1
|
Lv LX, Zhang Q, Zhao XF, Wang JX. Identification of COP9 signalosome (CSN) subunits and antiviral function analysis of CSN5 in shrimp. FISH & SHELLFISH IMMUNOLOGY 2024; 151:109679. [PMID: 38844185 DOI: 10.1016/j.fsi.2024.109679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 04/01/2024] [Accepted: 06/04/2024] [Indexed: 06/10/2024]
Abstract
The constitutive photomorphogenesis 9 (COP9) signalosome (CSN) typically composing of eight subunits (CSN1-8) mediates the process of deneddylation and deubiquitination. The fifth subunit of COP9 signalosome, CSN5, has special characteristics compared with the other seven subunits, and plays vital roles in the deneddylation activity and diverse cellular processes. However, the role of CSN5 in antiviral immunity is not clear. In this study, we identified 8 subunits (CSN1-8) of COP9 signalosome in shrimp Marsupenaeus japonicus. CSN1-6 were existed in all tested tissues, but CSN7-CSN8 were not detected in hepatopancreas. After WSSV challenged, the expression level of Csn1 to Csn4, and Csn6 to Csn8 were highly decreased, but the expression level of Csn5 was conspicuously increased in shrimp challenged by white spot syndrome virus (WSSV). The CSN5 was recombinantly expressed in Escherichia coli and its polyclonal antibody was prepared. The expression level of CSN5 was conspicuously increased at RNA and protein levels in the shrimp challenged by WSSV. After knockdown of Csn5 by RNA interference, the WSSV replication was obviously increased in shrimp. When injected the recombinant protein of CSN5 with the membrane penetrating peptide into shrimp, WSSV replication was inhibited and the survival rate of shrimp was significantly improved compared with control. We further analyzed the expression of antimicrobial peptides (AMPs) in Csn5-RNAi shrimp, and the results showed that the expression of several AMPs was declined significantly. These results indicate that CSN5 inhibits replication of WSSV via regulating expression of AMPs in shrimp, and the recombinant CSN5 might be used in shrimp aquaculture for the white spot syndrome disease control.
Collapse
Affiliation(s)
- Li-Xia Lv
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong, China
| | - Qiang Zhang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong, China
| | - Xiao-Fan Zhao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong, China.
| | - Jin-Xing Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong, China.
| |
Collapse
|
2
|
Klaus T, Hieber C, Bros M, Grabbe S. Integrins in Health and Disease-Suitable Targets for Treatment? Cells 2024; 13:212. [PMID: 38334604 PMCID: PMC10854705 DOI: 10.3390/cells13030212] [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: 12/27/2023] [Revised: 01/13/2024] [Accepted: 01/22/2024] [Indexed: 02/10/2024] Open
Abstract
Integrin receptors are heterodimeric surface receptors that play multiple roles regarding cell-cell communication, signaling, and migration. The four members of the β2 integrin subfamily are composed of an alternative α (CD11a-d) subunit, which determines the specific receptor properties, and a constant β (CD18) subunit. This review aims to present insight into the multiple immunological roles of integrin receptors, with a focus on β2 integrins that are specifically expressed by leukocytes. The pathophysiological role of β2 integrins is confirmed by the drastic phenotype of patients suffering from leukocyte adhesion deficiencies, most often resulting in severe recurrent infections and, at the same time, a predisposition for autoimmune diseases. So far, studies on the role of β2 integrins in vivo employed mice with a constitutive knockout of all β2 integrins or either family member, respectively, which complicated the differentiation between the direct and indirect effects of β2 integrin deficiency for distinct cell types. The recent generation and characterization of transgenic mice with a cell-type-specific knockdown of β2 integrins by our group has enabled the dissection of cell-specific roles of β2 integrins. Further, integrin receptors have been recognized as target receptors for the treatment of inflammatory diseases as well as tumor therapy. However, whereas both agonistic and antagonistic agents yielded beneficial effects in animal models, the success of clinical trials was limited in most cases and was associated with unwanted side effects. This unfavorable outcome is most probably related to the systemic effects of the used compounds on all leukocytes, thereby emphasizing the need to develop formulations that target distinct types of leukocytes to modulate β2 integrin activity for therapeutic applications.
Collapse
Affiliation(s)
| | | | | | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (T.K.); (C.H.); (M.B.)
| |
Collapse
|
3
|
Antony IR, Wong BHS, Kelleher D, Verma NK. Maladaptive T-Cell Metabolic Fitness in Autoimmune Diseases. Cells 2023; 12:2541. [PMID: 37947619 PMCID: PMC10650071 DOI: 10.3390/cells12212541] [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: 09/26/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
Immune surveillance and adaptive immune responses, involving continuously circulating and tissue-resident T-lymphocytes, provide host defense against infectious agents and possible malignant transformation while avoiding autoimmune tissue damage. Activation, migration, and deployment of T-cells to affected tissue sites are crucial for mounting an adaptive immune response. An effective adaptive immune defense depends on the ability of T-cells to dynamically reprogram their metabolic requirements in response to environmental cues. Inability of the T-cells to adapt to specific metabolic demands may skew cells to become either hyporesponsive (creating immunocompromised conditions) or hyperactive (causing autoimmune tissue destruction). Here, we review maladaptive T-cell metabolic fitness that can cause autoimmune diseases and discuss how T-cell metabolic programs can potentially be modulated to achieve therapeutic benefits.
Collapse
Affiliation(s)
- Irene Rose Antony
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- Vellore Institute of Technology, Vellore 632014, India; (I.R.A.); (B.H.S.W.); (D.K.)
| | - Brandon Han Siang Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- Interdisciplinary Graduate Programme, NTU Institute for Health Technologies (HealthTech-NTU), Nanyang Technological University, Singapore 637335, Singapore
| | - Dermot Kelleher
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- Skin Research Institute of Singapore, Singapore 308205, Singapore
| |
Collapse
|
4
|
Shi H, Shao B. LFA-1 Activation in T-Cell Migration and Immunological Synapse Formation. Cells 2023; 12:cells12081136. [PMID: 37190045 DOI: 10.3390/cells12081136] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/02/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Integrin LFA-1 plays a critical role in T-cell migration and in the formation of immunological synapses. LFA-1 functions through interacting with its ligands with differing affinities: low, intermediate, and high. Most prior research has studied how LFA-1 in the high-affinity state regulates the trafficking and functions of T cells. LFA-1 is also presented in the intermediate-affinity state on T cells, however, the signaling to activate LFA-1 to the intermediate-affinity state and the role of LFA-1 in this affinity state both remain largely elusive. This review briefly summarizes the activation and roles of LFA-1 with varied ligand-binding affinities in the regulation of T-cell migration and immunological synapse formation.
Collapse
Affiliation(s)
- Huiping Shi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Bojing Shao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| |
Collapse
|
5
|
Fazil MHUT, Chirumamilla CS, Perez-Novo C, Wong BHS, Kumar S, Sze SK, Vanden Berghe W, Verma NK. The steroidal lactone withaferin A impedes T-cell motility by inhibiting the kinase ZAP70 and subsequent kinome signaling. J Biol Chem 2021; 297:101377. [PMID: 34742736 PMCID: PMC8637146 DOI: 10.1016/j.jbc.2021.101377] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 02/07/2023] Open
Abstract
The steroidal lactone withaferin A (WFA) is a dietary phytochemical, derived from Withania somnifera. It exhibits a wide range of biological properties, including immunomodulatory, anti-inflammatory, antistress, and anticancer activities. Here we investigated the effect of WFA on T-cell motility, which is crucial for adaptive immune responses as well as autoimmune reactions. We found that WFA dose-dependently (within the concentration range of 0.3–1.25 μM) inhibited the ability of human T-cells to migrate via cross-linking of the lymphocyte function-associated antigen-1 (LFA-1) integrin with its ligand, intercellular adhesion molecule 1 (ICAM-1). Coimmunoprecipitation of WFA interacting proteins and subsequent tandem mass spectrometry identified a WFA-interactome consisting of 273 proteins in motile T-cells. In particular, our data revealed significant enrichment of the zeta-chain-associated protein kinase 70 (ZAP70) and cytoskeletal actin protein interaction networks upon stimulation. Phospho-peptide mapping and kinome analysis substantiated kinase signaling downstream of ZAP70 as a key WFA target, which was further confirmed by bait-pulldown and Western immunoblotting assays. The WFA-ZAP70 interaction was disrupted by a disulfide reducing agent dithiothreitol, suggesting an involvement of cysteine covalent binding interface. In silico docking predicted WFA binding to ZAP70 at cystine 560 and 564 residues. These findings provide a mechanistic insight whereby WFA binds to and inhibits the ZAP70 kinase and impedes T-cell motility. We therefore conclude that WFA may be exploited to pharmacologically control host immune responses and potentially prevent autoimmune-mediated pathologies.
Collapse
Affiliation(s)
| | - Chandra Sekhar Chirumamilla
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signaling (PPES) and Integrated Personalized and Precision Oncology Network (IPPON), Department of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Claudina Perez-Novo
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signaling (PPES) and Integrated Personalized and Precision Oncology Network (IPPON), Department of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Brandon Han Siang Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, Singapore; NTU Institute for Health Technologies (HealthTech NTU), Interdisciplinary Graduate Programme, Nanyang Technological University Singapore, Singapore
| | - Sunil Kumar
- Indian Council of Agricultural Research-National Bureau of Agriculturally Important Microorganisms, Kushmaur, Mau, Uttar Pradesh, India
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore
| | - Wim Vanden Berghe
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signaling (PPES) and Integrated Personalized and Precision Oncology Network (IPPON), Department of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium.
| | - Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, Singapore.
| |
Collapse
|
6
|
Wang H, Li X, Kajikawa T, Shin J, Lim JH, Kourtzelis I, Nagai K, Korostoff JM, Grossklaus S, Naumann R, Chavakis T, Hajishengallis G. Stromal cell-derived DEL-1 inhibits Tfh cell activation and inflammatory arthritis. J Clin Invest 2021; 131:e150578. [PMID: 34403362 PMCID: PMC8483759 DOI: 10.1172/jci150578] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 08/12/2021] [Indexed: 11/17/2022] Open
Abstract
The secreted protein developmental endothelial locus 1 (DEL-1) regulates inflammatory cell recruitment and protects against inflammatory pathologies in animal models. Here, we investigated DEL-1 in inflammatory arthritis using collagen-induced arthritis (CIA) and collagen Ab-induced arthritis (CAIA) models. In both models, mice with endothelium-specific overexpression of DEL-1 were protected from arthritis relative to WT controls, whereas arthritis was exacerbated in DEL-1-deficient mice. Compared with WT controls, mice with collagen VI promoter-driven overexpression of DEL-1 in mesenchymal cells were protected against CIA but not CAIA, suggesting a role for DEL-1 in the induction of the arthritogenic Ab response. Indeed, DEL-1 was expressed in perivascular stromal cells of the lymph nodes and inhibited Tfh and germinal center B cell responses. Mechanistically, DEL-1 inhibited DC-dependent induction of Tfh cells by targeting the LFA-1 integrin on T cells. Overall, DEL-1 restrained arthritis through a dual mechanism, one acting locally in the joints and associated with the anti-recruitment function of endothelial cell-derived DEL-1; the other mechanism acting systemically in the lymph nodes and associated with the ability of stromal cell-derived DEL-1 to restrain Tfh responses. DEL-1 may therefore be a promising therapeutic for the treatment of inflammatory arthritis.
Collapse
Affiliation(s)
- Hui Wang
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xiaofei Li
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tetsuhiro Kajikawa
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jieun Shin
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jong-Hyung Lim
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ioannis Kourtzelis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Hull York Medical School, York Biomedical Research Institute, University of York, York, United Kingdom
| | - Kosuke Nagai
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Jonathan M. Korostoff
- Department of Periodontics, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sylvia Grossklaus
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Ronald Naumann
- Transgenic Core Facility, Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - George Hajishengallis
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
7
|
Interleukin-1 β Enhances Umbilical Cord Mesenchymal Stem Cell Adhesion Ability on Human Umbilical Vein Endothelial Cells via LFA-1/ICAM-1 Interaction. Stem Cells Int 2019; 2019:7267142. [PMID: 31949440 PMCID: PMC6948307 DOI: 10.1155/2019/7267142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/13/2019] [Accepted: 11/19/2019] [Indexed: 12/13/2022] Open
Abstract
The migration of administered mesenchymal stem cells (MSCs) to sites of injury via the bloodstream has been demonstrated. However, the underlying mechanisms of umbilical cord MSC adhesion to endothelial cells during transendothelial migration are still unclear. In this study, our data showed that IL-1β induced LFA-1 expression on MSCs and ICAM-1 expression on HUVECs. We then pretreated MSCs with protein synthesis inhibitor cycloheximide. The results showed that IL-1β induced LFA-1 expression on the surface of MSCs via the protein synthesis pathway. Through the p38 MAPK signaling pathway inhibitor SB 203580, we found that IL-1β induces the expression of LFA-1 through p38 MAPK signaling and enhances ICAM-1 expression in HUVECs. In addition, IL-1β-induced MSC adhesion to HUVECs was found to be inhibited by IL-1RA and the LFA-1 inhibitor lovastatin. These results indicate that IL-1β promotes the cell adhesion of MSCs to HUVECs through LFA-1/ICAM-1 interaction. We address the evidence that the cell adhesion mechanism of IL-1β promotes MSC adhesion to HUVECs. The implications of these findings could enhance the therapeutic potential of MSCs.
Collapse
|
8
|
Verma NK, Chalasani MLS, Scott JD, Kelleher D. CG-NAP/Kinase Interactions Fine-Tune T Cell Functions. Front Immunol 2019; 10:2642. [PMID: 31781123 PMCID: PMC6861388 DOI: 10.3389/fimmu.2019.02642] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/24/2019] [Indexed: 01/04/2023] Open
Abstract
CG-NAP, also known as AKAP450, is an anchoring/adaptor protein that streamlines signal transduction in various cell types by localizing signaling proteins and enzymes with their substrates. Great efforts are being devoted to elucidating functional roles of this protein and associated macromolecular signaling complex. Increasing understanding of pathways involved in regulating T lymphocytes suggests that CG-NAP can facilitate dynamic interactions between kinases and their substrates and thus fine-tune T cell motility and effector functions. As a result, new binding partners of CG-NAP are continually being uncovered. Here, we review recent advances in CG-NAP research, focusing on its interactions with kinases in T cells with an emphasis on the possible role of this anchoring protein as a target for therapeutic intervention in immune-mediated diseases.
Collapse
Affiliation(s)
- Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | | | - John D Scott
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, United States
| | - Dermot Kelleher
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore.,Departments of Medicine and Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
9
|
Abstract
The inherent ability of T-cells to migrate is critical for a fully functional immune system, both in normal immune surveillance and for mounting an adaptive immune response. At the same time, inappropriate trafficking of T-cells can be a pathological factor for immune-mediated or chronic inflammatory diseases. T-cell motility is critically dependent on a series of ligand-receptor interactions, a precisely regulated intracellular signaling, an involvement of adaptor proteins, and dynamic remodeling of the cytoskeletal systems. The leukocyte integrin LFA-1 receptor present on T-cells binds to the ligand intercellular adhesion molecule 1 (ICAM-1) and this LFA-1/ICAM-1 contact acts as a trigger for T-cell motility. In this book, we present a collection of methods and protocols that are frequently used by researchers to better understand T-cell motility in health and diseases.
Collapse
Affiliation(s)
- Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore.
| | - Dermot Kelleher
- Lymphocyte Signalling Research Laboratory, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore.,Departments of Medicine and Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
10
|
Abstract
Regulated migration of T-lymphocytes through high endothelial venules and secondary lymphoid organs is necessary for an adaptive immune response. Uncontrolled trafficking of T-cells is implicated in many pathological conditions, including autoimmune disorders, such as psoriasis and inflammatory bowel disease. T-cell migration is regulated mainly by the αLβ2 integrin receptor LFA-1, which interacts primarily with its cognate ligand ICAM-1 expressed on the endothelium. This interaction triggers a plethora of downstream signaling pathways, which are not fully understood. Thus, in order to dissect the signal transduction processes at molecular levels and phenotypic changes in migrating T-cells, a laboratory model mimicking T-cell motility is important. Here, we describe a simple and highly reproducible in vitro model to study T-cell migration.
Collapse
|
11
|
Roy NH, MacKay JL, Robertson TF, Hammer DA, Burkhardt JK. Crk adaptor proteins mediate actin-dependent T cell migration and mechanosensing induced by the integrin LFA-1. Sci Signal 2018; 11:eaat3178. [PMID: 30538176 PMCID: PMC6333317 DOI: 10.1126/scisignal.aat3178] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
T cell entry into inflamed tissue involves firm adhesion, spreading, and migration of the T cells across endothelial barriers. These events depend on "outside-in" signals through which engaged integrins direct cytoskeletal reorganization. We investigated the molecular events that mediate this process and found that T cells from mice lacking expression of the adaptor protein Crk exhibited defects in phenotypes induced by the integrin lymphocyte function-associated antigen 1 (LFA-1), namely, actin polymerization, leading edge formation, and two-dimensional cell migration. Crk protein was an essential mediator of LFA-1 signaling-induced phosphorylation of the E3 ubiquitin ligase c-Cbl and its subsequent interaction with the phosphatidylinositol 3-kinase (PI3K) subunit p85, thus promoting PI3K activity and cytoskeletal remodeling. In addition, we found that Crk proteins were required for T cells to respond to changes in substrate stiffness, as measured by alterations in cell spreading and differential phosphorylation of the force-sensitive protein CasL. These findings identify Crk proteins as key intermediates coupling LFA-1 signals to actin remodeling and provide mechanistic insights into how T cells sense and respond to substrate stiffness.
Collapse
Affiliation(s)
- Nathan H Roy
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joanna L MacKay
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Tanner F Robertson
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel A Hammer
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Janis K Burkhardt
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA.
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
12
|
Plaumann J, Engelhardt M, Awwad MHS, Echchannaoui H, Amman E, Raab MS, Hillengass J, Halama N, Neuber B, Müller-Tidow C, Goldschmidt H, Hundemer M. IL-10 inducible CD8 + regulatory T-cells are enriched in patients with multiple myeloma and impact the generation of antigen-specific T-cells. Cancer Immunol Immunother 2018; 67:1695-1707. [PMID: 30128739 PMCID: PMC11028289 DOI: 10.1007/s00262-018-2230-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 08/09/2018] [Indexed: 01/07/2023]
Abstract
Tumor-mediated immunosuppression via regulatory T-cells is a key player among the various immune-escape mechanisms in multiple myeloma. We analyzed the generation, distribution, function and immunophenotype of CD8+CD28- regulatory T-cells in patients with multiple myeloma. Functionality of CD8+CD28- T-cells was assessed by immunological assays using ex vivo generated antigen-specific T-cells from patients with plasma cell dyscrasias and healthy donors. Detailed analysis of distribution, immunophenotype and cytotoxic potential of CD8+CD28- T-cells was performed by flow cytometry and ELISA. We found that the amount of CD8+CD28- T-cells was directly correlated with the suppression of antigen-specific T-cell responses in patients with plasma cell dyscrasia. Analyzing the CD8+CD28- T-cells in detail, increased numbers of these cells were observed in the bone marrow (i.e., tumor microenvironment) of patients with plasma cell dyscrasia. Furthermore, we identified the expression of lymphocyte function-associated antigen 1 (LFA-1) as a marker of immunosuppression and defined the CD8+CD28-CD57+LFA-1high population as the relevant immunosuppressive compartment. These regulatory T-cells act as immunosuppressors via soluble factors and incubation with IL-10 augmented their immunosuppressive capacity. The immunosuppressive regulatory network of IL-10 and the CD8+CD28-CD57+LFA-1high regulatory T-cells show unique characteristics and contribute to the tumor immune escape mechanism in patients with multiple myeloma.
Collapse
Affiliation(s)
- Julian Plaumann
- Department of Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - Melanie Engelhardt
- Department of Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - Mohamed H S Awwad
- Department of Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - Hakim Echchannaoui
- Department of Hematology, Oncology and Pneumology, University Medical Center (UMC) of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Eva Amman
- Department of Hematology, Oncology and Pneumology, University Medical Center (UMC) of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Marc S Raab
- Department of Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - Jens Hillengass
- Department of Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - Niels Halama
- National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Brigitte Neuber
- Department of Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - Hartmut Goldschmidt
- Department of Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
- National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Michael Hundemer
- Department of Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
13
|
Nichols KK, Donnenfeld ED, Karpecki PM, Hovanesian JA, Raychaudhuri A, Shojaei A, Zhang S. Safety and tolerability of lifitegrast ophthalmic solution 5.0%: Pooled analysis of five randomized controlled trials in dry eye disease. Eur J Ophthalmol 2018; 29:394-401. [PMID: 30112930 PMCID: PMC6625033 DOI: 10.1177/1120672118791936] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Purpose: Characterize the safety and tolerability of lifitegrast ophthalmic solution
5.0% for the treatment of dry eye disease. Methods: Pooled data from five randomized controlled trials were analyzed. Key
inclusion criteria were adults with dry eye disease (Schirmer tear test
score ⩾1 and ⩽10 mm, eye dryness score ⩾40 (visual analog scale 0–100),
corneal staining score ⩾2.0 (0–4 scale)). Participants were randomized to
lifitegrast ophthalmic solution 5.0% or placebo twice daily for 84 or
360 days. Treatment-emergent adverse events and drop comfort scores were
assessed. Results: Overall, 2464 participants (lifitegrast, n = 1287; placebo, n = 1177) were
included. Ocular treatment-emergent adverse events occurring in >5% in
either group were instillation site irritation (lifitegrast, 15.2%; placebo,
2.8%), instillation site reaction (lifitegrast, 12.3%; placebo, 2.3%), and
instillation site pain (lifitegrast, 9.8%; placebo, 2.1%); the most common
(> 5%) nonocular treatment-emergent adverse event was dysgeusia
(lifitegrast, 14.5%; placebo, 0.3%). The majority of treatment-emergent
adverse events were mild to moderate in severity. Discontinuation due to
treatment-emergent adverse events occurred in 7.0% (lifitegrast) versus 2.6%
(placebo) of participants (ocular: 5.5% vs 1.5%; nonocular: 1.9% vs 1.1%).
Drop comfort scores with lifitegrast improved within 3 min of instillation
and the score at 3 min improved across visits (12-week trials (both eyes,
day 84 vs 0): 2.0 vs 3.3; SONATA (day 360 vs 0): right eye, 1.2 vs 1.7; left
eye, 1.2 vs 1.8). Conclusion: Lifitegrast ophthalmic solution 5.0% appeared to be safe and well tolerated
for the treatment of dry eye disease. Drop comfort with lifitegrast improved
within 3 min of instillation.
Collapse
Affiliation(s)
- Kelly K Nichols
- 1 School of Optometry, The University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Verma NK, Kelleher D. Not Just an Adhesion Molecule: LFA-1 Contact Tunes the T Lymphocyte Program. THE JOURNAL OF IMMUNOLOGY 2017; 199:1213-1221. [PMID: 28784685 DOI: 10.4049/jimmunol.1700495] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 06/02/2017] [Indexed: 12/18/2022]
Abstract
The αLβ2 integrin LFA-1 is known to play a key role in T lymphocyte migration, which is necessary to mount a local immune response, and is also the main driver of autoimmune diseases. This migration-triggering signaling process in T cells is tightly regulated to permit an immune response that is appropriate to the local trigger, as well as to prevent deleterious tissue-damaging bystander effects. Emerging evidence shows that, in addition to prompting a diverse range of downstream signaling cascades, LFA-1 stimulation in T lymphocytes modulates gene-transcription programs, including genetic signatures of TGF-β and Notch pathways, with multifactorial biological outcomes. This review highlights recent findings and discusses molecular mechanisms by which LFA-1 signaling influence T lymphocyte differentiation into the effector subsets Th1, Th17, and induced regulatory T cells. We argue that LFA-1 contact with a cognate ligand, such as ICAM-1, independent of the immune synapse activates a late divergence in T cells' effector phenotypes, hence fine-tuning their functioning.
Collapse
Affiliation(s)
- Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore; and
| | - Dermot Kelleher
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore; and .,Faculty of Medicine, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| |
Collapse
|
15
|
Zhao S, Gu Z, Wang L, Guan L, Wang F, Yang N, Luo L, Gao Z, Song Y, Wang L, Liu D, Gao C. G-CSF inhibits LFA-1-mediated CD4 + T cell functions by inhibiting Lck and ZAP-70. Oncotarget 2017; 8:51578-51590. [PMID: 28881670 PMCID: PMC5584271 DOI: 10.18632/oncotarget.18194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 05/06/2017] [Indexed: 11/25/2022] Open
Abstract
In this study, we showed that G-CSF mobilization increased the frequency of T cells, specifically CD3+CD4+ T cells. G-CSF mobilization decreased the secretion of inflammatory cytokines of CD4+ T cells through the LFA-1/ICAM-1 signaling pathway, whereas it did not alter the TH1/TH2 ratio. We found that G-CSF mobilization inhibited LFA-1-mediated CD4+ T cell polarization and motility. In vitro, G-CSF stimulation also attenuated the polarization and adhesiveness of CD4+ T cells through the LFA-1/ICAM-1 interaction. Further investigation revealed that G-CSF mobilization suppressed LFA-1 signaling by down-regulating Lck and ZAP-70 expression in CD4+ T cells, similar results was also confirmed by in-vitro studies. These findings suggested that G-CSF directly suppressed LFA-1-mediated CD4+ T cell functions through the down-regulation of Lck and ZAP-70. The immunosuppressive effect of G-CSF mobilization deepened our understanding about peripheral blood hematopoietic stem cell transplantation. LFA-1/ICMA-1 pathway may become a potential target for graft-versus-host disease prophylaxis.
Collapse
Affiliation(s)
- Shasha Zhao
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China.,Medical School, Nankai University, Tianjin 300071, China
| | - Zhenyang Gu
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - Li Wang
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China.,Department of Hematology and Oncology, Laoshan Branch, No. 401 Hospital of Chinese PLA, Qingdao 266101, China
| | - Lixun Guan
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - Feiyan Wang
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - Nan Yang
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - Lan Luo
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - Zhe Gao
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - Yingwei Song
- Department of Blood Transfusion, Chinese PLA General Hospital, Beijing 100853, China
| | - Lili Wang
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - Daihong Liu
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - Chunji Gao
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
16
|
Xie Z, Chen J, Zheng C, Wu J, Cheng Y, Zhu S, Lin C, Cao Q, Zhu J, Jin T. 1,25-dihydroxyvitamin D 3 -induced dendritic cells suppress experimental autoimmune encephalomyelitis by increasing proportions of the regulatory lymphocytes and reducing T helper type 1 and type 17 cells. Immunology 2017; 152:414-424. [PMID: 28617989 DOI: 10.1111/imm.12776] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/01/2017] [Accepted: 06/05/2017] [Indexed: 12/26/2022] Open
Abstract
Dendritic cells (DCs), a bridge for innate and adaptive immune responses, play a key role in the development of multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE), an animal model for MS. Administration of tolerogenic DCs has been used as an immunotherapy in autoimmune diseases. Deficiency of vitamin D is an environmental risk factor of MS. In this study, we induced tolerogenic DCs by 1,25-dihydroxyvitamin D3 and transferred the tolerogenic DCs (VD3 -DCs) into EAE mice by adoptive transfer. We found that VD3 -DCs inhibited the infiltrations of T helper type 1 (Th1) and Th17 cells into spinal cord and increased the proportions of regulatory T cells (CD4+ CD25+ Foxp3+ ), CD4+ IL-10+ T cells and regulatory B cells (CD19+ CD5+ CD1d+ ) in peripheral immune organs, which resulted in attenuated EAE. However, the proportions of T helper type 1 (Th1) and Th17 cells in spleen and lymph nodes and the levels of pro-inflammatory cytokines and IgG in serum also increased after transfer of VD3 -DCs. We conclude that transfer of VD3 -DCs suppressed EAE by increasing proportions of regulatory T cells, CD4+ IL-10+ T cells and regulatory B cells in spleen and reducing infiltration of Th1 and Th17 cells into spinal cord, which suggests a possible immunotherapy method using VD3 -DCs in MS.
Collapse
Affiliation(s)
- Zhongxiang Xie
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China
| | - Jingtao Chen
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Chao Zheng
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China
| | - Jing Wu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yun Cheng
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China
| | - Shan Zhu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Chenhong Lin
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Qingqing Cao
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China
| | - Jie Zhu
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China.,Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
17
|
Currò D, Pugliese D, Armuzzi A. Frontiers in Drug Research and Development for Inflammatory Bowel Disease. Front Pharmacol 2017; 8:400. [PMID: 28690543 PMCID: PMC5481609 DOI: 10.3389/fphar.2017.00400] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 06/07/2017] [Indexed: 12/22/2022] Open
Abstract
Inflammatory bowel disease (IBD) is idiopathic, lifelong, immune-mediated diseases, for which curative therapies are not yet available. In the last 15 years, the introduction of monoclonal antibodies targeting tumor necrosis factor-α, a cytokine playing a key role in bowel inflammation, has revolutionized treatment paradigms for IBD. Despite their proven long-term efficacy, however, many patients do not respond or progressively lose response to these drugs. Major advances of knowledge in immunology and pathophysiology of intestinal inflammatory processes have made possible the identification of new molecular targets for drugs, thus opening several new potential therapeutic opportunities for IBD. The abnormal response of intestinal immunity to unknown antigens leads to the activation of T helper lymphocytes and triggers the inflammatory cascade. Sphingosine 1-phosphate receptor agonists negatively modulate the egress of lymphocytes, inducted by antigen-presenting cells, from secondary lymphoid tissues to intestinal wall. Leukocyte adhesion inhibitors (both anti-integrin and anti-Mucosal Vascular Addressin Cell Adhesion Molecule 1) interfere with the tissue homing processes. Activated T helper lymphocytes increase the levels of pro-inflammatory cytokines, such as interleukin 12, 23, and 6, offering several potential pharmacological interventions. The Janus kinases, intracellular enzymes mediating the transduction of several cytokine signals, are other explored targets for treating immune-mediated diseases. Finally, the impact of modulating Smad7 pathway, which is responsible for the down-regulation of the immunosuppressive cytokine transforming growth factor-β signaling, is currently under investigation. The purpose of this review is to discuss the most promising molecules in late-stage clinical development, with a special emphasis on pharmacological properties.
Collapse
Affiliation(s)
- Diego Currò
- Institute of Pharmacology, School of Medicine, Catholic University of the Sacred HeartRome, Italy
| | - Daniela Pugliese
- IBD Unit, Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario “A. Gemelli” Presidio Columbus, Catholic University of the Sacred HeartRome, Italy
| | - Alessandro Armuzzi
- IBD Unit, Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario “A. Gemelli” Presidio Columbus, Catholic University of the Sacred HeartRome, Italy
| |
Collapse
|
18
|
Qaqish A, Huang D, Chen CY, Zhang Z, Wang R, Li S, Yang E, Lu Y, Larsen MH, Jacobs WR, Qian L, Frencher J, Shen L, Chen ZW. Adoptive Transfer of Phosphoantigen-Specific γδ T Cell Subset Attenuates Mycobacterium tuberculosis Infection in Nonhuman Primates. THE JOURNAL OF IMMUNOLOGY 2017; 198:4753-4763. [PMID: 28526681 DOI: 10.4049/jimmunol.1602019] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 04/12/2017] [Indexed: 12/17/2022]
Abstract
The dominant Vγ2Vδ2 T cell subset recognizes phosphoantigen and exists only in humans and nonhuman primates. Despite the discovery of γδ T cells >30 y ago, a proof-of-concept study has not been done to prove the principle that the Vγ2Vδ2 T cell subset is protective against Mycobacterium tuberculosis and other infections. In this study, we used an adoptive cell-transfer strategy to define the protective role of Vγ2Vδ2 T cells in a primate tuberculosis (TB) model. Vγ2Vδ2 T cells for adoptive transfer displayed central/effector memory and mounted effector functions, including the production of anti-M. tuberculosis cytokines and inhibition of intracellular mycobacteria. They also expressed CXCR3/CCR5/LFA-1 trafficking/tissue-resident phenotypes and consistently trafficked to the airway, where they remained detectable from 6 h through 7 d after adoptive transfer. Interestingly, the test group of macaques receiving transfer of Vγ2Vδ2 T cells at weeks 1 and 3 after high-dose (500 CFU) M. tuberculosis infection exhibited significantly lower levels of M. tuberculosis infection burdens in lung lobes and extrapulmonary organs than did the control groups receiving PBLs or saline. Consistently, adoptive transfer of Vγ2Vδ2 T cells attenuated TB pathology and contained lesions primarily in the infection site of the right caudal lung lobe, with no or reduced TB dissemination to other lobes, spleen, or liver/kidney; in contrast, the controls showed widespread TB dissemination. The proof-of-concept finding supports the view that the dominant Vγ2Vδ2 T cell subset may be included in the rational design of a TB vaccine or host-directed therapy.
Collapse
Affiliation(s)
- Arwa Qaqish
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine Chicago, Chicago, IL 60612
| | - Dan Huang
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine Chicago, Chicago, IL 60612
| | - Crystal Y Chen
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine Chicago, Chicago, IL 60612
| | - Zhuoran Zhang
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine Chicago, Chicago, IL 60612
| | - Richard Wang
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine Chicago, Chicago, IL 60612
| | - Shengpu Li
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine Chicago, Chicago, IL 60612
| | - Enzhuoa Yang
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine Chicago, Chicago, IL 60612
| | - Yang Lu
- Department of Radiology, University of Illinois College of Medicine Chicago, Chicago, IL 60612; and
| | - Michelle H Larsen
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - William R Jacobs
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Lixia Qian
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine Chicago, Chicago, IL 60612
| | - James Frencher
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine Chicago, Chicago, IL 60612
| | - Ling Shen
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine Chicago, Chicago, IL 60612;
| | - Zheng W Chen
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine Chicago, Chicago, IL 60612;
| |
Collapse
|
19
|
Pflugfelder SC, Stern M, Zhang S, Shojaei A. LFA-1/ICAM-1 Interaction as a Therapeutic Target in Dry Eye Disease. J Ocul Pharmacol Ther 2016; 33:5-12. [PMID: 27906544 PMCID: PMC5240001 DOI: 10.1089/jop.2016.0105] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Dry eye disease (DED) is a common ocular disorder associated with inflammation of the lacrimal gland and ocular surface. The interaction of the integrin lymphocyte function-associated antigen-1 (LFA-1) with its cognate ligand intercellular adhesion molecule-1 (ICAM-1) is known to have important roles in the interaction of a variety of cells involved in immune responses and inflammation, including those prominent in ocular surface inflammation. Lifitegrast, an LFA-1 antagonist that blocks binding of ICAM-1 to LFA-1, has recently been approved in the United States for the treatment of signs and symptoms of DED. In this review, we evaluate research findings to explore the potential role of LFA-1/ICAM-1 interaction in the pathophysiology of DED, and the evidence supporting LFA-1/ICAM-1 interaction as a rational therapeutic target in DED. The results of our review suggest that LFA-1/ICAM-1 interaction may play important roles in the cell-mediated immune response and inflammation associated with DED, including facilitating the homing of dendritic cells to the lymph nodes, interaction of dendritic cells with T cells and subsequent T cell activation/differentiation, migration of activated CD4+ T cells from the lymph nodes to the ocular surface, reactivation of T cells by resident antigen-presenting cells at the ocular surface, and recruitment and retention of LFA-1-expressing T cells in the conjunctival epithelium. Based on the available evidence, inhibition of LFA-1/ICAM-1 interaction represents a rational targeted approach in treating DED. Notably, inhibition of LFA-1/ICAM-1 binding with lifitegrast offers a novel approach to reducing ocular surface inflammation in this condition.
Collapse
|
20
|
Heffernan DS, Monaghan SF, Ayala A. Lymphocyte integrin expression differences between SIRS and sepsis patients. Ir J Med Sci 2016; 186:981-987. [PMID: 27796667 DOI: 10.1007/s11845-016-1525-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 10/22/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND Systemic Inflammatory Response Syndrome (SIRS) and sepsis remain leading causes of death. Despite many similarities, the two entities are very distinct clinically and immunologically. T-Lymphocytes play a key pivotal role in the pathogenesis and ultimately outcome following both SIRS and sepsis. Integrins are essential in the trafficking and migration of lymphocytes. They also serve vital roles in efficient wound healing and clearance of infections. Here, we investigate whether integrin expression, specifically β1 (CD29) and β2 (CD18), are disrupted in SIRS and sepsis, and assess differences in integrin expression between these two critically ill clinical categories. METHODS T-Lymphocytes were isolated from whole blood collected from ICU patients exhibiting SIRS or sepsis. Samples were analyzed for CD18 (β2) and CD29 (β1) on CD3+ T cells through flow cytometry. Septic patients were stratified into either exclusively abdominal or non-abdominal sources of sepsis. RESULTS CD18 was almost ubiquitously expressed on CD3+ T cells irrespective of clinical condition. However, CD29 (β1 integrin) was lowest in SIRS patients (20.4% of CD3+ T cells) when compared with either septic patients (35.5%) or healthy volunteers (54.1%). Furthermore, there was evidence of compartmentalization in septic patients, where abdominal sources had a greater percentage of CD3+CD29+ T cells (41.7%) when compared with those with non-abdominal sources (29.5%). CONCLUSION Distinct differences in T-cell integrin expression exists between patients in SIRS versus sepsis, as well as relative to the source of sepsis. Further work is needed to understand cause and effect relative to the progression from SIRS into sepsis.
Collapse
Affiliation(s)
- D S Heffernan
- Division of Surgical Research, Department of Surgery, Warren Alpert Medical School of Brown UniversityRhode Island Hospital, 211 Aldrich Building, 593 Eddy Street, Providence, 02903, RI, USA.
| | - S F Monaghan
- Division of Surgical Research, Department of Surgery, Warren Alpert Medical School of Brown UniversityRhode Island Hospital, 211 Aldrich Building, 593 Eddy Street, Providence, 02903, RI, USA
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, Warren Alpert Medical School of Brown UniversityRhode Island Hospital, 211 Aldrich Building, 593 Eddy Street, Providence, 02903, RI, USA
| |
Collapse
|
21
|
Liao Z, Cantor JM. Endothelial Cells Require CD98 for Efficient Angiogenesis-Brief Report. Arterioscler Thromb Vasc Biol 2016; 36:2163-2166. [PMID: 27687603 DOI: 10.1161/atvbaha.116.308335] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 09/20/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE CD98 regulates integrin signaling and is critical for tumor cell proliferation. It is also expressed on endothelial cells (EC), but its role in angiogenesis is unclear. APPROACH AND RESULTS We used specific genetic targeting and antibody blockade approaches to examine the function of CD98 in EC proliferation, blood vessel growth, and tumor angiogenesis. It is upregulated on angiogenic ECs, and EC-specific deletion of CD98 in mice inhibited tumor growth, retinal angiogenesis, and EC proliferation. Reconstitution with CD98 mutants showed that integrin and CD98 interaction is necessary for EC survival and growth. Moreover, anti-CD98 treatment inhibited vessel formation and reversed EC-assisted tumor growth. CONCLUSIONS Our findings demonstrate a requirement for CD98 in EC growth and suggest that CD98-specific reagents could have a dual anticancer effect: directly by inhibiting tumor cell proliferation and indirectly by preventing tumor angiogenesis.
Collapse
Affiliation(s)
- Zhongji Liao
- From the Department of Medicine, University of California San Diego, La Jolla
| | - Joseph M Cantor
- From the Department of Medicine, University of California San Diego, La Jolla.
| |
Collapse
|
22
|
Verma NK, Fazil MHUT, Ong ST, Chalasani MLS, Low JH, Kottaiswamy A, P P, Kizhakeyil A, Kumar S, Panda AK, Freeley M, Smith SM, Boehm BO, Kelleher D. LFA-1/ICAM-1 Ligation in Human T Cells Promotes Th1 Polarization through a GSK3β Signaling–Dependent Notch Pathway. THE JOURNAL OF IMMUNOLOGY 2016; 197:108-18. [DOI: 10.4049/jimmunol.1501264] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 05/02/2016] [Indexed: 11/19/2022]
|
23
|
Iida T, Saito K, Katagiri K, Kinashi T, Ohta Y. The RacGAP protein FilGAP is a negative regulator of chemokine-promoted lymphocyte migration. FEBS Lett 2016; 590:1395-408. [PMID: 27130700 DOI: 10.1002/1873-3468.12189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 04/21/2016] [Accepted: 04/21/2016] [Indexed: 11/09/2022]
Abstract
Rho family small GTPases regulate lymphocyte migration induced by chemokines. However, how lymphocyte migration is regulated by Rho GTPases remains to be elucidated. Here, we identified FilGAP, a Rac-specific GAP, as a negative regulator of lymphocyte polarization and migration. Depletion of FilGAP in mouse pro-B BAF cells increased cellular elongation and membrane protrusion after stimulation of the cells with SDF-1α, which caused increased migration speed. Although FilGAP is detectable both at the front and rear of polarized cells, FilGAP appears to be concentrated at the tip of retracting lamellae of moving lymphocytes. Moreover, depletion of FilGAP increased activation of Rac at the front of polarized cells. Thus, FilGAP may inhibit lamellae extension at the front of moving lymphocytes.
Collapse
Affiliation(s)
- Toru Iida
- Division of Cell Biology, Department of Biosciences, School of Science, Kitasato University, Sagamihara, Kanagawa, Japan
| | - Koji Saito
- Division of Cell Biology, Department of Biosciences, School of Science, Kitasato University, Sagamihara, Kanagawa, Japan
| | - Koko Katagiri
- Division of Immunology, Department of Biosciences, School of Science, Kitasato University, Sagamihara, Kanagawa, Japan
| | - Tatsuo Kinashi
- Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka, Japan
| | - Yasutaka Ohta
- Division of Cell Biology, Department of Biosciences, School of Science, Kitasato University, Sagamihara, Kanagawa, Japan
| |
Collapse
|
24
|
Kim B, Nam S, Lim JH, Lim JS. NDRG2 Expression Decreases Tumor-Induced Osteoclast Differentiation by Down-regulating ICAM1 in Breast Cancer Cells. Biomol Ther (Seoul) 2016; 24:9-18. [PMID: 26759696 PMCID: PMC4703347 DOI: 10.4062/biomolther.2015.105] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 08/27/2015] [Accepted: 09/08/2015] [Indexed: 12/17/2022] Open
Abstract
Bone matrix is properly maintained by osteoclasts and osteoblasts. In the tumor microenvironment, osteoclasts are increasingly differentiated by the various ligands and cytokines secreted from the metastasized cancer cells at the bone metastasis niche. The activated osteoclasts generate osteolytic lesions. For this reason, studies focusing on the differentiation of osteoclasts are important to reduce bone destruction by tumor metastasis. The N-myc downstream-regulated gene 2 (NDRG2) has been known to contribute to the suppression of tumor growth and metastasis, but the precise role of NDRG2 in osteoclast differentiation induced by cancer cells has not been elucidated. In this study, we demonstrate that NDRG2 expression in breast cancer cells has an inhibitory effect on osteoclast differentiation. RAW 264.7 cells, which are monocytic preosteoclast cells, treated with the conditioned media (CM) of murine breast cancer cells (4T1) expressing NDRG2 are less differentiated into the multinucleated osteoclast-like cells than those treated with the CM of 4T1-WT or 4T1-mock cells. Interestingly, 4T1 cells stably expressing NDRG2 showed a decreased mRNA and protein level of intercellular adhesion molecule 1 (ICAM1), which is known to enhance osteoclast maturation. Osteoclast differentiation was also reduced by ICAM1 knockdown in 4T1 cells. In addition, blocking the interaction between soluble ICAM1 and ICAM1 receptors significantly decreased osteoclastogenesis of RAW 264.7 cells in the tumor environment. Collectively, these results suggest that the reduction of ICAM1 expression by NDRG2 in breast cancer cells decreases osteoclast differentiation, and demonstrate that excessive bone resorption could be inhibited via ICAM1 down-regulation by NDRG2 expression.
Collapse
Affiliation(s)
- Bomi Kim
- Department of Biological Science and the Research Center for Women's Disease, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Sorim Nam
- Department of Biological Science and the Research Center for Women's Disease, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Ji Hyun Lim
- Department of Biological Science and the Research Center for Women's Disease, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Jong-Seok Lim
- Department of Biological Science and the Research Center for Women's Disease, Sookmyung Women's University, Seoul 04310, Republic of Korea
| |
Collapse
|
25
|
Agarwal A, Afridi R, Hassan M, Sadiq MA, Sepah YJ, Do DV, Nguyen QD. Novel Therapies in Development for Diabetic Macular Edema. Curr Diab Rep 2015; 15:75. [PMID: 26294336 DOI: 10.1007/s11892-015-0652-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Diabetic macular edema (DME) secondary to diabetic retinopathy (DR) is a major cause for functional visual loss in the developed world. Laser photocoagulation has been used for decades in the treatment of DME. However, the advent of anti-vascular endothelial growth factor (anti-VEGF) has revolutionized the treatment of DME. Three important anti-VEGF agents whose efficacy has been well established via phase III clinical trials include ranibizumab, bevacizumab, and aflibercept. However, even in the era of anti-VEGF therapies, there are some challenges that retina specialists have to confront in managing patients with DME. These include the need for frequent treatment and an unpredictable response to therapy. There is evidence to suggest that pathways other than the VEGF pathway may be playing a role in the development of DME. Thus, extensive research is focused on development of novel agents that target these pathways. This review focuses on novel therapeutic agents in development, which may be used as a monotherapy or in combination with anti-VEGF agents, for the management of DME in the future.
Collapse
Affiliation(s)
- Aniruddha Agarwal
- Ocular Imaging Research and Reading Center (OIRRC), Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, USA
| | | | | | | | | | | | | |
Collapse
|