1
|
Yim J, Hope C, Huelse JM, Graham DK. Prospects of current AXL-targeting therapies in early phase cancer trials. Expert Opin Investig Drugs 2025:1-33. [PMID: 40413629 DOI: 10.1080/13543784.2025.2511178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/22/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025]
Abstract
INTRODUCTION AXL, a member of the TAM (TYRO3, AXL, and MERTK) family of receptor tyrosine kinases, controls pro-tumorigenic signaling cascades and cancer-immunological functions, and promotes drug resistance. Due to AXL's multifaceted role and therapeutic activity in preclinical studies, a variety of AXL inhibitors are being developed and tested in clinical trials for cancer treatment. Some clinical studies are showing promising results for AXL inhibitors as monotherapy and in combination with standard of care therapeutics. Currently, no selective AXL-targeting therapy has reached FDA-approval, but several compounds have entered phase II and III studies. AREA COVERED We elaborate on the role of AXL in cancer progression and suppressing anti-cancer immunity at both the molecular level and immune cell interaction level. Additionally, we review pre-clinical and clinical data of AXL-targeting agents. EXPERT OPINION Preclinical and several early clinical trials demonstrated the safety of AXL-targeting monotherapies with some evidence of efficacy. Additionally, multiple novel combination regimens including AXL-targeting agents to overcome resistance mechanisms are being actively examined with some promising results. However, patient selection and companion biomarkers may be critical for the success of AXL-targeting therapies.
Collapse
Affiliation(s)
- Juhye Yim
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Chloe Hope
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Justus M Huelse
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Douglas K Graham
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, USA
| |
Collapse
|
2
|
Rosenstein I, Novakova L, Kvartsberg H, Nordin A, Rasch S, Rembeza E, Sandgren S, Malmeström C, Fruhwürth S, Axelsson M, Blennow K, Zetterberg H, Lycke J. Tyro3 and Gas6 are associated with white matter and myelin integrity in multiple sclerosis. J Neuroinflammation 2024; 21:320. [PMID: 39673059 PMCID: PMC11645787 DOI: 10.1186/s12974-024-03315-0] [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] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/29/2024] [Indexed: 12/15/2024] Open
Abstract
BACKGROUND The Gas6/TAM (Tyro3, Axl, and Mer) receptor system has been implicated in demyelination and delayed remyelination in experimental animal models, but data in humans are scarce. We aimed to investigate the role of Gas6/TAM in neurodegenerative processes in multiple sclerosis (MS). METHODS From a prospective 5-year follow-up study, soluble Gas6/TAM biomarkers were analyzed in cerebrospinal fluid (CSF) by enzyme-linked immunosorbent assay (ELISA) at baseline in patients with relapsing-remitting MS (RRMS) (n = 40), progressive MS (PMS) (n = 20), and healthy controls (HC) (n = 25). Brain volumes, including myelin content (MyC) and white matter (WM) were measured by synthetic magnetic resonance imaging at baseline, 12 months, and 60-month follow-up. Associations with brain volume changes were investigated in multivariable linear regression models. Gas6/TAM concentrations were also determined at 12 months follow-up in RRMS to assess treatment response. RESULTS Baseline concentrations of Tyro3, Axl, and Gas6 were significantly higher in PMS vs. RRMS and HC. Mer was higher in PMS vs. HC. Tyro3 and Gas6 were associated with reduced WM (β = 25.5, 95% confidence interval [CI] [6.11-44.96, p = 0.012; β = 11.4, 95% CI [0.42-22.4], p = 0.042, respectively) and MyC (β = 7.95, 95%CI [1.84-14.07], p = 0.012; β = 4.4, 95%CI [1.04-7.75], p = 0.012 respectively) at 60 months. Patients with evidence of remyelination at last follow-up had lower baseline soluble Tyro3 (p = 0.033) and Gas6 (p = 0.014). Except Mer, Gas6/TAM concentrations did not change with treatment in RRMS. DISCUSSION Our data indicate a potential role for the Gas6/TAM receptor system in neurodegenerative processes influencing demyelination and ineffective remyelination.
Collapse
Affiliation(s)
- Igal Rosenstein
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Blå Stråket 7, 413 45, Gothenburg, Sweden.
- Region Västra Götaland, Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Lenka Novakova
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Blå Stråket 7, 413 45, Gothenburg, Sweden
- Region Västra Götaland, Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Hlin Kvartsberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
| | - Anna Nordin
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Blå Stråket 7, 413 45, Gothenburg, Sweden
| | - Sofia Rasch
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Elzbieta Rembeza
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Sofia Sandgren
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Blå Stråket 7, 413 45, Gothenburg, Sweden
- Region Västra Götaland, Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Clas Malmeström
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Blå Stråket 7, 413 45, Gothenburg, Sweden
- Region Västra Götaland, Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Stefanie Fruhwürth
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
| | - Markus Axelsson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Blå Stråket 7, 413 45, Gothenburg, Sweden
- Region Västra Götaland, Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- Hong Kong Centre for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Jan Lycke
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Blå Stråket 7, 413 45, Gothenburg, Sweden
- Region Västra Götaland, Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
3
|
Mor G, Singh A, Yang J, Adzibolosu N, Cai S, Kauf E, Yang L, Li Q, Li H, Werner A, Parthasarathy S, Ding J, Fortier J, Rodriguez-Garcia M, Diao LH. Decoding Functional and Developmental Trajectories of Tissue-Resident Uterine Dendritic Cells Through Integrative Omics. RESEARCH SQUARE 2024:rs.3.rs-5424920. [PMID: 39606471 PMCID: PMC11601813 DOI: 10.21203/rs.3.rs-5424920/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Uterine dendritic cells (uDCs) are critical for endometrial function, yet their origin, molecular characteristics, and specific roles during the pre- and post-implantation periods in the human endometrium remain largely unknown. The complexity of the endometrial environment makes defining the contributions of uDCs subtypes challenging. We hypothesize that distinct uDC subsets carry out specialized functions, and that resident progenitor DCs generate these subtypes. Employing single-cell RNA sequencing on uterine tissues collected across different menstrual phases and during early pregnancy, we identify several uDCs subtypes, including resident progenitor DCs. CITE-seq was performed on endometrial single-cell suspensions to link surface protein expression with key genes identified by the RNAseq analysis. Our analysis revealed the developmental trajectory of the uDCs along with the distinct functional roles of each uDC subtype, including immune regulation, antigen presentation, and creating a conducive environment for embryo implantation. This study provides a comprehensive characterization of uDCs, serving as a foundational reference for future studies for better understanding female reproductive disorders such as infertility and pregnancy complications.
Collapse
Affiliation(s)
| | | | - Jing Yang
- National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, Xiamen 361102, China
| | | | - Songchen Cai
- Shenzhen Zhongshan Obstetrics & Gynecology Hospital
| | | | | | - Qiyuan Li
- National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Hanjie Li
- Shenzhen Institutes of Advanced Technology
| | | | | | | | | | | | | |
Collapse
|
4
|
Perez F, Iribarren ML, Olexen CM, Ruera CN, Errasti AE, Guzman L, Garbi L, Carrera Silva EA, Chirdo FG. Duodenal mucosa of untreated celiac disease patients has altered expression of the GAS6 and PROS1 and the negative regulator tyrosine kinase TAM receptors subfamily. Clin Immunol 2024; 263:110202. [PMID: 38575045 DOI: 10.1016/j.clim.2024.110202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/14/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024]
Abstract
Celiac disease (CD) is an immune-driven disease characterized by tissue damage in the small intestine of genetically-susceptible individuals. We evaluated here a crucial immune regulatory pathway involving TYRO3, AXL, and MERTK (TAM) receptors and their ligands PROS1 and GAS6 in duodenal biopsies of controls and CD patients. We found increased GAS6 expression associated with downregulation of PROS1 and variable TAM receptors levels in duodenum tissue of CD patients. Interestingly, CD3+ lymphocytes, CD68+, CD11c+ myeloid and epithelial cells, showed differential expressions of TAM components comparing CD vs controls. Principal component analysis revealed a clear segregation of two groups of CD patients based on TAM components and IFN signaling. In vitro validation demonstrated that monocytes, T lymphocytes and epithelial cells upregulated TAM components in response to IFN stimulation. Our findings highlight a dysregulated TAM axis in CD related to IFN signaling and contribute to a deeper understanding of the pathophysiology of CD.
Collapse
Affiliation(s)
- Federico Perez
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, CIC PBA, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - María Luz Iribarren
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, CIC PBA, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Cinthia Mariel Olexen
- Instituto de Medicina Experimental (IMEX), Academia Nacional de Medicina (ANM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) de Argentina, Buenos Aires, Argentina
| | - Carolina Naymé Ruera
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, CIC PBA, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Andrea Emilse Errasti
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Luciana Guzman
- Servicio de gastroenterología del Hospital de Niños Sor María Ludovica de La Plata, Argentina
| | - Laura Garbi
- Servicio de gastroenterología del Hospital San Martín de la Plata, Argentina
| | - Eugenio Antonio Carrera Silva
- Instituto de Medicina Experimental (IMEX), Academia Nacional de Medicina (ANM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) de Argentina, Buenos Aires, Argentina.
| | - Fernando Gabriel Chirdo
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, CIC PBA, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina.
| |
Collapse
|
5
|
Suzuki H, Iwamoto H, Tanaka T, Sakaue T, Imamura Y, Masuda A, Nakamura T, Koga H, Hoshida Y, Kawaguchi T. Fibroblast growth factor inhibition by molecular-targeted agents mitigates immunosuppressive tissue microenvironment in hepatocellular carcinoma. Hepatol Int 2024; 18:610-622. [PMID: 37864726 PMCID: PMC11014819 DOI: 10.1007/s12072-023-10603-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/24/2023] [Indexed: 10/23/2023]
Abstract
BACKGROUND & AIMS Combination immunotherapy refers to the use of immune checkpoint inhibitors (ICI) and molecular-targeted agents (MTA), which have recently been approved for the treatment of advanced hepatocellular carcinoma (HCC). Owing to its relatively low antitumor effect (up to 30%), sequential therapy following ICIs treatment is required in patients with HCC. This study aimed to determine the impact of MTAs on the tumor immune microenvironment (TIME). METHODS We established immune syngeneic orthotopic HCC mouse models using Hep-55.1C and Hep-53.4, and treated them with MTAs (lenvatinib, sorafenib, regorafenib, cabozantinib, and DC101 as anti-vascular endothelial growth factor receptor-2 antibodies, and AZD4547 as a fibroblast growth factor receptor (FGFR)-1/2/3/4 inhibitor) for 2 weeks. Subsequently, alterations in the TIME caused by MTAs were evaluated using immunohistochemistry (antibodies for CD3, CD8, Foxp3, Granzyme B, Arginase-1, NK1.1, F4/80, CD11c, PD-1, and PD-L1). We conducted RNA-seq analysis using lenvatinib- and AZD4547-treated tumors. To confirm the clinical relevance of these findings, we analyzed the transcriptome data of human HCC cells (MHCC-97H) treated with various concentrations of lenvatinib for 24 h using RNA-seq data from the Gene Expression Omnibus database. RESULTS The number of Foxp3- and F4/80-positive cells in the TIME was decreased in many MTAs. Cabozantinib increased the numbers in NK1.1-, Granzyme B, and CD11c-positive cells. Lenvatinib and AZD4547 increased the number of CD8, Granzyme B, and PD-L1-positive cells. Gene ontology enrichment analysis revealed that lipid metabolism-related genes were downregulated by lenvatinib and AZD4547. In total, 161 genes downregulated by FGFR inhibition in rodent models overlapped with those downregulated by lenvatinib in human HCC cells. CONCLUSIONS In this study, we showed that cabozantinib activated the innate immune system, and lenvatinib and AZD4547, which commonly inhibit FGFR signaling, altered TIME to a hot immune state by downregulating lipid metabolism-related genes. These findings support the therapeutic use of combination immunotherapies.
Collapse
Affiliation(s)
- Hiroyuki Suzuki
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan.
- Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, 830-0011, Japan.
| | - Hideki Iwamoto
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan.
- Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, 830-0011, Japan.
- Iwamoto Internal Medicine Clinic, Kitakyushu, 802-0832, Japan.
| | - Toshimitsu Tanaka
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
- Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, 830-0011, Japan
| | - Takahiko Sakaue
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
- Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, 830-0011, Japan
| | - Yasuko Imamura
- Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, 830-0011, Japan
| | - Atsutaka Masuda
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
- Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, 830-0011, Japan
| | - Toru Nakamura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
- Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, 830-0011, Japan
| | - Hironori Koga
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
- Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, 830-0011, Japan
| | - Yujin Hoshida
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| |
Collapse
|
6
|
Nerviani A, Boutet MA, Ghirardi GM, Goldmann K, Sciacca E, Rivellese F, Pontarini E, Prediletto E, Abatecola F, Caliste M, Pagani S, Mauro D, Bellan M, Cubuk C, Lau R, Church SE, Hudson BM, Humby F, Bombardieri M, Lewis MJ, Pitzalis C. Axl and MerTK regulate synovial inflammation and are modulated by IL-6 inhibition in rheumatoid arthritis. Nat Commun 2024; 15:2398. [PMID: 38493215 PMCID: PMC10944458 DOI: 10.1038/s41467-024-46564-6] [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] [Scholar Register] [Received: 03/08/2023] [Accepted: 02/27/2024] [Indexed: 03/18/2024] Open
Abstract
The TAM tyrosine kinases, Axl and MerTK, play an important role in rheumatoid arthritis (RA). Here, using a unique synovial tissue bioresource of patients with RA matched for disease stage and treatment exposure, we assessed how Axl and MerTK relate to synovial histopathology and disease activity, and their topographical expression and longitudinal modulation by targeted treatments. We show that in treatment-naive patients, high AXL levels are associated with pauci-immune histology and low disease activity and inversely correlate with the expression levels of pro-inflammatory genes. We define the location of Axl/MerTK in rheumatoid synovium using immunohistochemistry/fluorescence and digital spatial profiling and show that Axl is preferentially expressed in the lining layer. Moreover, its ectodomain, released in the synovial fluid, is associated with synovial histopathology. We also show that Toll-like-receptor 4-stimulated synovial fibroblasts from patients with RA modulate MerTK shedding by macrophages. Lastly, Axl/MerTK synovial expression is influenced by disease stage and therapeutic intervention, notably by IL-6 inhibition. These findings suggest that Axl/MerTK are a dynamic axis modulated by synovial cellular features, disease stage and treatment.
Collapse
Affiliation(s)
- Alessandra Nerviani
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London & NIHR BRC Barts Health NHS Trust, London, UK
| | - Marie-Astrid Boutet
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London & NIHR BRC Barts Health NHS Trust, London, UK
- Nantes Université, Oniris, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000, Nantes, France
| | - Giulia Maria Ghirardi
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London & NIHR BRC Barts Health NHS Trust, London, UK
| | - Katriona Goldmann
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London & NIHR BRC Barts Health NHS Trust, London, UK
| | - Elisabetta Sciacca
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London & NIHR BRC Barts Health NHS Trust, London, UK
| | - Felice Rivellese
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London & NIHR BRC Barts Health NHS Trust, London, UK
| | - Elena Pontarini
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London & NIHR BRC Barts Health NHS Trust, London, UK
| | - Edoardo Prediletto
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London & NIHR BRC Barts Health NHS Trust, London, UK
| | - Federico Abatecola
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London & NIHR BRC Barts Health NHS Trust, London, UK
| | - Mattia Caliste
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London & NIHR BRC Barts Health NHS Trust, London, UK
| | - Sara Pagani
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London & NIHR BRC Barts Health NHS Trust, London, UK
| | - Daniele Mauro
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London & NIHR BRC Barts Health NHS Trust, London, UK
| | - Mattia Bellan
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London & NIHR BRC Barts Health NHS Trust, London, UK
- Department of Rheumatology, University of Eastern Piedmont and Maggiore della Carita Hospital, Novara, Italy
| | - Cankut Cubuk
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London & NIHR BRC Barts Health NHS Trust, London, UK
| | - Rachel Lau
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London & NIHR BRC Barts Health NHS Trust, London, UK
| | | | | | - Frances Humby
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London & NIHR BRC Barts Health NHS Trust, London, UK
| | - Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London & NIHR BRC Barts Health NHS Trust, London, UK
| | - Myles J Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London & NIHR BRC Barts Health NHS Trust, London, UK
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London & NIHR BRC Barts Health NHS Trust, London, UK.
- Department of Biomedical Sciences, Humanitas University & IRCCS Humanitas Research Hospital, Milan, Italy.
| |
Collapse
|
7
|
Gao L, He C, Yang A, Zhou H, Lu Q, Birge RB, Wu Y. Receptor tyrosine kinases Tyro3, Axl, and Mertk differentially contribute to antibody-induced arthritis. Cell Commun Signal 2023; 21:195. [PMID: 37537628 PMCID: PMC10398921 DOI: 10.1186/s12964-023-01133-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 04/20/2023] [Indexed: 08/05/2023] Open
Abstract
Tyro3, Axl, and Mertk (abbreviated TAMs) comprise a family of homologous type 1 receptor tyrosine kinases (RTKs) that have been implicated as inhibitory receptors that dampen inflammation, but their roles in the pathogenesis of rheumatoid arthritis remains understudied. Here, to investigate TAMs in an inflammatory arthritis model, antibody-induced arthritis in single TAM-deficient mice (Tyro3- KO, Axl-KO, Mertk-KO) was induced by K/BxN serum injection. Subsequently, joint inflammation and cytokine levels, as well as the expression of Fcγ Rs and complement receptors were assessed in WT and TAM-deficient mice. Compared with littermate control mice, Axl-/- and Mertk-/- mice developed more severe antibody-induced arthritis, while in contrast, Tyro3-/- mice showed diminished joint inflammation. Concomitantly, the levels of cytokines in joints of Axl-/- and Mertk-/- mice were also significantly increased, while cytokines in the Tyro3-/- joint tissues were decreased. At the molecular and cellular level, TAMs showed distinct expression patterns, whereby monocytes expressed Axl and Mertk, but no Tyro3, while neutrophils expressed Axl and Tyro3 but little Mertk. Moreover, expression of Fcγ receptors and C5aR showed different patterns with TAMs expression, whereby FcγRIV was higher in monocytes of Axl-/- and Mertk-/- mice compared to wild-type mice, while Tyro3-/- neutrophils showed lower expression levels of FcγRI, FcγRIII and FcγRIV. Finally, expression of C5aR was increased in Mertk-/- monocytes, and was decreased in Tyro3-/- neutrophils. These data indicate that Axl, Mertk and Tyro3 have distinct functions in antibody-induced arthritis, due in part to the differential regulation of cytokines production, as well as expression of FcγRs and C5aR. Video Abstract.
Collapse
Affiliation(s)
- Liang Gao
- Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, National Clinical Research Center for Hematologic Diseases, Cyrus Tang Medical Institute, Soochow University, Suzhou, 215123, China
| | - Chao He
- Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, National Clinical Research Center for Hematologic Diseases, Cyrus Tang Medical Institute, Soochow University, Suzhou, 215123, China
| | - Aizhen Yang
- Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, National Clinical Research Center for Hematologic Diseases, Cyrus Tang Medical Institute, Soochow University, Suzhou, 215123, China.
| | - Haibin Zhou
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215123, China
| | - Qingxian Lu
- Department of Ophthalmology and Visual Sciences, University of Louisville, Louisville, KY, 40202, USA
| | - Raymond B Birge
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School Cancer Center, Rutgers University, Newark, NJ, USA.
| | - Yi Wu
- Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, National Clinical Research Center for Hematologic Diseases, Cyrus Tang Medical Institute, Soochow University, Suzhou, 215123, China.
- Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
8
|
Song Q, Datta S, Liang X, Xu X, Pavicic P, Zhang X, Zhao Y, Liu S, Zhao J, Xu Y, Xu J, Wu L, Wu Z, Zhang M, Zhao Z, Lin C, Wang Y, Han P, Jiang P, Qin Y, Li W, Zhang Y, Luo Y, Sen G, Stark GR, Zhao C, Hamilton T, Yang J. Type I interferon signaling facilitates resolution of acute liver injury by priming macrophage polarization. Cell Mol Immunol 2023; 20:143-157. [PMID: 36596875 PMCID: PMC9886918 DOI: 10.1038/s41423-022-00966-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 12/08/2022] [Indexed: 01/05/2023] Open
Abstract
Due to their broad functional plasticity, myeloid cells contribute to both liver injury and recovery during acetaminophen overdose-induced acute liver injury (APAP-ALI). A comprehensive understanding of cellular diversity and intercellular crosstalk is essential to elucidate the mechanisms and to develop therapeutic strategies for APAP-ALI treatment. Here, we identified the function of IFN-I in the myeloid compartment during APAP-ALI. Utilizing single-cell RNA sequencing, we characterized the cellular atlas and dynamic progression of liver CD11b+ cells post APAP-ALI in WT and STAT2 T403A mice, which was further validated by immunofluorescence staining, bulk RNA-seq, and functional experiments in vitro and in vivo. We identified IFN-I-dependent transcriptional programs in a three-way communication pathway that involved IFN-I synthesis in intermediate restorative macrophages, leading to CSF-1 production in aging neutrophils that ultimately enabled Trem2+ restorative macrophage maturation, contributing to efficient liver repair. Overall, we uncovered the heterogeneity of hepatic myeloid cells in APAP-ALI at single-cell resolution and the therapeutic potential of IFN-I in the treatment of APAP-ALI.
Collapse
Affiliation(s)
- Qiaoling Song
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Shyamasree Datta
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Xue Liang
- Lars Bolund Institute of Regenerative Medicine, Qingdao-Europe Advanced Institute for Life Sciences, BGI-Qingdao, BGI-Shenzhen, Shenzhen, China
| | - Xiaohan Xu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Paul Pavicic
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Xiaonan Zhang
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Yuanyuan Zhao
- Department of Radiation, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shan Liu
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jun Zhao
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Yuting Xu
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jing Xu
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Lihong Wu
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Zhihua Wu
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Minghui Zhang
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Zhan Zhao
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Chunhua Lin
- Department of Urology, Yantai Yuhuangding Hospital, Yantai, China
| | - Yuxin Wang
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Peng Han
- Lars Bolund Institute of Regenerative Medicine, Qingdao-Europe Advanced Institute for Life Sciences, BGI-Qingdao, BGI-Shenzhen, Shenzhen, China
| | - Peng Jiang
- Department of Radiation, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yating Qin
- Lars Bolund Institute of Regenerative Medicine, Qingdao-Europe Advanced Institute for Life Sciences, BGI-Qingdao, BGI-Shenzhen, Shenzhen, China
| | - Wei Li
- Department of Radiation, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yingying Zhang
- Lars Bolund Institute of Regenerative Medicine, Qingdao-Europe Advanced Institute for Life Sciences, BGI-Qingdao, BGI-Shenzhen, Shenzhen, China
| | - Yonglun Luo
- Lars Bolund Institute of Regenerative Medicine, Qingdao-Europe Advanced Institute for Life Sciences, BGI-Qingdao, BGI-Shenzhen, Shenzhen, China
| | - Ganes Sen
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - George R Stark
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Chenyang Zhao
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China.
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| | - Thomas Hamilton
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
| | - Jinbo Yang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China.
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
9
|
Zheng L, Xu L, Hu F, Xue J, Bai M, Yao R, Zhu H, Zhong H, Su Y. Elevated expression of TAM receptor tyrosine kinase in synovial fluid and synovial tissue of rheumatoid arthritis. Clin Exp Immunol 2022; 209:270-279. [PMID: 35951003 PMCID: PMC9521663 DOI: 10.1093/cei/uxac071] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/17/2022] [Accepted: 08/08/2022] [Indexed: 01/23/2023] Open
Abstract
To investigate the expression and roles of TAM (Tyro3/Axl/Mer) receptor tyrosine kinases (TK) in synovial fluid and synovial tissue of patients with rheumatoid arthritis (RA). The expression of TAM TKs in the synovial fluid and synovial tissues of RA and osteoarthritis (OA) patients was measured by ELISA and immunohistochemistry. The relationships between soluble TAM TKs (sTAM TKs) levels and the clinical features, laboratory parameters and disease activity were analyzed in RA. The concentrations of sTAM TK in the synovial fluids of RA patients were increased in comparison to those of OA patients. Compared with OA patients, the expression of membrane Tyro3 TK (mTyro3 TK) and mMer TK in RA patient synovial tissue were significantly increased, which may partly explain the possible mechanism of elevated levels of sTAM TK in RA patient synovial fluid. sAxl TK levels were decreased in RA patients under sulfasalazine treatment and elevated in patients under Iguratimod treatment. Furthermore, sTyro3 TK levels were positively correlated with erythrocyte sedimentation rate (ESR) and negatively correlated with white blood cells (WBCs), red blood cells (RBCs), and hemoglobin (HB) in RA patients. The levels of sMer TK were positively associated with disease duration and rheumatoid factor (RF) and negatively correlated with HB, complement 3 (C3), and C4. Taken together, TAM TKs might be involved in RA synovial tissue inflammation.
Collapse
Affiliation(s)
- Li Zheng
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Beijing, China
| | - Liling Xu
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Fanlei Hu
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Jimeng Xue
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Mingxin Bai
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Ranran Yao
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Huaqun Zhu
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Hua Zhong
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yin Su
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| |
Collapse
|
10
|
Li H, Liu Z, Liu L, Zhang H, Han C, Girard L, Park H, Zhang A, Dong C, Ye J, Rayford A, Peyton M, Li X, Avila K, Cao X, Hu S, Alam MM, Akbay EA, Solis LM, Behrens C, Hernandez-Ruiz S, Lu W, Wistuba I, Heymach JV, Chisamore M, Micklem D, Gabra H, Gausdal G, Lorens JB, Li B, Fu YX, Minna JD, Brekken RA. AXL targeting restores PD-1 blockade sensitivity of STK11/LKB1 mutant NSCLC through expansion of TCF1 + CD8 T cells. Cell Rep Med 2022; 3:100554. [PMID: 35492873 PMCID: PMC9040166 DOI: 10.1016/j.xcrm.2022.100554] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 10/22/2021] [Accepted: 02/08/2022] [Indexed: 12/14/2022]
Abstract
Mutations in STK11/LKB1 in non-small cell lung cancer (NSCLC) are associated with poor patient responses to immune checkpoint blockade (ICB), and introduction of a Stk11/Lkb1 (L) mutation into murine lung adenocarcinomas driven by mutant Kras and Trp53 loss (KP) resulted in an ICB refractory syngeneic KPL tumor. Mechanistically this occurred because KPL mutant NSCLCs lacked TCF1-expressing CD8 T cells, a phenotype recapitulated in human STK11/LKB1 mutant NSCLCs. Systemic inhibition of Axl results in increased type I interferon secretion from dendritic cells that expanded tumor-associated TCF1+PD-1+CD8 T cells, restoring therapeutic response to PD-1 ICB in KPL tumors. This was observed in syngeneic immunocompetent mouse models and in humanized mice bearing STK11/LKB1 mutant NSCLC human tumor xenografts. NSCLC-affected individuals with identified STK11/LKB1 mutations receiving bemcentinib and pembrolizumab demonstrated objective clinical response to combination therapy. We conclude that AXL is a critical targetable driver of immune suppression in STK11/LKB1 mutant NSCLC.
Collapse
Affiliation(s)
- Huiyu Li
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-8593, USA
- Cancer Biology Graduate Program, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zhida Liu
- Department of Pathology, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9072, USA
| | - Longchao Liu
- Department of Pathology, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9072, USA
| | - Hongyi Zhang
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chuanhui Han
- Department of Pathology, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9072, USA
| | - Luc Girard
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-8593, USA
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hyunsil Park
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-8593, USA
| | - Anli Zhang
- Department of Pathology, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9072, USA
| | - Chunbo Dong
- Department of Pathology, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9072, USA
| | - Jianfeng Ye
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Austin Rayford
- BerGenBio ASA, Bergen, Norway
- Department of Biomedicine, Centre for Cancer Biomarkers, Norwegian Centre of Excellence, University of Bergen, Bergen, Norway
| | - Michael Peyton
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-8593, USA
| | - Xiaoguang Li
- Department of Pathology, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9072, USA
| | - Kimberley Avila
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-8593, USA
| | - Xuezhi Cao
- Department of Pathology, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9072, USA
| | - Shuiqing Hu
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Md Maksudul Alam
- Department of Pathology, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9072, USA
| | - Esra A. Akbay
- Department of Pathology, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9072, USA
| | - Luisa M. Solis
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Carmen Behrens
- Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sharia Hernandez-Ruiz
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wei Lu
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ignacio Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - John V. Heymach
- Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | - James B. Lorens
- Department of Biomedicine, Centre for Cancer Biomarkers, Norwegian Centre of Excellence, University of Bergen, Bergen, Norway
| | - Bo Li
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yang-Xin Fu
- Department of Pathology, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9072, USA
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - John D. Minna
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-8593, USA
- Cancer Biology Graduate Program, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rolf A. Brekken
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-8593, USA
- Cancer Biology Graduate Program, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
11
|
Mollet I, Martins C, Ângelo-Dias M, Carvalho AS, Aloria K, Matthiesen R, Baptista MV, Borrego LM, Vieira HL. Pilot study in human healthy volunteers on the mechanisms underlying remote ischemic conditioning (RIC) – Targeting circulating immune cells and immune-related proteins. J Neuroimmunol 2022; 367:577847. [DOI: 10.1016/j.jneuroim.2022.577847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/03/2022] [Accepted: 03/15/2022] [Indexed: 11/29/2022]
|
12
|
Kasper M, Heming M, Schafflick D, Li X, Lautwein T, Meyer zu Horste M, Bauer D, Walscheid K, Wiendl H, Loser K, Heiligenhaus A, Meyer zu Hörste G. Intraocular dendritic cells characterize HLA-B27-associated acute anterior uveitis. eLife 2021; 10:e67396. [PMID: 34783307 PMCID: PMC8594918 DOI: 10.7554/elife.67396] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 09/21/2021] [Indexed: 12/30/2022] Open
Abstract
Uveitis describes a heterogeneous group of inflammatory eye diseases characterized by infiltration of leukocytes into the uveal tissues. Uveitis associated with the HLA haplotype B27 (HLA-B27) is a common subtype of uveitis and a prototypical ocular immune-mediated disease. Local immune mechanisms driving human uveitis are poorly characterized mainly due to the limited available biomaterial and subsequent technical limitations. Here, we provide the first high-resolution characterization of intraocular leukocytes in HLA-B27-positive (n = 4) and -negative (n = 2) anterior uveitis and an infectious endophthalmitis control (n = 1) by combining single-cell RNA-sequencing with flow cytometry and protein analysis. Ocular cell infiltrates consisted primarily of lymphocytes in both subtypes of uveitis and of myeloid cells in infectious endophthalmitis. HLA-B27-positive uveitis exclusively featured a plasmacytoid and classical dendritic cell (cDC) infiltrate. Moreover, cDCs were central in predicted local cell-cell communication. This suggests a unique pattern of ocular leukocyte infiltration in HLA-B27-positive uveitis with relevance to DCs.
Collapse
Affiliation(s)
- Maren Kasper
- Ophtha-Lab, Department of Ophthalmology, and Uveitis Centre at St. Franziskus HospitalMünsterGermany
| | - Michael Heming
- Department of Neurology with Institute of Translational Neurology, University Hospital MuensterMuensterGermany
| | - David Schafflick
- Department of Neurology with Institute of Translational Neurology, University Hospital MuensterMuensterGermany
| | - Xiaolin Li
- Department of Neurology with Institute of Translational Neurology, University Hospital MuensterMuensterGermany
| | - Tobias Lautwein
- Department of Neurology with Institute of Translational Neurology, University Hospital MuensterMuensterGermany
| | | | - Dirk Bauer
- Ophtha-Lab, Department of Ophthalmology, and Uveitis Centre at St. Franziskus HospitalMünsterGermany
| | - Karoline Walscheid
- Ophtha-Lab, Department of Ophthalmology, and Uveitis Centre at St. Franziskus HospitalMünsterGermany
- Department of Ophthalmology, University of Duisburg-EssenEssenGermany
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University Hospital MuensterMuensterGermany
| | - Karin Loser
- Department of Human Medicine, University of OldenburgOldenburgGermany
| | - Arnd Heiligenhaus
- Ophtha-Lab, Department of Ophthalmology, and Uveitis Centre at St. Franziskus HospitalMünsterGermany
- University of Duisburg-EssenEssenGermany
| | - Gerd Meyer zu Hörste
- Department of Neurology with Institute of Translational Neurology, University Hospital MuensterMuensterGermany
| |
Collapse
|
13
|
Abstract
Extensive interest in cancer immunotherapy is reported according to the clinical importance of CTLA-4 and (PD-1/PD-L1) [programmed death (PD) and programmed death-ligand (PD-L1)] in immune checkpoint therapies. AXL is a receptor tyrosine kinase expressed in different types of cancer and in relation to resistance against various anticancer therapeutics due to poor clinical prognosis. AXL and its ligand, i.e., growth arrest-specific 6 (GAS6) proteins, are expressed on many cancer cells, and the GAS6/AXL pathway is reported to promote cancer cell proliferation, survival, migration, invasion, angiogenesis, and immune evasion. AXL is an attractive and novel therapeutic target for impairing tumor progression from immune cell contracts in the tumor microenvironment. The GAS6/AXL pathway is also of interest immunologically because it targets fewer antitumor immune responses. In effect, several targeted therapies are selective and nonselective for AXL, which are in preclinical and clinical development in multiple cancer types. Therefore, this review focuses on the role of the GAS6/AXL signaling pathway in triggering the immunosuppressive tumor microenvironment as immune evasion. This includes regulating its composition and activating T-cell exclusion with the immune-suppressive activity of regulatory T cells, which is related to one of the hallmarks of cancer survival. Finally, this article discusses the GAS6/AXL signaling pathway in the context of several immune responses such as NK cell activation, apoptosis, and tumor-specific immunity, especially PD-1/PDL-1 signaling.
Collapse
Affiliation(s)
- Hye-Youn Son
- Department of Breast and Endocrine Surgery, Center for Medical Innovation, Seoul National University Hospital, Seoul, South Korea
| | - Hwan-Kyu Jeong
- School of Biosystems and Biomedical Sciences, Korea University, Seoul, South Korea
| |
Collapse
|
14
|
Yan D, Earp HS, DeRyckere D, Graham DK. Targeting MERTK and AXL in EGFR Mutant Non-Small Cell Lung Cancer. Cancers (Basel) 2021; 13:5639. [PMID: 34830794 PMCID: PMC8616094 DOI: 10.3390/cancers13225639] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 12/20/2022] Open
Abstract
MERTK and AXL are members of the TAM family of receptor tyrosine kinases and are abnormally expressed in 69% and 93% of non-small cell lung cancers (NSCLCs), respectively. Expression of MERTK and/or AXL provides a survival advantage for NSCLC cells and correlates with lymph node metastasis, drug resistance, and disease progression in patients with NSCLC. The TAM receptors on host tumor infiltrating cells also play important roles in the immunosuppressive tumor microenvironment. Thus, MERTK and AXL are attractive biologic targets for NSCLC treatment. Here, we will review physiologic and oncologic roles for MERTK and AXL with an emphasis on the potential to target these kinases in NSCLCs with activating EGFR mutations.
Collapse
Affiliation(s)
- Dan Yan
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, GA 30322, USA; (D.Y.); (D.D.)
| | - H. Shelton Earp
- UNC Lineberger Comprehensive Cancer Center, Department of Medicine, Chapel Hill, NC 27599, USA;
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Deborah DeRyckere
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, GA 30322, USA; (D.Y.); (D.D.)
| | - Douglas K. Graham
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, GA 30322, USA; (D.Y.); (D.D.)
| |
Collapse
|
15
|
Rui J, Deng S, Perdigoto AL, Ponath G, Kursawe R, Lawlor N, Sumida T, Levine-Ritterman M, Stitzel ML, Pitt D, Lu J, Herold KC. Tet2 Controls the Responses of β cells to Inflammation in Autoimmune Diabetes. Nat Commun 2021; 12:5074. [PMID: 34417463 PMCID: PMC8379260 DOI: 10.1038/s41467-021-25367-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/06/2021] [Indexed: 01/02/2023] Open
Abstract
β cells may participate and contribute to their own demise during Type 1 diabetes (T1D). Here we report a role of their expression of Tet2 in regulating immune killing. Tet2 is induced in murine and human β cells with inflammation but its expression is reduced in surviving β cells. Tet2-KO mice that receive WT bone marrow transplants develop insulitis but not diabetes and islet infiltrates do not eliminate β cells even though immune cells from the mice can transfer diabetes to NOD/scid recipients. Tet2-KO recipients are protected from transfer of disease by diabetogenic immune cells.Tet2-KO β cells show reduced expression of IFNγ-induced inflammatory genes that are needed to activate diabetogenic T cells. Here we show that Tet2 regulates pathologic interactions between β cells and immune cells and controls damaging inflammatory pathways. Our data suggests that eliminating TET2 in β cells may reduce activating pathologic immune cells and killing of β cells.
Collapse
Affiliation(s)
- Jinxiu Rui
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, CT, USA
| | - Songyan Deng
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, CT, USA
| | - Ana Luisa Perdigoto
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, CT, USA
| | - Gerald Ponath
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Romy Kursawe
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Nathan Lawlor
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Tomokazu Sumida
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | | | - Michael L Stitzel
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Department of Genetics and Genome Sciences and Institute for Systems Genomics, University of Connecticut, Farmington, CT, USA
| | - David Pitt
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Jun Lu
- Department of Genetics, Yale University, New Haven, CT, USA
| | - Kevan C Herold
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, CT, USA.
| |
Collapse
|
16
|
Zhou L, Matsushima GK. Tyro3, Axl, Mertk receptor-mediated efferocytosis and immune regulation in the tumor environment. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 361:165-210. [PMID: 34074493 DOI: 10.1016/bs.ircmb.2021.02.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Three structurally related tyrosine receptor cell surface kinases, Tyro3, Axl, and Mertk (TAM) have been recognized to modulate immune function, tissue homeostasis, cardiovasculature, and cancer. The TAM receptor family appears to operate in adult mammals across multiple cell types, suggesting both widespread and specific regulation of cell functions and immune niches. TAM family members regulate tissue homeostasis by monitoring the presence of phosphatidylserine expressed on stressed or apoptotic cells. The detection of phosphatidylserine on apoptotic cells requires intermediary molecules that opsonize the dying cells and tether them to TAM receptors on phagocytes. This complex promotes the engulfment of apoptotic cells, also known as efferocytosis, that leads to the resolution of inflammation and tissue healing. The immune mechanisms dictating these processes appear to fall upon specific family members or may involve a complex of different receptors acting cooperatively to resolve and repair damaged tissues. Here, we focus on the role of TAM receptors in triggering efferocytosis and its consequences in the regulation of immune responses in the context of inflammation and cancer.
Collapse
Affiliation(s)
- Liwen Zhou
- UNC Neuroscience Center, University of North Carolina-CH, Chapel Hill, NC, United States
| | - Glenn K Matsushima
- UNC Neuroscience Center, University of North Carolina-CH, Chapel Hill, NC, United States; UNC Department of Microbiology & Immunology, University of North Carolina-CH, Chapel Hill, NC, United States; UNC Integrative Program for Biological & Genome Sciences, University of North Carolina-CH, Chapel Hill, NC, United States.
| |
Collapse
|
17
|
Li M, Xue W, Li X, Song Y, Liu X, Qin L. Axl is related to inflammation in hemodialysis patients. Mol Immunol 2021; 133:146-153. [PMID: 33667984 DOI: 10.1016/j.molimm.2021.02.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 02/10/2021] [Accepted: 02/23/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Hemodialysis (HD) patients often develop chronic inflammation, which is associated with an increased risk of cardiovascular complications and mortality. Axl and its ligand, growth arrest 6 (Gas6), have been reported to play key roles in regulating the immune response. However, the function of Axl in HD patients has not been clarified. METHODS In the present study, we enrolled 130 HD patients and 117 normal controls (NCs) and evaluated the levels of inflammatory markers, soluble Axl (sAxl), membrane Axl (mAxl), and Gas6 in all participants. The potential downstream cascades of Gas6-Axl signaling in HD patients were identified by quantitative real time polymerase chain reaction and western blotting. RESULTS The levels of inflammatory cytokines-tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ)-plasma sAxl, and Gas6, were significantly increased in HD patients compared to NCs. Additionally, sAxl was positively associated with the inflammatory factor, interleukin-6 (IL-6), in HD patients. Moreover, we found that mAxl in CD14+ mononuclear cells and CD19+ B cells was increased upon HD. Furthermore, we discovered that the metalloproteinase ADAM17, also called TACE, contributed to the cleavage of mAxl into sAxl, and not ADAM10, in the peripheral blood mononuclear cells (PBMCs) of HD patients. The upregulation of Gas6-mAxl signaling caused the activation of the STAT1-SOCS3 pathway in the PBMCs of HD patients. After two years follow-up, patients with lower sAxl levels had longer survival time than those with higher sAxl levels. CONCLUSION Our results suggest that Axl may play a significant role in systemic inflammation in HD patients.
Collapse
Affiliation(s)
- Mengyuan Li
- Department of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301, Middle Yanchang Road, Shanghai, 200072, China
| | - Wen Xue
- Department of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301, Middle Yanchang Road, Shanghai, 200072, China
| | - Xinhua Li
- Department of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301, Middle Yanchang Road, Shanghai, 200072, China
| | - Yaxiang Song
- Department of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301, Middle Yanchang Road, Shanghai, 200072, China
| | - Xinying Liu
- Department of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301, Middle Yanchang Road, Shanghai, 200072, China.
| | - Ling Qin
- Department of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301, Middle Yanchang Road, Shanghai, 200072, China.
| |
Collapse
|
18
|
Jin S, Guerrero-Juarez CF, Zhang L, Chang I, Ramos R, Kuan CH, Myung P, Plikus MV, Nie Q. Inference and analysis of cell-cell communication using CellChat. Nat Commun 2021; 12:1088. [PMID: 33597522 PMCID: PMC7889871 DOI: 10.1038/s41467-021-21246-9] [Citation(s) in RCA: 3708] [Impact Index Per Article: 927.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 01/08/2021] [Indexed: 01/31/2023] Open
Abstract
Understanding global communications among cells requires accurate representation of cell-cell signaling links and effective systems-level analyses of those links. We construct a database of interactions among ligands, receptors and their cofactors that accurately represent known heteromeric molecular complexes. We then develop CellChat, a tool that is able to quantitatively infer and analyze intercellular communication networks from single-cell RNA-sequencing (scRNA-seq) data. CellChat predicts major signaling inputs and outputs for cells and how those cells and signals coordinate for functions using network analysis and pattern recognition approaches. Through manifold learning and quantitative contrasts, CellChat classifies signaling pathways and delineates conserved and context-specific pathways across different datasets. Applying CellChat to mouse and human skin datasets shows its ability to extract complex signaling patterns. Our versatile and easy-to-use toolkit CellChat and a web-based Explorer ( http://www.cellchat.org/ ) will help discover novel intercellular communications and build cell-cell communication atlases in diverse tissues.
Collapse
Affiliation(s)
- Suoqin Jin
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
| | - Christian F Guerrero-Juarez
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Lihua Zhang
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
| | - Ivan Chang
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
- Research Cyberinfrastructure Center, University of California, Irvine, Irvine, CA, USA
| | - Raul Ramos
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Chen-Hsiang Kuan
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Division of Plastic Surgery, Department of Surgery, National Taiwan University, Taipei, Taiwan
| | - Peggy Myung
- Department of Dermatology, Yale University, New Haven, CT, USA
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Maksim V Plikus
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA.
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA.
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA.
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA.
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA.
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
19
|
Kvedaraite E, Hertwig L, Sinha I, Ponzetta A, Hed Myrberg I, Lourda M, Dzidic M, Akber M, Klingström J, Folkesson E, Muvva JR, Chen P, Gredmark-Russ S, Brighenti S, Norrby-Teglund A, Eriksson LI, Rooyackers O, Aleman S, Strålin K, Ljunggren HG, Ginhoux F, Björkström NK, Henter JI, Svensson M. Major alterations in the mononuclear phagocyte landscape associated with COVID-19 severity. Proc Natl Acad Sci U S A 2021; 118:e2018587118. [PMID: 33479167 PMCID: PMC8017719 DOI: 10.1073/pnas.2018587118] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Dendritic cells (DCs) and monocytes are crucial mediators of innate and adaptive immune responses during viral infection, but misdirected responses by these cells may contribute to immunopathology. Here, we performed high-dimensional flow cytometry-analysis focusing on mononuclear phagocyte (MNP) lineages in SARS-CoV-2-infected patients with moderate and severe COVID-19. We provide a deep and comprehensive map of the MNP landscape in COVID-19. A redistribution of monocyte subsets toward intermediate monocytes and a general decrease in circulating DCs was observed in response to infection. Severe disease coincided with the appearance of monocytic myeloid-derived suppressor cell-like cells and a higher frequency of pre-DC2. Furthermore, phenotypic alterations in MNPs, and their late precursors, were cell-lineage-specific and associated either with the general response against SARS-CoV-2 or COVID-19 severity. This included an interferon-imprint in DC1s observed in all patients and a decreased expression of the coinhibitory molecule CD200R in pre-DCs, DC2s, and DC3 subsets of severely sick patients. Finally, unsupervised analysis revealed that the MNP profile, alone, pointed to a cluster of COVID-19 nonsurvivors. This study provides a reference for the MNP response to SARS-CoV-2 infection and unravels mononuclear phagocyte dysregulations associated with severe COVID-19.
Collapse
Affiliation(s)
- Egle Kvedaraite
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden;
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Laura Hertwig
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Indranil Sinha
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Andrea Ponzetta
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Ida Hed Myrberg
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Magda Lourda
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Majda Dzidic
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Mira Akber
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Jonas Klingström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Elin Folkesson
- Department of Infectious Diseases, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Jagadeeswara Rao Muvva
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Puran Chen
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Sara Gredmark-Russ
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Susanna Brighenti
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Anna Norrby-Teglund
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Lars I Eriksson
- Department of Physiology and Pharmacology, Section for Anesthesiology and Intensive Care, Karolinska Institutet, 171 77 Stockholm, Sweden
- Function Perioperative Medicine and Intensive Care, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Olav Rooyackers
- Function Perioperative Medicine and Intensive Care, Karolinska University Hospital, 171 77 Stockholm, Sweden
- Division of Anesthesiology and Intensive Care, Department of Clinical Science, Intervention, and Technology, Karolinska Institutet, 141 52 Huddinge, Sweden
| | - Soo Aleman
- Department of Infectious Diseases, Karolinska University Hospital, 171 77 Stockholm, Sweden
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Kristoffer Strålin
- Department of Infectious Diseases, Karolinska University Hospital, 171 77 Stockholm, Sweden
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Hans-Gustaf Ljunggren
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Florent Ginhoux
- Singapore Immunology Network, Agency for Science, Technology and Research, BIOPOLIS, 138648 Singapore, Singapore
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, 200240 Shanghai, China
- Translational Immunology Institute, SingHealth Duke-National University of Singapore Academic Medical Centre, 168753 Singapore, Singapore
| | - Niklas K Björkström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Jan-Inge Henter
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, 171 77 Stockholm, Sweden
- Pediatric Oncology, Theme of Children's Health, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Mattias Svensson
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden
| |
Collapse
|
20
|
Abstract
Over the last few years, cancer immunotherapy experienced tremendous developments and it is nowadays considered a promising strategy against many types of cancer. However, the exclusion of lymphocytes from the tumor nest is a common phenomenon that limits the efficiency of immunotherapy in solid tumors. Despite several mechanisms proposed during the years to explain the immune excluded phenotype, at present, there is no integrated understanding about the role played by different models of immune exclusion in human cancers. Hypoxia is a hallmark of most solid tumors and, being a multifaceted and complex condition, shapes in a unique way the tumor microenvironment, affecting gene transcription and chromatin remodeling. In this review, we speculate about an upstream role for hypoxia as a common biological determinant of immune exclusion in solid tumors. We also discuss the current state of ex vivo and in vivo imaging of hypoxic determinants in relation to T cell distribution that could mechanisms of immune exclusion and discover functional-morphological tumor features that could support clinical monitoring.
Collapse
|
21
|
Li XL, Zhang B, Liu W, Sun MJ, Zhang YL, Liu H, Wang MX. Rapamycin Alleviates the Symptoms of Multiple Sclerosis in Experimental Autoimmune Encephalomyelitis (EAE) Through Mediating the TAM-TLRs-SOCS Pathway. Front Neurol 2020; 11:590884. [PMID: 33329339 PMCID: PMC7728797 DOI: 10.3389/fneur.2020.590884] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/09/2020] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS). Our research aimed to find an immunomodulatory therapy for MS. An experimental autoimmune encephalomyelitis (EAE) mouse model of MS was established induced with the syntheticmyelin oligodendrocyte glycoprotein peptide 35-55 (MOG35-55). Fifty C57BL/6 mice were randomly divided into the Normal group, EAE group, and Rapamycin group (EAE mice treated with three different doses of rapamycin). Hematoxylin and eosin staining and Weil myelin staining were performed on the brain tissues of mice after 21 days post-immunization. The protein expression of Gas6, Tyro3, Axl, Mer in paraventricular tissues were analyzed by immunohistochemistry. The mRNA and protein expression of Gas6, Tyro3, Axl, Mer, SOCS1, SOCS3, Toll-like receptor (TLR) 3, and TLR4 were detected by quantitative real-time PCR (qRT-PCR) and Western blot, respectively. An enzyme-linked immunosorbent assay (ELISA) was used to detect the secretion of the inflammatory factors IFN-γ and IL-17. Rapamycin treatment could ameliorate the behavior impairment in EAE mice induced by MOG35-55. The expression of Gas6, Tyro3, Axl, Mer, SOCS1, and SOCS3 were decreased in EAE mice at 21 days post-immunization, while the expression of Gas6, Tyro3, Axl, and Mer in rapamycin group was higher than that in EAE group. It was accompanied by an increase in anti-inflammatory proteins SOCS1 and SOCS3, a decrease in the inflammatory proteins TLR-3, TLR-4 and in the amount of IFN-γ, and IL-17. Rapamycin injection relieved the nerve function of and the loss of myelin sheath in the EAE mice, mainly through mediating the TAM-TLRs-SOCS signaling pathway to regulate natural immunity.
Collapse
Affiliation(s)
- Xiao-ling Li
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Bo Zhang
- Department of Cardiology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Wei Liu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Meng-jiao Sun
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Ya-lan Zhang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Hui Liu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Man-xia Wang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
22
|
Di Stasi R, De Rosa L, D'Andrea LD. Therapeutic aspects of the Axl/Gas6 molecular system. Drug Discov Today 2020; 25:2130-2148. [PMID: 33002607 DOI: 10.1016/j.drudis.2020.09.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/14/2020] [Accepted: 09/21/2020] [Indexed: 12/17/2022]
Abstract
Axl receptor tyrosine kinase (RTK) and its ligand, growth arrest-specific protein 6 (Gas6), are involved in several biological functions and participate in the development and progression of a range of malignancies and autoimmune disorders. In this review, we present this molecular system from a drug discovery perspective, highlighting its therapeutic implications and challenges that need to be addressed. We provide an update on Axl/Gas6 axis biology, exploring its role in fields ranging from angiogenesis, cancer development and metastasis, immune response and inflammation to viral infection. Finally, we summarize the molecules that have been developed to date to target the Axl/Gas6 molecular system for therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Rossella Di Stasi
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| | - Lucia De Rosa
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| | - Luca D D'Andrea
- Istituto di Biostrutture e Bioimmagini, CNR, Via Nizza 52, 10126 Torino, Italy.
| |
Collapse
|
23
|
Giroud P, Renaudineau S, Gudefin L, Calcei A, Menguy T, Rozan C, Mizrahi J, Caux C, Duong V, Valladeau-Guilemond J. Expression of TAM-R in Human Immune Cells and Unique Regulatory Function of MerTK in IL-10 Production by Tolerogenic DC. Front Immunol 2020; 11:564133. [PMID: 33101282 PMCID: PMC7546251 DOI: 10.3389/fimmu.2020.564133] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 08/13/2020] [Indexed: 11/24/2022] Open
Abstract
Tumor-infiltrating myeloid cells are a key component of the immune infiltrate often correlated with a poor prognosis due to their capacities to sustain an immunosuppressive environment. Among membrane receptors implicated in myeloid cell functions, Tyro3, Axl, and MerTK, which are a family of tyrosine kinase receptors (TAM-R), have been described in the regulation of innate cell functions. Here, we have identified MerTK among TAM-R as the major marker of both human M2 macrophages and tolerogenic dendritic cells (DC). In situ, MerTK expression was found within the immune infiltrate in multiple solid tumors, highlighting its potential role in cancer immunity. TAM-R ligands Gas6 and PROS1 were found to be constitutively produced by myeloid cells in vitro. Importantly, we describe a novel function of MerTK/PROS1 axis in the regulation of IL-10 production by tolerogenic DC. Finally, the analysis of TAM-R expression within the lymphoid compartment following activation revealed that MerTK, but not Axl or Tyro3, is expressed on activated B lymphocytes and regulatory T cells, as well as CD4+ and CD8+ T cells. Thus, our findings deepen the implication of MerTK in the regulation of myeloid cell-mediated immunosuppression and identified new cellular targets expressing MerTK that could participate in the antitumor immune response.
Collapse
Affiliation(s)
- Paul Giroud
- Elsalys Biotech SA, Lyon, France.,Université Claude Bernard Lyon 1, INSERM U1052 CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | | | | | | | | | | | | | - Christophe Caux
- Université Claude Bernard Lyon 1, INSERM U1052 CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | | | - Jenny Valladeau-Guilemond
- Université Claude Bernard Lyon 1, INSERM U1052 CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| |
Collapse
|
24
|
Selective Increment of Synovial Soluble TYRO3 Correlates with Disease Severity and Joint Inflammation in Patients with Rheumatoid Arthritis. J Immunol Res 2020; 2020:9690832. [PMID: 32964059 PMCID: PMC7502136 DOI: 10.1155/2020/9690832] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/08/2020] [Accepted: 07/22/2020] [Indexed: 12/22/2022] Open
Abstract
Objective To investigate the role of TAM receptors in rheumatoid arthritis (RA) by determining synovial tissue TAM receptor expression, synovial fluid levels of soluble TAM receptors, and the relationship between soluble TAM receptors, joint inflammation and disease activity. Methods TAM receptor expression was determined by immunohistochemistry on the synovium from RA and osteoarthritis (OA) patients. Soluble (s) Tyro3, sAxl, sMer, and their ligand Gas6 were measured by ELISA in the synovial fluid of RA (n = 28) and OA (n = 12) patients and cytokine levels by multiplex immunoassay in RA samples. Correlation analyses were performed among sTAM receptors with local cytokine levels; systemic disease parameters like erythrocyte sedimentation rate (ESR), rheumatoid factor (RF), and anticyclic citrullinated peptide antibodies (ACPA); and disease activity scores (DAS28-ESR) in RA patients. Results TAM receptors were expressed on different locations in the synovial tissue (lining, sublining, and blood vessels), and a similar expression pattern was observed in RA and OA patients. Synovial fluid sTyro3 and sMer were significantly enhanced in RA compared to OA patients, whereas no significant differences in sAxl and Gas6 levels were found. In RA samples, sTyro3 levels, but not sMer, correlated positively with proinflammatory local cytokines and the systemic factor erythrocyte sedimentation rate. Moreover, stratification analysis showed high sTyro3 levels positively correlated with higher DAS28-ESR and in RF and ACPA double positive RA patients. Conclusion sTyro3 in the synovial fluid of RA patients correlates with local inflammatory molecules and systemic disease activity. These findings suggest that the reduced negative control of cell activation by TAM receptors due to their shedding in the synovial fluid, mainly sTyro3, favoring joint inflammation in RA patients.
Collapse
|
25
|
Gas6/Axl Signaling Pathway in the Tumor Immune Microenvironment. Cancers (Basel) 2020; 12:cancers12071850. [PMID: 32660000 PMCID: PMC7408754 DOI: 10.3390/cancers12071850] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 01/19/2023] Open
Abstract
Receptor tyrosine kinases have been shown to dysregulate a number of pathways associated with tumor development, progression, and metastasis. Axl is a receptor tyrosine kinase expressed in many cancer types and has been associated with therapy resistance and poor clinical prognosis and outcomes. In addition, Axl and its ligand growth arrest specific 6 (Gas6) protein are expressed by a number of host cells. The Gas6/Axl signaling pathway has been implicated in the promotion of tumor cell proliferation, survival, migration, invasion, angiogenesis, and immune evasion. As a result, Axl is an attractive, novel therapeutic target to impair multiple stages of tumor progression from both neoplastic and host cell axes. This review focuses on the role of the Gas6/Axl signaling pathway in promoting the immunosuppressive tumor microenvironment, as immune evasion is considered one of the hallmarks of cancer. The review discusses the structure and activation of the Gas6/Axl signaling pathway, GAS6 and AXL expression patterns in the tumor microenvironment, mechanisms of Axl-mediated tumor immune response, and the role of Gas6/Axl signaling in immune cell recruitment.
Collapse
|
26
|
Dayoub AS, Brekken RA. TIMs, TAMs, and PS- antibody targeting: implications for cancer immunotherapy. Cell Commun Signal 2020; 18:29. [PMID: 32087708 PMCID: PMC7036251 DOI: 10.1186/s12964-020-0521-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 01/29/2020] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy for cancer is making impressive strides at improving survival of a subset of cancer patients. To increase the breadth of patients that benefit from immunotherapy, new strategies that combat the immunosuppressive microenvironment of tumors are needed. Phosphatidylserine (PS) signaling is exploited by tumors to enhance tumor immune evasion and thus strategies to inhibit PS-mediated immune suppression have potential to increase the efficacy of immunotherapy. PS is a membrane lipid that flips to the outer surface of the cell membrane during apoptosis and/or cell stress. Externalized PS can drive efferocytosis or engage PS receptors (PSRs) to promote local immune suppression. In the tumor microenvironment (TME) PS-mediated immune suppression is often termed apoptotic mimicry. Monoclonal antibodies (mAbs) targeting PS or PSRs have been developed and are in preclinical and clinical testing. The TIM (T-cell/transmembrane, immunoglobulin, and mucin) and TAM (Tyro3, AXL, and MerTK) family of receptors are PSRs that have been shown to drive PS-mediated immune suppression in tumors. This review will highlight the development of mAbs targeting PS, TIM-3 and the TAM receptors. Video Abstract
Collapse
Affiliation(s)
- Adam S Dayoub
- Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX, 75390-8593, USA
| | - Rolf A Brekken
- Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX, 75390-8593, USA. .,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
27
|
New Insights into the Role of Tyro3, Axl, and Mer Receptors in Rheumatoid Arthritis. DISEASE MARKERS 2020; 2020:1614627. [PMID: 32051695 PMCID: PMC6995487 DOI: 10.1155/2020/1614627] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 12/01/2019] [Accepted: 01/04/2020] [Indexed: 12/14/2022]
Abstract
Rheumatoid Arthritis (RA) is the most common chronic inflammatory autoimmune disease involving joints. Among several pathogenic mechanisms, the impairment of homeostatic regulators of inflammation seems to be critically important to sustain persistent infiltration and activation of immune and stromal cells within the diseased synovium. Tyrosine kinase receptors Tyro3, Axl, and Mer are members of the TAM family. Upon binding their ligands Growth Arrest-Specific gene 6 (Gas6) and Protein S (ProS1), TAM receptors (TAMs) exert numerous and diverse biologic functions. Activated Axl and Mer, for instance, can negatively regulate the inflammatory cascade and mediate phagocytosis of apoptotic cells, contributing to prevent the development of autoimmunity. Thus, a role for TAMs has been hypothesized in RA. In this review, we will summarise unmet clinical needs in RA, depict the biology of TAMs and TAM ligands, focussing on their ability to regulate the immune system and inflammation cascade, and finally offer an overview of the state-of-the-art literature about the putative role of TAM axis in RA.
Collapse
|
28
|
Yan S, Vandewalle N, De Beule N, Faict S, Maes K, De Bruyne E, Menu E, Vanderkerken K, De Veirman K. AXL Receptor Tyrosine Kinase as a Therapeutic Target in Hematological Malignancies: Focus on Multiple Myeloma. Cancers (Basel) 2019; 11:E1727. [PMID: 31694201 PMCID: PMC6896070 DOI: 10.3390/cancers11111727] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/28/2019] [Accepted: 10/31/2019] [Indexed: 01/13/2023] Open
Abstract
AXL belongs to the TAM (TYRO3, AXL, and MERTK) receptor family, a unique subfamily of the receptor tyrosine kinases. Their common ligand is growth arrest-specific protein 6 (GAS6). The GAS6/TAM signaling pathway regulates many important cell processes and plays an essential role in immunity, hemostasis, and erythropoiesis. In cancer, AXL overexpression and activation has been associated with cell proliferation, chemotherapy resistance, tumor angiogenesis, invasion, and metastasis; and has been correlated with a poor prognosis. In hematological malignancies, the expression and function of AXL is highly diverse, not only between the different tumor types but also in the surrounding tumor microenvironment. Most research and clinical evidence has been provided for AXL inhibitors in acute myeloid leukemia. However, recent studies also revealed an important role of AXL in lymphoid leukemia, lymphoma, and multiple myeloma. In this review, we summarize the basic functions of AXL in various cell types and the role of AXL in different hematological cancers, with a focus on AXL in the dormancy of multiple myeloma. In addition, we provide an update on the most promising AXL inhibitors currently in preclinical/clinical evaluation and discuss future perspectives in this emerging field.
Collapse
Affiliation(s)
- Siyang Yan
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090 Brussel, Belgium; (S.Y.); (N.V.); (N.D.B.); (S.F.); (K.M.); (E.D.B.); (E.M.); (K.V.)
- Department of Hematology, Tianjin Medical University, Tianjin 300060, China
| | - Niels Vandewalle
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090 Brussel, Belgium; (S.Y.); (N.V.); (N.D.B.); (S.F.); (K.M.); (E.D.B.); (E.M.); (K.V.)
| | - Nathan De Beule
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090 Brussel, Belgium; (S.Y.); (N.V.); (N.D.B.); (S.F.); (K.M.); (E.D.B.); (E.M.); (K.V.)
| | - Sylvia Faict
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090 Brussel, Belgium; (S.Y.); (N.V.); (N.D.B.); (S.F.); (K.M.); (E.D.B.); (E.M.); (K.V.)
| | - Ken Maes
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090 Brussel, Belgium; (S.Y.); (N.V.); (N.D.B.); (S.F.); (K.M.); (E.D.B.); (E.M.); (K.V.)
| | - Elke De Bruyne
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090 Brussel, Belgium; (S.Y.); (N.V.); (N.D.B.); (S.F.); (K.M.); (E.D.B.); (E.M.); (K.V.)
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090 Brussel, Belgium; (S.Y.); (N.V.); (N.D.B.); (S.F.); (K.M.); (E.D.B.); (E.M.); (K.V.)
| | - Karin Vanderkerken
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090 Brussel, Belgium; (S.Y.); (N.V.); (N.D.B.); (S.F.); (K.M.); (E.D.B.); (E.M.); (K.V.)
| | - Kim De Veirman
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090 Brussel, Belgium; (S.Y.); (N.V.); (N.D.B.); (S.F.); (K.M.); (E.D.B.); (E.M.); (K.V.)
| |
Collapse
|
29
|
TAM Receptor Pathways at the Crossroads of Neuroinflammation and Neurodegeneration. DISEASE MARKERS 2019; 2019:2387614. [PMID: 31636733 PMCID: PMC6766163 DOI: 10.1155/2019/2387614] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 06/04/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023]
Abstract
Increasing evidence suggests that pathogenic mechanisms underlying neurodegeneration are strongly linked with neuroinflammatory responses. Tyro3, Axl, and Mertk (TAM receptors) constitute a subgroup of the receptor tyrosine kinase family, cell surface receptors which transmit signals from the extracellular space to the cytoplasm and nucleus. TAM receptors and the corresponding ligands, Growth Arrest Specific 6 and Protein S, are expressed in different tissues, including the nervous system, playing complex roles in tissue repair, inflammation and cell survival, proliferation, and migration. In the nervous system, TAM receptor signalling modulates neurogenesis and neuronal migration, synaptic plasticity, microglial activation, phagocytosis, myelination, and peripheral nerve repair, resulting in potential interest in neuroinflammatory and neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Multiple Sclerosis. In Alzheimer and Parkinson diseases, a role of TAM receptors in neuronal survival and pathological protein aggregate clearance has been suggested, while in Multiple Sclerosis TAM receptors are involved in myelination and demyelination processes. To better clarify roles and pathways involving TAM receptors may have important therapeutic implications, given the fine modulation of multiple molecular processes which could be reached. In this review, we summarise the roles of TAM receptors in the central nervous system, focusing on the regulation of immune responses and microglial activities and analysing in vitro and in vivo studies regarding TAM signalling involvement in neurodegeneration.
Collapse
|
30
|
Mechanism of gut microbiota and Axl/SOCS3 in experimental autoimmune encephalomyelitis. Biosci Rep 2019; 39:BSR20190228. [PMID: 31221818 PMCID: PMC6603274 DOI: 10.1042/bsr20190228] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 05/08/2019] [Accepted: 05/24/2019] [Indexed: 01/09/2023] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated disease of the central nervous system (CNS). The present study explored the role of intestinal microbiota in the initiation and propagation of mice induced by experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. 48 C57BL/6 were randomly divided into control group and EAE group. The changes of body weight and the scores of neurological function were recorded. The mRNA expression of the receptor tyrosine kinase subfamily (AXL) was detected by real-time quantitative PCR. The levels of IL-17 and IFN-γ in blood samples were examined by ELISA. The intestinal microbial composition of mice at different time points during the EAE induction was analyzed by 16S rRNA gene-based sequencing. In EAE group, the body weight began to reduce at day 3 and neurological symptoms began to appear at day 7 after EAE induction. The levels of IL-17 and IFN-γ in EAE group reached the peak at day 21 and then decreased gradually. However, the expression of Axl and SOCS3 reached the lowest level at day 21 and then increased gradually. The microbiome analyses revealed that the abundances of Alistipes, Blautia, and Lachnospiraceae_NK4A136_group were significantly changed at day 14, whereas the abundances of Allobaculum, Eubacterium and Helicobacter were significantly changed at day 30 of EAE induction. The prevotellaceae_NK3B31_group may be key bacteria that contribute to the development of MS. Regulation of intestinal microbiota composition can become a new therapeutic target for the treatment of MS.
Collapse
|
31
|
Um E, Oh JM, Park J, Song T, Kim TE, Choi Y, Shin C, Kolygina D, Jeon JH, Grzybowski BA, Cho YK. Immature dendritic cells navigate microscopic mazes to find tumor cells. LAB ON A CHIP 2019; 19:1665-1675. [PMID: 30931468 DOI: 10.1039/c9lc00150f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Dendritic cells (DCs) are potent antigen-presenting cells with high sentinel ability to scan their neighborhood and to initiate an adaptive immune response. Whereas chemotactic migration of mature DCs (mDCs) towards lymph nodes is relatively well documented, the migratory behavior of immature DCs (imDCs) in tumor microenvironments is still poorly understood. Here, microfluidic systems of various geometries, including mazes, are used to investigate how the physical and chemical microenvironment influences the migration pattern of imDCs. Under proper degree of confinement, the imDCs are preferentially recruited towards cancer vs. normal cells, accompanied by increased cell speed and persistence. Furthermore, a systematic screen of cytokines, reveals that Gas6 is a major chemokine responsible for the chemotactic preference. These results and the accompanying theoretical model suggest that imDC migration in complex tissue environments is tuned by a proper balance between the strength of the chemical gradients and the degree of spatial confinement.
Collapse
Affiliation(s)
- Eujin Um
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
PD-1 immunobiology in systemic lupus erythematosus. J Autoimmun 2018; 97:1-9. [PMID: 30396745 DOI: 10.1016/j.jaut.2018.10.025] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/22/2018] [Accepted: 10/28/2018] [Indexed: 01/22/2023]
Abstract
Programmed death (PD)-1 receptors and their ligands have been identified in the pathogenesis and development of systemic lupus erythematosus (SLE). Two key pathways, toll-like receptor and type I interferon, are significant to SLE pathogenesis and modulate the expression of PD-1 and the ligands (PD-L1, PD-L2) through activation of NF-κB and/or STAT1. These cell signals are regulated by tyrosine kinase (Tyro, Axl, Mer) receptors (TAMs) that are aberrantly activated in SLE. STAT1 and NF-κB also exhibit crosstalk with the aryl hydrocarbon receptor (AHR). Ligands to AHR are identified in SLE etiology and pathogenesis. These ligands also regulate the activity of the Epstein-Barr virus (EBV), which is an identified factor in SLE and PD-1 immunobiology. AHR is important in the maintenance of immune tolerance and the development of distinct immune subsets, highlighting a potential role of AHR in PD-1 immunobiology. Understanding the functions of AHR ligands as well as AHR crosstalk with STAT1, NF-κB, and EBV may provide insight into disease development, the PD-1 axis and immunotherapies that target PD-1 and its ligand, PD-L1.
Collapse
|
33
|
Du W, Brekken RA. Does Axl have potential as a therapeutic target in pancreatic cancer? Expert Opin Ther Targets 2018; 22:955-966. [PMID: 30244621 PMCID: PMC6292430 DOI: 10.1080/14728222.2018.1527315] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Pancreatic cancer is a leading cause of cancer-related death. Metastasis, therapy resistance, and immunosuppression are dominant characteristics of pancreatic tumors. Strategies that enhance the efficacy of standard of care and/or immune therapy are likely the most efficient route to improve overall survival in this disease. Areas covered: Axl, a member of the TAM (Tyro3, Axl, MerTK) family of receptor tyrosine kinases, is involved in cell plasticity, chemoresistance, immune suppression, and metastasis in various cancers, including pancreatic cancer. This review provides an overview of Axl and its function in normal conditions, summarizes the regulation and function of Axl in cancer, and highlights the contribution of Axl to pancreatic cancer as well as its potential as a therapeutic target. Expert opinion: Axl is an attractive therapeutic target in pancreatic cancer because it contributes to many of the roadblocks that hamper therapeutic efficacy. Clinical evidence supporting Axl inhibition in pancreatic cancer is currently limited; however, multiple clinical trials have been initiated or are in the planning phase to test the effect of inhibiting Axl in conjunction with standard therapy in pancreatic cancer patients. We anticipate that these studies will provide robust validation of Axl as a therapeutic target in pancreatic cancer.
Collapse
|
34
|
The Dual Role of TAM Receptors in Autoimmune Diseases and Cancer: An Overview. Cells 2018; 7:cells7100166. [PMID: 30322068 PMCID: PMC6210017 DOI: 10.3390/cells7100166] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/05/2018] [Accepted: 10/09/2018] [Indexed: 01/01/2023] Open
Abstract
Receptor tyrosine kinases (RTKs) regulate cellular processes by converting signals from the extracellular environment to the cytoplasm and nucleus. Tyro3, Axl, and Mer (TAM) receptors form an RTK family that plays an intricate role in tissue maintenance, phagocytosis, and inflammation as well as cell proliferation, survival, migration, and development. Defects in TAM signaling are associated with numerous autoimmune diseases and different types of cancers. Here, we review the structure of TAM receptors, their ligands, and their biological functions. We discuss the role of TAM receptors and soluble circulating TAM receptors in the autoimmune diseases systemic lupus erythematosus (SLE) and multiple sclerosis (MS). Lastly, we discuss the effect of TAM receptor deregulation in cancer and explore the therapeutic potential of TAM receptors in the treatment of diseases.
Collapse
|
35
|
Xu L, Hu F, Zhu H, Liu X, Shi L, Li Y, Zhong H, Su Y. Soluble TAM receptor tyrosine kinases in rheumatoid arthritis: correlation with disease activity and bone destruction. Clin Exp Immunol 2017; 192:95-103. [PMID: 29148078 DOI: 10.1111/cei.13082] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2017] [Indexed: 12/28/2022] Open
Abstract
The TAM receptor tyrosine kinases (TAM RTK) are a subfamily of receptor tyrosine kinases, the role of which in autoimmune diseases such as systemic lupus erythematosus has been well explored, while their functions in rheumatoid arthritis (RA) remain largely unknown. In this study, we investigated the role of soluble TAM receptor tyrosine kinases (sAxl/sMer/sTyro3) in patients with RA. A total of 306 RA patients, 100 osteoarthritis (OA) patients and 120 healthy controls (HCs) were enrolled into this study. The serum concentrations of sAxl/sMer/sTyro3 were measured by enzyme-linked immunosorbent assay (ELISA), then the associations between sAxl/sMer/sTyro3 levels and clinical features of RA patients were analysed. We also investigated whether sTyro3 could promote osteoclast differentiation in vitro in RA patients. The results showed that compared with healthy controls (HCs), sTyro3 levels in the serum of RA patients were elevated remarkably and sMer levels were decreased significantly, whereas there was no difference between HCs and RA patients on sAxl levels. The sTyro3 levels were correlated weakly but positively with white blood cells (WBC), immunoglobulin (Ig)M, rheumatoid factor (RF), swollen joint counts, tender joint counts, total sharp scores and joint erosion scores. Conversely, there were no significant correlations between sMer levels and the above indices. Moreover, RA patients with high disease activity also showed higher sTyro3 levels. In-vitro osteoclast differentiation assay showed further that tartrate-resistant acid phosphatase (TRAP)+ osteoclasts were increased significantly in the presence of sTyro3. Collectively, our study indicated that serum sTyro3 levels were elevated in RA patients and correlated positively with disease activity and bone destruction, which may serve as an important participant in RA pathogenesis.
Collapse
Affiliation(s)
- L Xu
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - F Hu
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - H Zhu
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - X Liu
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - L Shi
- Department of Rheumatology and Immunology, Peking University International Hospital, Beijing, China
| | - Y Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - H Zhong
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Y Su
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| |
Collapse
|
36
|
Trahtemberg U, Mevorach D. Apoptotic Cells Induced Signaling for Immune Homeostasis in Macrophages and Dendritic Cells. Front Immunol 2017; 8:1356. [PMID: 29118755 PMCID: PMC5661053 DOI: 10.3389/fimmu.2017.01356] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 10/03/2017] [Indexed: 12/24/2022] Open
Abstract
Inefficient and abnormal clearance of apoptotic cells (efferocytosis) contributes to systemic autoimmune disease in humans and mice, and inefficient chromosomal DNA degradation by DNAse II leads to systemic polyarthritis and a cytokine storm. By contrast, efficient clearance allows immune homeostasis, generally leads to a non-inflammatory state for both macrophages and dendritic cells (DCs), and contributes to maintenance of peripheral tolerance. As many as 3 × 108 cells undergo apoptosis every hour in our bodies, and one of the primary “eat me” signals expressed by apoptotic cells is phosphatidylserine (PtdSer). Apoptotic cells themselves are major contributors to the “anti-inflammatory” nature of the engulfment process, some by secreting thrombospondin-1 (TSP-1) or adenosine monophosphate and possibly other immune modulating “calm-down” signals that interact with macrophages and DCs. Apoptotic cells also produce “find me” and “tolerate me” signals to attract and immune modulate macrophages and DCs that express specific receptors for some of these signals. Neither macrophages nor DCs are uniform, and each cell type may variably express membrane proteins that function as receptors for PtdSer or for opsonins like complement or opsonins that bind to PtdSer, such as protein S and growth arrest-specific 6. Macrophages and DCs also express scavenger receptors, CD36, and integrins that function via bridging molecules such as TSP-1 or milk fat globule-EGF factor 8 protein and that differentially engage in various multi-ligand interactions between apoptotic cells and phagocytes. In this review, we describe the anti-inflammatory and pro-homeostatic nature of apoptotic cell interaction with the immune system. We do not review some forms of immunogenic cell death. We summarize the known apoptotic cell signaling events in macrophages and DCs that are related to toll-like receptors, nuclear factor kappa B, inflammasome, the lipid-activated nuclear receptors, Tyro3, Axl, and Mertk receptors, as well as induction of signal transducer and activator of transcription 1 and suppressor of cytokine signaling that lead to immune system silencing and DC tolerance. These properties of apoptotic cells are the mechanisms that enable their successful use as therapeutic modalities in mice and humans in various autoimmune diseases, organ transplantation, graft-versus-host disease, and sepsis.
Collapse
Affiliation(s)
- Uriel Trahtemberg
- General Intensive Care Unit, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Dror Mevorach
- Rheumatology Research Center, Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
37
|
Tong LS, Shao AW, Ou YB, Guo ZN, Manaenko A, Dixon BJ, Tang J, Lou M, Zhang JH. Recombinant Gas6 augments Axl and facilitates immune restoration in an intracerebral hemorrhage mouse model. J Cereb Blood Flow Metab 2017; 37:1971-1981. [PMID: 27389179 PMCID: PMC5464693 DOI: 10.1177/0271678x16658490] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Axl, a tyrosine kinase receptor, was recently identified as an essential component regulating innate immune response. Suppressor of cytokine signaling 1 and suppressor of cytokine signaling 3 are potent Axl-inducible negative inflammatory regulators. This study investigated the role of Axl signaling pathway in immune restoration in an autologous blood-injection mouse model of intracerebral hemorrhage. Recombinant growth arrest-specific 6 (Gas6) and R428 were administrated as specific agonist and antagonist. In vivo knockdown of Axl or suppressor of cytokine signaling 1 and suppressor of cytokine signaling 3 by siRNA was applied. After intracerebral hemorrhage, the expression of endogenous Axl, soluble Axl, and Gas6 was increased, whereas the expression of suppressor of cytokine signaling 1 and suppressor of cytokine signaling 3 was inhibited. Recombinant growth arrest-specific 6 administration alleviated brain edema and improved neurobehavioral performances. Moreover, enhanced Axl phosphorylation with cleavage of soluble Axl (sAxl), and an upregulation of suppressor of cytokine signaling 1 and suppressor of cytokine signaling 3 were observed. In vivo knockdown of Axl and R428 administration both abolished the effect of recombinant growth arrest-specific 6 on brain edema and also decreased the expression suppressor of cytokine signaling 1 and suppressor of cytokine signaling 3. In vivo knockdown of suppressor of cytokine signaling 1 and suppressor of cytokine signaling 3 aggravated cytokine releasing despite of recombinant growth arrest-specific 6. In conclusion, Axl plays essential role in immune restoration after intracerebral hemorrhage. And recombinant growth arrest-specific 6 attenuated brain injury after intracerebral hemorrhage, probably by enhancing Axl phosphorylation and production of suppressor of cytokine signaling 1 and suppressor of cytokine signaling 3.
Collapse
Affiliation(s)
- Lu-Sha Tong
- 1 Department of Anesthesiology, School of Medicine, Loma Linda University, CA, USA.,2 Department of Neurology, School of Medicine, the 2nd Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - An-Wen Shao
- 1 Department of Anesthesiology, School of Medicine, Loma Linda University, CA, USA.,3 Department of Neurosurgery, School of Medicine, the 2nd Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Yi-Bo Ou
- 1 Department of Anesthesiology, School of Medicine, Loma Linda University, CA, USA.,4 Department of Neurosurgery, Tong-ji Hospital, Wuhan, China
| | - Zhen-Ni Guo
- 1 Department of Anesthesiology, School of Medicine, Loma Linda University, CA, USA.,5 Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Anatol Manaenko
- 1 Department of Anesthesiology, School of Medicine, Loma Linda University, CA, USA
| | - Brandon J Dixon
- 1 Department of Anesthesiology, School of Medicine, Loma Linda University, CA, USA
| | - Jiping Tang
- 2 Department of Neurology, School of Medicine, the 2nd Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Min Lou
- 2 Department of Neurology, School of Medicine, the 2nd Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - John H Zhang
- 1 Department of Anesthesiology, School of Medicine, Loma Linda University, CA, USA
| |
Collapse
|
38
|
Hastings AK, Yockey LJ, Jagger BW, Hwang J, Uraki R, Gaitsch HF, Parnell LA, Cao B, Mysorekar IU, Rothlin CV, Fikrig E, Diamond MS, Iwasaki A. TAM Receptors Are Not Required for Zika Virus Infection in Mice. Cell Rep 2017; 19:558-568. [PMID: 28423319 DOI: 10.1016/j.celrep.2017.03.058] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 03/16/2017] [Accepted: 03/20/2017] [Indexed: 01/26/2023] Open
Abstract
Tyro3, Axl, and Mertk (TAM) receptors are candidate entry receptors for infection with the Zika virus (ZIKV), an emerging flavivirus of global public health concern. To investigate the requirement of TAM receptors for ZIKV infection, we used several routes of viral inoculation and compared viral replication in wild-type versus Axl-/-, Mertk-/-, Axl-/-Mertk-/-, and Axl-/-Tyro3-/- mice in various organs. Pregnant and non-pregnant mice treated with interferon-α-receptor (IFNAR)-blocking (MAR1-5A3) antibody and infected subcutaneously with ZIKV showed no reliance on TAMs for infection. In the absence of IFNAR-blocking antibody, adult female mice challenged intravaginally with ZIKV showed no difference in mucosal viral titers. Similarly, in young mice that were infected with ZIKV intracranially or intraperitoneally, ZIKV replication occurred in the absence of TAM receptors, and no differences in cell tropism were observed. These findings indicate that, in mice, TAM receptors are not required for ZIKV entry and infection.
Collapse
Affiliation(s)
- Andrew K Hastings
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Laura J Yockey
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Brett W Jagger
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Jesse Hwang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ryuta Uraki
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Hallie F Gaitsch
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Lindsay A Parnell
- Department of Obstetrics and Gynecology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Bin Cao
- Department of Obstetrics and Gynecology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Indira U Mysorekar
- Department of Obstetrics and Gynecology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Carla V Rothlin
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - Michael S Diamond
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Obstetrics and Gynecology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
39
|
Laurance S, Bertin FR, Ebrahimian T, Kassim Y, Rys RN, Lehoux S, Lemarié CA, Blostein MD. Gas6 Promotes Inflammatory (CCR2 hiCX3CR1 lo) Monocyte Recruitment in Venous Thrombosis. Arterioscler Thromb Vasc Biol 2017; 37:1315-1322. [PMID: 28450294 DOI: 10.1161/atvbaha.116.308925] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 04/17/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Coagulation and inflammation are inter-related. Gas6 (growth arrest-specific 6) promotes venous thrombosis and participates to inflammation through endothelial-innate immune cell interactions. Innate immune cells can provide the initiating stimulus for venous thrombus development. We hypothesize that Gas6 promotes monocyte recruitment during venous thrombosis. APPROACH AND RESULTS Deep venous thrombosis was induced in wild-type and Gas6-deficient (-/-) mice using 5% FeCl3 and flow reduction in the inferior vena cava. Total monocyte depletion was achieved by injection of clodronate before deep venous thrombosis. Inflammatory monocytes were depleted using an anti-C-C chemokine receptor type 2 (CCR2) antibody. Similarly, injection of an anti-chemokine ligand 2 (CCL2) antibody induced CCL2 depletion. Flow cytometry and immunofluorescence were used to characterize the monocytes recruited to the thrombus. In vivo, absence of Gas6 was associated with a reduction of monocyte recruitment in both deep venous thrombosis models. Global monocyte depletion by clodronate leads to smaller thrombi in wild-type mice. Compared with wild type, the thrombi from Gas6-/- mice contain less inflammatory (CCR2hiCX3CR1lo) monocytes, consistent with a Gas6-dependent recruitment of this monocyte subset. Correspondingly, selective depletion of CCR2hiCX3CR1lo monocytes reduced the formation of venous thrombi in wild-type mice demonstrating a predominant role of the inflammatory monocytes in thrombosis. In vitro, the expression of both CCR2 and CCL2 were Gas6 dependent in monocytes and endothelial cells, respectively, impacting monocyte migration. Moreover, Gas6-dependent CCL2 expression and monocyte migration were mediated via JNK (c-Jun N-terminal kinase). CONCLUSIONS This study demonstrates that Gas6 specifically promotes the recruitment of inflammatory CCR2hiCX3CR1lo monocytes through the regulation of both CCR2 and CCL2 during deep venous thrombosis.
Collapse
MESH Headings
- Animals
- CX3C Chemokine Receptor 1
- Cells, Cultured
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Chemotaxis, Leukocyte/drug effects
- Clodronic Acid/pharmacology
- Disease Models, Animal
- Endothelial Cells/metabolism
- Genetic Predisposition to Disease
- Inflammation/genetics
- Inflammation/metabolism
- Inflammation/pathology
- Inflammation/prevention & control
- Intercellular Signaling Peptides and Proteins/deficiency
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/metabolism
- JNK Mitogen-Activated Protein Kinases/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Monocytes/drug effects
- Monocytes/metabolism
- Paracrine Communication
- Phenotype
- Receptors, CCR2/genetics
- Receptors, CCR2/metabolism
- Receptors, Chemokine/metabolism
- Signal Transduction
- Vena Cava, Inferior/drug effects
- Vena Cava, Inferior/metabolism
- Vena Cava, Inferior/pathology
- Venous Thrombosis/genetics
- Venous Thrombosis/metabolism
- Venous Thrombosis/pathology
- Venous Thrombosis/prevention & control
Collapse
Affiliation(s)
- Sandrine Laurance
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| | - François-René Bertin
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| | - Talin Ebrahimian
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| | - Yusra Kassim
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| | - Ryan N Rys
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| | - Stéphanie Lehoux
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| | - Catherine A Lemarié
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada.
| | - Mark D Blostein
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| |
Collapse
|
40
|
Growth Arrest-Specific 6 Enhances the Suppressive Function of CD4 +CD25 + Regulatory T Cells Mainly through Axl Receptor. Mediators Inflamm 2017; 2017:6848430. [PMID: 28270700 PMCID: PMC5320320 DOI: 10.1155/2017/6848430] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/07/2016] [Accepted: 12/05/2016] [Indexed: 11/17/2022] Open
Abstract
Background. Growth arrest-specific (Gas) 6 is one of the endogenous ligands of TAM receptors (Tyro3, Axl, and Mertk), and its role as an immune modulator has been recently emphasized. Naturally occurring CD4+CD25+ regulatory T cells (Tregs) are essential for the active suppression of autoimmunity. The present study was designed to investigate whether Tregs express TAM receptors and the potential role of Gas6-TAM signal in regulating the suppressive function of Tregs. Methods. The protein and mRNA levels of TAM receptors were determined by using Western blot, immunofluorescence, flow cytometry, and RT-PCR. Then, TAM receptors were silenced using targeted siRNA or blocked with specific antibody. The suppressive function of Tregs was assessed by using a CFSE-based T cell proliferation assay. Flow cytometry was used to determine the expression of Foxp3 and CTLA4 whereas cytokines secretion levels were measured by ELISA assay. Results. Tregs express both Axl and Mertk receptors. Gas6 increases the suppressive function of Tregs in vitro and in mice. Both Foxp3 and CTLA-4 expression on Tregs are enhanced after Gas6 stimulation. Gas6 enhances the suppressive activity of Tregs mainly through Axl receptor. Conclusion. Gas6 has a direct effect on the functions of CD4+CD25+Tregs mainly through its interaction with Axl receptor.
Collapse
|
41
|
The Gas6/TAM System and Multiple Sclerosis. Int J Mol Sci 2016; 17:ijms17111807. [PMID: 27801848 PMCID: PMC5133808 DOI: 10.3390/ijms17111807] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 10/22/2016] [Accepted: 10/26/2016] [Indexed: 01/25/2023] Open
Abstract
Growth arrest specific 6 (Gas6) is a multimodular circulating protein, the biological actions of which are mediated by the interaction with three transmembrane tyrosine kinase receptors: Tyro3, Axl, and MerTK, collectively named TAM. Over the last few decades, many progresses have been done in the understanding of the biological activities of this highly pleiotropic system, which plays a role in the regulation of immune response, inflammation, coagulation, cell growth, and clearance of apoptotic bodies. Recent findings have further related Gas6 and TAM receptors to neuroinflammation in general and, specifically, to multiple sclerosis (MS). In this paper, we review the biology of the Gas6/TAM system and the current evidence supporting its potential role in the pathogenesis of MS.
Collapse
|
42
|
Stalder G, Que YA, Calzavarini S, Burnier L, Kosinski C, Ballabeni P, Roger T, Calandra T, Duchosal MA, Liaudet L, Eggimann P, Angelillo-Scherrer A. Study of Early Elevated Gas6 Plasma Level as a Predictor of Mortality in a Prospective Cohort of Patients with Sepsis. PLoS One 2016; 11:e0163542. [PMID: 27788141 PMCID: PMC5082941 DOI: 10.1371/journal.pone.0163542] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 09/09/2016] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Growth arrest-specific gene 6 (Gas6), a vitamin K-dependent protein interacting with anionic phospholipids and TAM tyrosine kinase receptors, is elevated in plasma of septic patients. Previous studies did not find different levels between survivors and non-survivors at admission because either they included a low number of patients (<50) or a low number of non-survivors (5%). OBJECTIVES To determine, in a larger cohort of septic patients comprising an expected number of non-survivors, the performance of the plasma level of Gas6 and its soluble receptor Axl (sAxl) within 24 hours of admission to predict in-ICU mortality. PATIENTS Septic adults with or without shock. METHODS Gas6 and sAxl were prospectively measured by ELISA at day 0, 3, 7, and then weekly until discharge or death. RESULTS We evaluated 129 septic patients, including 82 with and 47 without shock, with in-ICU mortality rate of 19.4% and in-hospital mortality rate of 26%. Gas6 level was higher in non-survivors than in survivors (238 vs. 167%, P = 0.003); this difference remained constant during the ICU stay. The area under the ROC curve for Gas6 (0.695 [95% CI: 0.58-0.81]) was higher than for sAxl, procalcitonin, CRP, IL-1beta, IL-6 and-alpha, and slightly higher than for IL-8, IL-10, SOFA and APACHEII scores in predicting in-ICU mortality. Considering 249% as a cut-off value, Gas6 measurement had a negative predictive value for mortality of 87%. CONCLUSION It seems that Gas6 plasma level within 24 hours of ICU admission may predicts in-ICU mortality in patients with sepsis. If our result are confirmed in external validation, Gas6 plasma level measurement could contribute to the identification of patients who may benefit most from more aggressive management.
Collapse
Affiliation(s)
- Grégoire Stalder
- Service and Central Laboratory of Hematology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Yok Ai Que
- Service of Adult Intensive Care Medicine, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Sara Calzavarini
- Service and Central Laboratory of Hematology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Laurent Burnier
- Service and Central Laboratory of Hematology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Christophe Kosinski
- Service and Central Laboratory of Hematology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Pierluigi Ballabeni
- Clinical Research Centre, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Thierry Roger
- Infectious Diseases Service, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Thierry Calandra
- Infectious Diseases Service, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Michel A. Duchosal
- Service and Central Laboratory of Hematology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Lucas Liaudet
- Service of Adult Intensive Care Medicine, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Philippe Eggimann
- Service of Adult Intensive Care Medicine, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Anne Angelillo-Scherrer
- Service and Central Laboratory of Hematology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of Clinical Research, University of Bern, Bern, Switzerland
- * E-mail:
| |
Collapse
|
43
|
The Role of TAM Family Receptors in Immune Cell Function: Implications for Cancer Therapy. Cancers (Basel) 2016; 8:cancers8100097. [PMID: 27775650 PMCID: PMC5082387 DOI: 10.3390/cancers8100097] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/13/2016] [Accepted: 10/14/2016] [Indexed: 01/30/2023] Open
Abstract
The TAM receptor protein tyrosine kinases-Tyro3, Axl, and Mer-are essential regulators of immune homeostasis. Guided by their cognate ligands Growth arrest-specific gene 6 (Gas6) and Protein S (Pros1), these receptors ensure the resolution of inflammation by dampening the activation of innate cells as well as by restoring tissue function through promotion of tissue repair and clearance of apoptotic cells. Their central role as negative immune regulators is highlighted by the fact that deregulation of TAM signaling has been linked to the pathogenesis of autoimmune, inflammatory, and infectious diseases. Importantly, TAM receptors have also been associated with cancer development and progression. In a cancer setting, TAM receptors have a dual regulatory role, controlling the initiation and progression of tumor development and, at the same time, the associated anti-tumor responses of diverse immune cells. Thus, modulation of TAM receptors has emerged as a potential novel strategy for cancer treatment. In this review, we discuss our current understanding of how TAM receptors control immunity, with a particular focus on the regulation of anti-tumor responses and its implications for cancer immunotherapy.
Collapse
|
44
|
Klase ZA, Khakhina S, Schneider ADB, Callahan MV, Glasspool-Malone J, Malone R. Zika Fetal Neuropathogenesis: Etiology of a Viral Syndrome. PLoS Negl Trop Dis 2016; 10:e0004877. [PMID: 27560129 PMCID: PMC4999274 DOI: 10.1371/journal.pntd.0004877] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The ongoing Zika virus epidemic in the Americas and the observed association with both fetal abnormalities (primary microcephaly) and adult autoimmune pathology (Guillain-Barré syndrome) has brought attention to this neglected pathogen. While initial case studies generated significant interest in the Zika virus outbreak, larger prospective epidemiology and basic virology studies examining the mechanisms of Zika viral infection and associated pathophysiology are only now starting to be published. In this review, we analyze Zika fetal neuropathogenesis from a comparative pathology perspective, using the historic metaphor of "TORCH" viral pathogenesis to provide context. By drawing parallels to other viral infections of the fetus, we identify common themes and mechanisms that may illuminate the observed pathology. The existing data on the susceptibility of various cells to both Zika and other flavivirus infections are summarized. Finally, we highlight relevant aspects of the known molecular mechanisms of flavivirus replication.
Collapse
Affiliation(s)
- Zachary A Klase
- Department of Biological Sciences, University of the Sciences, Philadelphia, Pennsylvania, United States of America
| | - Svetlana Khakhina
- Department of Biological Sciences, University of the Sciences, Philadelphia, Pennsylvania, United States of America
| | - Adriano De Bernardi Schneider
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, North Carolina, United States of America
| | - Michael V Callahan
- Department of Medicine, Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Zika Foundation, College Station, Texas, United States of America
| | - Jill Glasspool-Malone
- Atheric Pharmaceutical, Scottsville, Virginia, United States of America
- Global Clinical Scholars Research Training Program, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Robert Malone
- Atheric Pharmaceutical, Scottsville, Virginia, United States of America
- Global Clinical Scholars Research Training Program, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
45
|
Schmid ET, Pang IK, Carrera Silva EA, Bosurgi L, Miner JJ, Diamond MS, Iwasaki A, Rothlin CV. AXL receptor tyrosine kinase is required for T cell priming and antiviral immunity. eLife 2016; 5. [PMID: 27350258 PMCID: PMC4924996 DOI: 10.7554/elife.12414] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 06/01/2016] [Indexed: 12/21/2022] Open
Abstract
The receptor tyrosine kinase (RTK) AXL is induced in response to type I interferons (IFNs) and limits their production through a negative feedback loop. Enhanced production of type I IFNs in Axl-/-dendritic cells (DCs) in vitro have led to speculation that inhibition of AXL would promote antiviral responses. Notwithstanding, type I IFNs also exert potent immunosuppressive functions. Here we demonstrate that ablation of AXL enhances the susceptibility to infection by influenza A virus and West Nile virus. The increased type I IFN response in Axl-/- mice was associated with diminished DC maturation, reduced production of IL-1β, and defective antiviral T cell immunity. Blockade of type I IFN receptor or administration of IL-1β to Axl-/- mice restored the antiviral adaptive response and control of infection. Our results demonstrate that AXL is essential for limiting the immunosuppressive effects of type I IFNs and enabling the induction of protective antiviral adaptive immunity. DOI:http://dx.doi.org/10.7554/eLife.12414.001 The immune system must be ever vigilant to ward off infections. Immune cells called T-cells can identify and eliminate microbes, but if they are too aggressive, they can damage the body. To prevent this, the body has systems that control immune responses. For example, another type of immune cell called a dendritic cell produces proteins known as type 1 interferons, which help to fight viral infections while limiting other immune responses. An enzyme called AXL blocks the production of type 1 interferons. Many scientists believe that this activity reduces the ability of individual cells in the body to defend themselves against attacking viruses. In fact, experiments with cells grown in the laboratory have shown that some viruses activate the AXL enzyme to help them infect. Similar studies have also shown that inhibiting AXL and related enzymes can make cells more able to fight off certain types of viral infection. These and other studies suggested that some drugs that block AXL might be useful treatments for viral infections, however it was not clear if this was the case for all viruses. Now, Schmid et al. show that the loss of AXL actually makes mice more prone to infections by the influenza virus and West Nile Virus. In the experiments, mice genetically engineered to lack AXL were more likely than normal mice to become ill after exposure to one of the viruses. Furthermore, fewer T cells matured to the stage where they could attack the virus in these mice. Next, Schmid et al. show that blocking the production of type 1 interferons in the mice that lack AXL restores their ability to fight off these viral infections. This is because type 1 interferons limit the production of a protein that helps the dendritic cells to mature. Therefore, Schmid et al.’s findings show that AXL is vital for mice to fight off viral infections because it helps to balance the antiviral and immune suppressing activities of type 1 interferons. The findings also suggest that using drugs that block AXL to treat infections with certain viruses, including influenza and West Nile Virus, might do more harm than good. DOI:http://dx.doi.org/10.7554/eLife.12414.002
Collapse
Affiliation(s)
- Edward T Schmid
- Department of Immunobiology, School of Medicine, Yale University, New Haven, United States
| | - Iris K Pang
- Department of Immunobiology, School of Medicine, Yale University, New Haven, United States
| | | | - Lidia Bosurgi
- Department of Immunobiology, School of Medicine, Yale University, New Haven, United States
| | - Jonathan J Miner
- Department of Medicine, Washington University School of Medicine, St Louis, United States
| | - Michael S Diamond
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, United States.,Department of Pathology and Immunology, Washington University School of Medicine, St Louis, United States.,The Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, United States
| | - Akiko Iwasaki
- Department of Immunobiology, School of Medicine, Yale University, New Haven, United States.,Howard Hughes Medical Institute, Yale University, New Haven, United States
| | - Carla V Rothlin
- Department of Immunobiology, School of Medicine, Yale University, New Haven, United States
| |
Collapse
|
46
|
Malawista A, Wang X, Trentalange M, Allore HG, Montgomery RR. Coordinated expression of tyro3, axl, and mer receptors in macrophage ontogeny. ACTA ACUST UNITED AC 2016; 3. [PMID: 27695708 PMCID: PMC5040214 DOI: 10.14800/macrophage.1261] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The TAM receptors (Tyro3, Axl, and Mer) are a family of homologous receptor-tyrosine kinases that inhibit Toll-like receptor signaling to regulate downstream pathways and restore homeostasis. TAM triple mutant mice (Tyro3−/−, Axl−/−, Mer−/−) have elevated levels of pro-inflammatory cytokines and are prone to developing lymphoproliferative disorders and autoimmunity. Understanding differential expression of TAM receptors among human subjects is critical to harnessing this pathway for therapeutic interventions. We have quantified changes in TAM expression during the ontogeny of human macrophages using paired samples of monocytes and macrophages to take advantage of characteristic expression within an individual. No significant differences in levels of Tyro3 were found between monocytes and macrophages (flow cytometry: p=0.652, immunoblot: p=0.231, qPCR: p=0.389). Protein levels of Axl were reduced (flow cytometry: p=0.049, immunoblot: p<0.001) when monocytes matured to macrophages. No significant differences in the levels of Axl mRNA transcripts were found (qPCR: p=0.082), however, Tyro3 and Axl were proportionate. The most striking difference was upregulation of expression of Mer with both protein and mRNA being significantly increased when monocytes developed into macrophages (flow cytometry: p<0.001, immunoblot: p<0.001, qPCR: p=0.004). A fuller characterization of TAM receptor expression in macrophage ontogeny informs our understanding of their function and potential therapeutic interventions.
Collapse
Affiliation(s)
- Anna Malawista
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Xiaomei Wang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Mark Trentalange
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Heather G Allore
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Ruth R Montgomery
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06519, USA; Program on Human Translational Immunology, Yale University School of Medicine, New Haven, CT, 06519, USA
| |
Collapse
|
47
|
Bryant C, Fromm PD, Kupresanin F, Clark G, Lee K, Clarke C, Silveira PA, Suen H, Brown R, Newman E, Cunningham I, Ho PJ, Gibson J, Bradstock K, Joshua D, Hart DN. A CD2 high-expressing stress-resistant human plasmacytoid dendritic-cell subset. Immunol Cell Biol 2016; 94:447-57. [PMID: 26791160 DOI: 10.1038/icb.2015.116] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 12/15/2015] [Accepted: 12/15/2015] [Indexed: 01/22/2023]
Abstract
Human plasmacytoid dendritic cells (pDCs) were considered to be a phenotypically and functionally homogeneous cell population; however, recent analyses indicate potential heterogeneity. This is of major interest, given their importance in the induction of anti-viral responses and their role in creating immunologically permissive environments for human malignancies. For this reason, we investigated the possible presence of human pDC subsets in blood and bone marrow, using unbiased cell phenotype clustering and functional studies. This defined two major functionally distinct human pDC subsets, distinguished by differential expression of CD2. The CD2(hi) and CD2(lo) pDCs represent discontinuous subsets, each with hallmark pDC functionality, including interferon-alpha production. The rarer CD2(hi) pDC subset demonstrated a significant survival advantage over CD2(lo) pDC during stress and upon exposure to glucocorticoids (GCs), which was associated with higher expression of the anti-apoptotic molecule BCL2. The differential sensitivity of these two human pDC subsets to GCs is demonstrated in vivo by a relative increase in CD2(hi) pDC in multiple myeloma patients treated with GCs. Hence, the selective apoptosis of CD2(lo) pDC during stress represents a novel mechanism for the control of innate responses.
Collapse
Affiliation(s)
- Christian Bryant
- Dendritic Cell Research, ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, NSW, Australia.,Concord Clinical School, University of Sydney, Sydney, NSW, Australia.,Department of Haematology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Phillip D Fromm
- Dendritic Cell Research, ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, NSW, Australia.,Concord Clinical School, University of Sydney, Sydney, NSW, Australia
| | - Fiona Kupresanin
- Dendritic Cell Research, ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, NSW, Australia
| | - Georgina Clark
- Dendritic Cell Research, ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, NSW, Australia.,Concord Clinical School, University of Sydney, Sydney, NSW, Australia
| | - Kenneth Lee
- Concord Clinical School, University of Sydney, Sydney, NSW, Australia.,Department of Anatomical Pathology, Concord Repatriation General Hospital, Sydney, NSW, Australia
| | - Candice Clarke
- Department of Anatomical Pathology, Concord Repatriation General Hospital, Sydney, NSW, Australia
| | - Pablo A Silveira
- Dendritic Cell Research, ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, NSW, Australia.,Concord Clinical School, University of Sydney, Sydney, NSW, Australia
| | - Hayley Suen
- Department of Haematology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Ross Brown
- Department of Haematology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Elizabeth Newman
- Department of Haematology, Concord Hospital, Sydney, NSW, Australia
| | - Ilona Cunningham
- Department of Haematology, Concord Hospital, Sydney, NSW, Australia
| | - P Joy Ho
- Department of Haematology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - John Gibson
- Department of Haematology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Kenneth Bradstock
- Dendritic Cell Research, ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, NSW, Australia.,Blood and Bone Marrow Transplant Service, Westmead Hospital, Sydney, NSW, Australia
| | - Douglas Joshua
- Department of Haematology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Derek Nj Hart
- Dendritic Cell Research, ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, NSW, Australia.,Concord Clinical School, University of Sydney, Sydney, NSW, Australia.,Department of Haematology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.,Department of Haematology, Concord Hospital, Sydney, NSW, Australia
| |
Collapse
|
48
|
Read SA, Tay ES, Shahidi M, O’Connor KS, Booth DR, George J, Douglas MW. Hepatitis C Virus Driven AXL Expression Suppresses the Hepatic Type I Interferon Response. PLoS One 2015; 10:e0136227. [PMID: 26313459 PMCID: PMC4551482 DOI: 10.1371/journal.pone.0136227] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Accepted: 07/30/2015] [Indexed: 01/06/2023] Open
Abstract
Treatment of chronic hepatitis C virus (HCV) infection is evolving rapidly with the development of novel direct acting antivirals (DAAs), however viral clearance remains intimately linked to the hepatic innate immune system. Patients demonstrating a high baseline activation of interferon stimulated genes (ISGs), termed interferon refractoriness, are less likely to mount a strong antiviral response and achieve viral clearance when placed on treatment. As a result, suppressor of cytokine signalling (SOCS) 3 and other regulators of the IFN response have been identified as key candidates for the IFN refractory phenotype due to their regulatory role on the IFN response. AXL is a receptor tyrosine kinase that has been identified as a key regulator of interferon (IFN) signalling in myeloid cells of the immune system, but has not been examined in the context of chronic HCV infection. Here, we show that AXL is up-regulated following HCV infection, both in vitro and in vivo and is likely induced by type I/III IFNs and inflammatory signalling pathways. AXL inhibited type IFNα mediated ISG expression resulting in a decrease in its antiviral efficacy against HCV in vitro. Furthermore, patients possessing the favourable IFNL3 rs12979860 genotype associated with treatment response, showed lower AXL expression in the liver and a stronger induction of AXL in the blood, following their first dose of IFN. Together, these data suggest that elevated AXL expression in the liver may mediate an IFN-refractory phenotype characteristic of patients possessing the unfavourable rs12979860 genotype, which is associated with lower rates of viral clearance.
Collapse
Affiliation(s)
- Scott A. Read
- Storr Liver Centre, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, Australia
| | - Enoch S. Tay
- Storr Liver Centre, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, Australia
| | - Mahsa Shahidi
- Storr Liver Centre, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, Australia
| | - Kate S. O’Connor
- Centre for Immunology and Allergy Research, University of Sydney at Westmead Hospital, Westmead, Australia
| | - David R. Booth
- Centre for Immunology and Allergy Research, University of Sydney at Westmead Hospital, Westmead, Australia
| | - Jacob George
- Storr Liver Centre, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, Australia
| | - Mark W. Douglas
- Storr Liver Centre, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, Australia
- Centre for Infectious Diseases and Microbiology, Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney at Westmead Hospital, Westmead, Australia
| |
Collapse
|
49
|
Rothlin CV, Carrera-Silva EA, Bosurgi L, Ghosh S. TAM receptor signaling in immune homeostasis. Annu Rev Immunol 2015; 33:355-91. [PMID: 25594431 DOI: 10.1146/annurev-immunol-032414-112103] [Citation(s) in RCA: 345] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The TAM receptor tyrosine kinases (RTKs)-TYRO3, AXL, and MERTK-together with their cognate agonists GAS6 and PROS1 play an essential role in the resolution of inflammation. Deficiencies in TAM signaling have been associated with chronic inflammatory and autoimmune diseases. Three processes regulated by TAM signaling may contribute, either independently or collectively, to immune homeostasis: the negative regulation of the innate immune response, the phagocytosis of apoptotic cells, and the restoration of vascular integrity. Recent studies have also revealed the function of TAMs in infectious diseases and cancer. Here, we review the important milestones in the discovery of these RTKs and their ligands and the studies that underscore the functional importance of this signaling pathway in physiological immune settings and disease.
Collapse
|
50
|
Abstract
Three receptor tyrosine kinases, Tyro3, Axl, and Mertk (TAM) and their ligands Gas6 and Protein S, have emerged as potent negative regulators of innate immune responses. A number of studies using genetic ablation of TAM loci in mice have elucidated the mechanism of TAM engagement and function during the immune response and removal of apoptotic cells. Following phagocytosis of apoptotic cells or the induction of T-cell dependent adaptive immune responses, ligand-induced TAM signaling dampens proinflammatory cytokine production and thus prevents exaggerated or prolonged inflammation. It is believed that the TAM pathway may play an important role in the pathogenesis of inflammatory bowel disease. Suppression of inflammation and removal of apoptotic cells followed by tissue repair are essential processes for disease remission and the successful management of inflammatory bowel disease. In light of the key role of TAMs in controlling inflammatory responses, here, we review the recent advances on TAM research vis-à-vis the resolution of intestinal inflammation. Targeted activation of TAM receptor tyrosine kinases may represent a potent therapeutic opportunity in inflammatory bowel disease.
Collapse
|