors in the mucosa. Pathologic states, such as inflammatory bowel disease, are associated with a leaky epithelial barrier, resulting in excessive exposure to microbial antigens, recruitment of leukocytes, release of soluble mediators, and ultimately mucosal damage. An inflammatory microenvironment affects epithelial barrier properties and mucosal homeostasis by altering the structure and function of epithelial intercellular junctions through direct and indirect mechanisms. We review our current understanding of complex interactions between the intestinal epithelium and immune cells, with a focus on pathologic mucosal inflammation and mechanisms of epithelial repair. We discuss leukocyte-epithelial interactions, as well as inflammatory mediators that affect the epithelial barrier and mucosal repair. Increased knowledge of communication networks between the epithelium and immune system will lead to tissue-specific strategies for treating pathologic intestinal inflammation.
Collapse
Affiliation(s)
- Anny-Claude Luissint
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Charles A Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Asma Nusrat
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan.
| |
Collapse
|
32
|
Sumagin R, Brazil JC, Nava P, Nishio H, Alam A, Luissint AC, Weber DA, Neish AS, Nusrat A, Parkos CA. Neutrophil interactions with epithelial-expressed ICAM-1 enhances intestinal mucosal wound healing. Mucosal Immunol 2016; 9:1151-62. [PMID: 26732677 PMCID: PMC4935657 DOI: 10.1038/mi.2015.135] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/06/2015] [Accepted: 11/06/2015] [Indexed: 02/04/2023]
Abstract
A characteristic feature of gastrointestinal tract inflammatory disorders, such as inflammatory bowel disease, is polymorphonuclear neutrophil (PMN) transepithelial migration (TEM) and accumulation in the gut lumen. PMN accumulation within the intestinal mucosa contributes to tissue injury. Although epithelial infiltration by large numbers of PMNs results in mucosal injury, we found that PMN interactions with luminal epithelial membrane receptors may also play a role in wound healing. Intercellular adhesion molecule-1 (ICAM-1) is a PMN ligand that is upregulated on apical surfaces of intestinal epithelial cells under inflammatory conditions. In our study, increased expression of ICAM-1 resulted in enhanced PMN binding to the apical epithelium, which was associated with reduced PMN apoptosis. Following TEM, PMN adhesion to ICAM-1 resulted in activation of Akt and β-catenin signaling, increased epithelial-cell proliferation, and wound healing. Such responses were ICAM-1 dependent as engagement of epithelial ICAM-1 by antibody-mediated cross-linking yielded similar results. Furthermore, using an in-vivo biopsy-based, colonic-mucosal-injury model, we demonstrated epithelial ICAM-1 has an important role in activation of epithelial Akt and β-catenin signaling and wound healing. These findings suggest that post-migrated PMNs within the intestinal lumen can regulate epithelial homeostasis, thereby identifying ICAM-1 as a potential therapeutic target for promoting mucosal wound healing.
Collapse
Affiliation(s)
- R Sumagin
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - J C Brazil
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - P Nava
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, Mexico, Mexico
| | - H Nishio
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - A Alam
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - A C Luissint
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - D A Weber
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - A S Neish
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - A Nusrat
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - C A Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
33
|
Butin-Israeli V, Houser MC, Feng M, Thorp EB, Nusrat A, Parkos CA, Sumagin R. Deposition of microparticles by neutrophils onto inflamed epithelium: a new mechanism to disrupt epithelial intercellular adhesions and promote transepithelial migration. FASEB J 2016; 30:4007-4020. [PMID: 27553226 DOI: 10.1096/fj.201600734r] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/13/2016] [Accepted: 08/08/2016] [Indexed: 12/22/2022]
Abstract
Neutrophil [polymorphonuclear leukocyte (PMN)] transepithelial migration (TEM) is a hallmark of inflammatory mucosal disorders. PMN TEM is associated with epithelial injury; however, mechanisms involved in this process are not well defined. The current work describes a new mechanism whereby deposition of PMN membrane-derived microparticles (PMN-MPs) onto intestinal epithelial cells (IECs) during TEM leads to loss of epithelial cadherins, thus promoting epithelial injury and increased PMN recruitment. PMN-MPs secreted by activated PMNs during TEM displayed a high level of enzymatically active matrix metalloproteinase 9 (MMP-9), and were capable of mediating potent effects on IEC integrity. Isolated PMN-MPs efficiently bound to IEC monolayers and induced cleavage of desmoglein-2 (DSG-2) but not E-cadherin, leading to disruption of IEC intercellular adhesions. Furthermore, PMN-MP binding to intestinal epithelium in vitro in transwell assays and in vivo in ligated intestinal loop preparations facilitated increases in PMN TEM. These effects were MMP-9 dependent and were reversed in the presence of specific pharmacological inhibitors. Finally, we demonstrated that IEC Dsg-2 serves as a barrier for migrating PMNs, and its removal by PMN-MP-associated MMP-9 facilitates PMN trafficking across epithelial layers. Our findings thus implicate PMN-MPs in PMN-mediated inflammation and epithelial damage, as observed in inflammatory disorders of mucosal surfaces.-Butin-Israeli, V., Houser, M. C., Feng, M., Thorp, E. B., Nusrat, A., Parkos, C. A, Sumagin, R. Deposition of microparticles by neutrophils onto inflamed epithelium: a new mechanism to disrupt epithelial intercellular adhesions and promote transepithelial migration.
Collapse
Affiliation(s)
- Veronika Butin-Israeli
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Madelyn C Houser
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia, USA; and
| | - Mingli Feng
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Edward B Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Asma Nusrat
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Charles A Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Ronen Sumagin
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA;
| |
Collapse
|
34
|
Morton PE, Hicks A, Ortiz-Zapater E, Raghavan S, Pike R, Noble A, Woodfin A, Jenkins G, Rayner E, Santis G, Parsons M. TNFα promotes CAR-dependent migration of leukocytes across epithelial monolayers. Sci Rep 2016; 6:26321. [PMID: 27193388 PMCID: PMC4872059 DOI: 10.1038/srep26321] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/08/2016] [Accepted: 05/03/2016] [Indexed: 12/24/2022] Open
Abstract
Trans-epithelial migration (TEpM) of leukocytes during inflammation requires engagement with receptors expressed on the basolateral surface of the epithelium. One such receptor is Coxsackie and Adenovirus Receptor (CAR) that binds to Junctional Adhesion Molecule-like (JAM-L) expressed on leukocytes. Here we provide the first evidence that efficient TEpM of monocyte-derived THP-1 cells requires and is controlled by phosphorylation of CAR. We show that TNFα acts in a paracrine manner on epithelial cells via a TNFR1-PI3K-PKCδ pathway leading to CAR phosphorylation and subsequent transmigration across cell junctions. Moreover, we show that CAR is hyper-phosphorylated in vivo in acute and chronic lung inflammation models and this response is required to facilitate immune cell recruitment. This represents a novel mechanism of feedback between leukocytes and epithelial cells during TEpM and may be important in controlling responses to pro-inflammatory cytokines in pathological settings.
Collapse
Affiliation(s)
- Penny E Morton
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guys Campus, London, SE1 1UL, UK
| | - Alexander Hicks
- Division of Asthma, Allergy &Lung Biology, King's College London, 5th Floor Tower Wing, Guy's Hospital Campus, London, SE1 1UL, UK
| | - Elena Ortiz-Zapater
- Division of Asthma, Allergy &Lung Biology, King's College London, 5th Floor Tower Wing, Guy's Hospital Campus, London, SE1 1UL, UK
| | - Swetavalli Raghavan
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guys Campus, London, SE1 1UL, UK
| | - Rosemary Pike
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guys Campus, London, SE1 1UL, UK
| | - Alistair Noble
- Division of Asthma, Allergy &Lung Biology, King's College London, 5th Floor Tower Wing, Guy's Hospital Campus, London, SE1 1UL, UK
| | - Abigail Woodfin
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary, University of London, EC1M 6BQ, UK
| | - Gisli Jenkins
- Respiratory Research Unit, University of Nottingham, Clinical Sciences Building, City Campus, Nottingham, NG5 1PB, UK
| | - Emma Rayner
- Public Health England, Salisbury, Wiltshire, SP4 0JG, UK
| | - George Santis
- Division of Asthma, Allergy &Lung Biology, King's College London, 5th Floor Tower Wing, Guy's Hospital Campus, London, SE1 1UL, UK
| | - Maddy Parsons
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guys Campus, London, SE1 1UL, UK
| |
Collapse
|
35
|
Parkos CA. Neutrophil-Epithelial Interactions: A Double-Edged Sword. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1404-16. [PMID: 27083514 DOI: 10.1016/j.ajpath.2016.02.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 12/09/2015] [Revised: 01/25/2016] [Accepted: 02/01/2016] [Indexed: 02/07/2023]
Abstract
In recent years, it has become clear that innate immune cells termed neutrophils act as double-edged swords by playing essential roles in clearing infection but also causing tissue damage, yet being critical for wound healing. Neutrophil recruitment to sites of injured tissue or infection has been well studied, and many of the molecular events that regulate passage of leukocytes out of the microcirculation are now understood. However, after exiting the circulation, the molecular details that regulate neutrophil passage to end targets, such mucosal surfaces, are just beginning to be appreciated. Given that migration of neutrophils across mucosal epithelia is associated with disease symptoms and disruption of critical barrier function in disorders such as inflammatory bowel disease, there has been long-standing interest in understanding the molecular basis and functional consequences of neutrophil-epithelial interactions. It is a great honor that my work was recognized by the Rous-Whipple Award this past year, giving me the opportunity to summarize what we have learned during the past few decades about leukocyte interactions with epithelial cells.
Collapse
Affiliation(s)
- Charles A Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan.
| |
Collapse
|
36
|
Singh V, Yeoh BS, Chassaing B, Zhang B, Saha P, Xiao X, Awasthi D, Shashidharamurthy R, Dikshit M, Gewirtz A, Vijay-Kumar M. Microbiota-inducible Innate Immune, Siderophore Binding Protein Lipocalin 2 is Critical for Intestinal Homeostasis. Cell Mol Gastroenterol Hepatol 2016; 2:482-498.e6. [PMID: 27458605 PMCID: PMC4957954 DOI: 10.1016/j.jcmgh.2016.03.007] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Lipocalin 2 (Lcn2) is a multifunctional innate immune protein whose expression closely correlates with extent of intestinal inflammation. However, whether Lcn2 plays a role in the pathogenesis of gut inflammation is unknown. Herein, we investigated the extent to which Lcn2 regulates inflammation and gut bacterial dysbiosis in mouse models of IBD. METHODS Lcn2 expression was monitored in murine colitis models and upon microbiota ablation/restoration. WT and Lcn2 knockout (Lcn2KO) mice were analyzed for gut bacterial load, composition by 16S rRNA gene pyrosequencing and, their colitogenic potential by co-housing with Il-10KO mice. Acute (dextran sodium sulfate) and chronic (IL-10R neutralization and T-cell adoptive transfer) colitis was induced in WT and Lcn2KO mice with or without antibiotics. RESULTS Lcn2 expression was dramatically induced upon inflammation and was dependent upon presence of a gut microbiota and MyD88 signaling. Use of bone-marrow chimeric mice revealed non-immune cells are the major contributors of circulating Lcn2. Lcn2KO mice exhibited elevated levels of entA-expressing gut bacteria burden and, moreover, a broadly distinct bacterial community relative to WT littermates. Lcn2KO mice developed highly colitogenic T-cells and exhibited exacerbated colitis upon exposure to DSS or neutralization of IL-10. Such exacerbated colitis could be prevented by antibiotic treatment. Moreover, exposure to the microbiota of Lcn2KO mice, via cohousing, resulted in severe colitis in Il-10KO mice. CONCLUSION Lcn2 is a bacterially-induced, MyD88-dependent, protein that play an important role in gut homeostasis and a pivotal role upon challenge. Hence, therapeutic manipulation of Lcn2 levels may provide a strategy to help manage diseases driven by alteration of the gut microbiota.
Collapse
Affiliation(s)
- Vishal Singh
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania
| | - Beng San Yeoh
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania
| | - Benoit Chassaing
- Center for Inflammation, Immunity and Infection, Institute of Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Benyue Zhang
- Center for Inflammation, Immunity and Infection, Institute of Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Piu Saha
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania
| | - Xia Xiao
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania
| | - Deepika Awasthi
- Pharmacology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India
| | | | - Madhu Dikshit
- Pharmacology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India
| | - Andrew Gewirtz
- Center for Inflammation, Immunity and Infection, Institute of Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Matam Vijay-Kumar
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania,Department of Medicine, The Pennsylvania State University Medical Center, Hershey, Pennsylvania,Correspondence Address correspondence to: Matam Vijay-Kumar, PhD, Department of Nutritional Sciences 222, Chandlee Laboratory, The Pennsylvania State University, University Park, Pennsylvania 16802. fax: (814) 863-6103.Department of Nutritional Sciences 222Chandlee LaboratoryThe Pennsylvania State UniversityUniversity ParkPennsylvania 16802
| |
Collapse
|
37
|
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
|
38
|
IL-21 and probiotic therapy improve Th17 frequencies, microbial translocation, and microbiome in ARV-treated, SIV-infected macaques. Mucosal Immunol 2016; 9:458-67. [PMID: 26286233 PMCID: PMC4760912 DOI: 10.1038/mi.2015.75] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/20/2015] [Accepted: 07/19/2015] [Indexed: 02/04/2023]
Abstract
Increased mortality in antiretroviral (ARV)-treated, HIV-infected individuals has been attributed to persistent immune dysfunction, in part due to abnormalities at the gastrointestinal barrier. In particular, the poor reconstitution of gastrointestinal Th17 cells correlates with residual translocation of dysbiotic, immunostimulatory microflora across a compromised intestinal epithelial barrier. We have previously demonstrated that oral probiotics promote increased intestinal CD4(+) T-cell reconstitution during ARV treatment in a non-human primate model of HIV infection; however, essential mucosal T-cell subsets, such as Th17 cells, had limited recovery. Here, we sought to promote Th17 cell recovery by administering interleukin (IL)-21 to a limited number of ARV-treated, probiotic-supplemented, Simian Immunodeficiency Virus (SIV)-infected pigtailed macaques. We demonstrate that probiotic and IL-21 supplementation of ARVs are associated with enhanced polyfunctional Th17 expansion and reduced markers of microbial translocation and dysbiosis as compared with infected controls receiving ARVs alone. Importantly, treatment resulted in fewer morbidities compared with controls, and was independent of increased immune activation or loss of viral suppression. We propose that combining ARVs with therapeutics aimed at restoring intestinal stasis may significantly improve disease prognosis of ARV-treated, HIV-infected individuals.
Collapse
|
39
|
Reeves RK, Burgener A, Klatt NR. Targeting the gastrointestinal tract to develop novel therapies for HIV. Clin Pharmacol Ther 2015; 98:381-6. [PMID: 26179624 DOI: 10.1002/cpt.186] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/19/2015] [Accepted: 07/10/2015] [Indexed: 01/10/2023]
Abstract
Despite the use of antiretroviral therapy (ART), which delays and/or prevents AIDS pathogenesis, human immunodeficiency virus (HIV)-infected individuals continue to face increased morbidities and mortality rates compared with uninfected individuals. Gastrointestinal (GI) mucosal dysfunction is a key feature of HIV infection, and is associated with mortality. In this study, we review current knowledge about mucosal dysfunction in HIV infection, and describe potential avenues for therapeutic targets to enhance mucosal function and decrease morbidities and mortalities in HIV-infected individuals.
Collapse
Affiliation(s)
- R K Reeves
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - A Burgener
- National Laboratory for HIV Immunology, Public Health Agency of Canada, Winnipeg, Canada.,Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institute, Sweden
| | - N R Klatt
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA.,Washington National Primate Research Center, Seattle, Washington, USA
| |
Collapse
|
40
|
Hao XP, Lucero CM, Turkbey B, Bernardo ML, Morcock DR, Deleage C, Trubey CM, Smedley J, Klatt NR, Giavedoni LD, Kristoff J, Xu A, Del Prete GQ, Keele BF, Rao SS, Alvord WG, Choyke PL, Lifson JD, Brenchley JM, Apetrei C, Pandrea I, Estes JD. Experimental colitis in SIV-uninfected rhesus macaques recapitulates important features of pathogenic SIV infection. Nat Commun 2015; 6:8020. [PMID: 26282376 PMCID: PMC4544774 DOI: 10.1038/ncomms9020] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/05/2015] [Accepted: 07/08/2015] [Indexed: 02/07/2023] Open
Abstract
Mucosal damage to the gastrointestinal (GI) tract with resulting microbial translocation is hypothesized to significantly contribute to the heightened and persistent chronic inflammation and immune activation characteristic to HIV infection. Here we employ a non-human primate model of chemically induced colitis in SIV-uninfected rhesus macaques that we developed using dextran sulfate sodium (DSS), to directly test this hypothesis. DSS treatment results in GI barrier damage with associated microbial translocation, inflammation and immune activation. The progression and severity of colitis are longitudinally monitored by a magnetic resonance imaging approach. DSS treatment of SIV-infected African green monkeys, a natural host species for SIV that does not manifest GI tract damage or chronic immune activation during infection, results in colitis with elevated levels of plasma SIV RNA, sCD14, LPS, CRP and mucosal CD4+ T-cell loss. Together these results support the hypothesis that GI tract damage leading to local and systemic microbial translocation, and associated immune activation, are important determinants of AIDS pathogenesis.
Collapse
Affiliation(s)
- Xing Pei Hao
- Pathology and Histotechnology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 539, Post Office Box B, Frederick, Maryland 21702, USA
| | - Carissa M. Lucero
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 535, Post Office Box B, Frederick, Maryland 21702, USA
| | - Baris Turkbey
- Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B69F, Bethesda, Maryland 20814, USA
| | - Marcelino L. Bernardo
- Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B69F, Bethesda, Maryland 20814, USA
| | - David R. Morcock
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 535, Post Office Box B, Frederick, Maryland 21702, USA
| | - Claire Deleage
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 535, Post Office Box B, Frederick, Maryland 21702, USA
| | - Charles M. Trubey
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 535, Post Office Box B, Frederick, Maryland 21702, USA
| | - Jeremy Smedley
- Laboratory Animal Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 14D RM 233, 14 Service RD West, Bethesda, Maryland 20814, USA
- Washington National Primate Research Center, University of Washington, 1705 NE Pacific Street, Box 357330, Seattle, Washington 98195, USA
| | - Nichole R. Klatt
- Department of Pharmaceutics, WaNPRC, University of Washington, 3018 Western Avenue, Box 357331, Seattle, Washington 98121, USA
| | - Luis D. Giavedoni
- Department of Virology and Immunology, Southwest National Primate Research Center, Texas Biomedical Research Institute, 7620 NW Loop 410, San Antonio, Texas 78227, USA
| | - Jan Kristoff
- Center for Vaccine Research, University of Pittsburgh, 9044 BST3, 3501 Fifth Avenue, Pittsburgh, Pennsylvania 15261, USA
- School of Public Health, University of Pittsburgh, 9044 BST3, 3501 Fifth Avenue, Pittsburgh, Pennsylvania 15261, USA
| | - Amy Xu
- Center for Vaccine Research, University of Pittsburgh, 9044 BST3, 3501 Fifth Avenue, Pittsburgh, Pennsylvania 15261, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, 9044 BST3, 3501 Fifth Avenue, Pittsburgh, Pennsylvania 15261, USA
| | - Gregory Q. Del Prete
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 535, Post Office Box B, Frederick, Maryland 21702, USA
| | - Brandon F. Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 535, Post Office Box B, Frederick, Maryland 21702, USA
| | - Srinivas S. Rao
- Laboratory Animal Medicine, Vaccine Research Center, NIAID, NIH, BG 40, 40 Convent Drive, Bethesda, Maryland 20814, USA
| | - W. Gregory Alvord
- Statistical Consulting, Data Management Services, Inc., National Cancer Institute at Frederick, Post Office Box B, Frederick, Maryland 21702, USA
| | - Peter L. Choyke
- Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B69F, Bethesda, Maryland 20814, USA
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 535, Post Office Box B, Frederick, Maryland 21702, USA
| | - Jason M. Brenchley
- Immunopathogenesis Section, Lab of Molecular Microbiology, NIAID, NIH, BG 4 RM 201, 4 Memorial Drive, Bethesda, Maryland 20814, USA
| | - Cristian Apetrei
- Center for Vaccine Research, University of Pittsburgh, 9044 BST3, 3501 Fifth Avenue, Pittsburgh, Pennsylvania 15261, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, 9044 BST3, 3501 Fifth Avenue, Pittsburgh, Pennsylvania 15261, USA
| | - Ivona Pandrea
- Center for Vaccine Research, University of Pittsburgh, 9044 BST3, 3501 Fifth Avenue, Pittsburgh, Pennsylvania 15261, USA
- Department of Pathology and School of Medicine, University of Pittsburgh, 9017 Biomedical Science Tower 3, 3501 Fifth Avenue, Pittsburgh, Pennsylvania 15261, USA
| | - Jacob D. Estes
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 535, Post Office Box B, Frederick, Maryland 21702, USA
| |
Collapse
|
41
|
Sumagin R, Parkos CA. Epithelial adhesion molecules and the regulation of intestinal homeostasis during neutrophil transepithelial migration. Tissue Barriers 2015; 3:e969100. [PMID: 25838976 DOI: 10.4161/21688362.2014.969100] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/19/2014] [Accepted: 08/14/2014] [Indexed: 12/19/2022] Open
Abstract
Epithelial adhesion molecules play essential roles in regulating cellular function and maintaining mucosal tissue homeostasis. Some form epithelial junctional complexes to provide structural support for epithelial monolayers and act as a selectively permeable barrier separating luminal contents from the surrounding tissue. Others serve as docking structures for invading viruses and bacteria, while also regulating the immune response. They can either obstruct or serve as footholds for the immune cells recruited to mucosal surfaces. Currently, it is well appreciated that adhesion molecules collectively serve as environmental cue sensors and trigger signaling events to regulate epithelial function through their association with the cell cytoskeleton and various intracellular adapter proteins. Immune cells, particularly neutrophils (PMN) during transepithelial migration (TEM), can modulate adhesion molecule expression, conformation, and distribution, significantly impacting epithelial function and tissue homeostasis. This review discusses the roles of key intestinal epithelial adhesion molecules in regulating PMN trafficking and outlines the potential consequences on epithelial function.
Collapse
Key Words
- AJs, adherens junctions
- CAR, coxsackie and adenovirus receptor
- CLMP, CAR-like protein
- CTLs, cytotoxic T lymphocytes
- CTX, thymocyte Xenopus
- DMs, Desmosomes
- Dsc-2, desmocollin-2
- Dsg-2, desmoglein-2
- E-cadherin, epithelial cadherin
- EGFR, Epithelial growth factor receptor
- EMT, epithelial-mesenchymal transition
- EpCAM, epithelial cell adhesion molecule
- IBD, inflammatory bowel diseases
- ICAM-1, intercellular adhesion molecule-1
- IECs, intestinal epithelial cells
- JAM, junctional adhesion molecules
- LAD, leukocyte adhesion deficiency
- LTB-4, lipid leukotriene B4
- MIP1 α, macrophage inflammatory protein 1 alpha
- MLCK, myosin light chain kinase
- MMPs, matrix metalloproteases
- NF-κB, nuclear factor kappa B
- NO, nitric oxide
- PARS, protease-activated receptors
- PI3K, phosphatidylinositol 3-kinase
- PMN, polymorphonuclear cells
- SGD, specific granule deficiency
- SIRPa, signal regulatory protein alpha
- TEM, transepithelial migration
- TGF-β, transforming growth factor beta
- TIAM1, metastasis-inducing protein 1
- TJs, tight junctions
- TSP-1, thrombospondin-1
- adhesion molecules
- barrier
- cell migration
- epithelial cells
- neutrophils
- sLea, sialyl Lewis A
Collapse
Affiliation(s)
- Ronen Sumagin
- Department of Pathology and Laboratory Medicine; Epithelial Pathobiology and Mucosal Inflammation Unit; Emory University ; Atlanta, GA USA
| | - Charles A Parkos
- Department of Pathology and Laboratory Medicine; Epithelial Pathobiology and Mucosal Inflammation Unit; Emory University ; Atlanta, GA USA
| |
Collapse
|
42
|
Kishida K, Kohyama M, Kurashima Y, Kogure Y, Wang J, Hirayasu K, Suenaga T, Kiyono H, Kunisawa J, Arase H. Negative regulation of DSS-induced experimental colitis by PILRα. Int Immunol 2015; 27:307-14. [DOI: 10.1093/intimm/dxv004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/08/2014] [Accepted: 01/23/2015] [Indexed: 12/16/2022] Open
|
43
|
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: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution 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
|