1
|
Dash UC, Nayak V, Navani HS, Samal RR, Agrawal P, Singh AK, Majhi S, Mogare DG, Duttaroy AK, Jena AB. Understanding the molecular bridges between the drugs and immune cell. Pharmacol Ther 2025; 267:108805. [PMID: 39908660 DOI: 10.1016/j.pharmthera.2025.108805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/11/2025] [Accepted: 01/21/2025] [Indexed: 02/07/2025]
Abstract
The interactions of drugs with the host's immune cells determine the drug's efficacy and adverse effects in patients. Nonsteroidal Anti-Inflammatory Drugs (NSAID), such as corticosteroids, NSAIDs, and immunosuppressants, affect the immune cells and alter the immune response. Molecularly, drugs can interact with immune cells via cell surface receptors, changing the antigen presentation by modifying the co-stimulatory molecules and interacting with the signaling pathways of T cells, B cells, Natural killer (NK) cells, mast cells, basophils, and macrophages. Immunotoxicity, resulting from drug-induced changes in redox status, generation of Reactive Oxygen Species (ROS)/Reactive Nitrogen Species (RNS), and alterations in antioxidant enzymes within immune cells, leads to immunodeficiency. This, in turn, causes allergic reactions, autoimmune diseases, and cytokine release syndrome (CRS). The treatment options should include the evaluation of immune status and utilization of the concept of pharmacogenomics to minimize the chances of immunotoxicity. Many strategies in redox, like targeting the redox pathway or using redox-active agents, are available for the modulation of the immune system and developing drugs. Case studies highlight significant drug-immune cell interactions and patient outcomes, underscoring the importance of understanding these complexities. The future direction focuses on the drugs to deliver antiviral therapy, new approaches to immunomodulation, and modern technologies for increasing antidote effects with reduced toxicity. In conclusion, in-depth knowledge of the interaction between drugs and immune cells is critical to protect the patient from the adverse effects of the drug and improve therapeutic outcomes of the treatment process. This review focuses on the multifaceted interactions of drugs and their consequences at the cellular levels of immune cells.
Collapse
Affiliation(s)
- Umesh Chandra Dash
- School of Biotechnology, Campus 11, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, Odisha 751024, India
| | - Vinayak Nayak
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, India
| | - Hiten Shanker Navani
- Biological Materials Laboratory, CSIR- Central Leather Research Institute, Adyar, Chennai 600020, India
| | - Rashmi Rekha Samal
- CSIR-Institute of Minerals & Materials Technology, Bhubaneswar 751 013, India
| | - Palak Agrawal
- Unit de Microbiologie Structurale, Institut Pasteur, Paris, France
| | - Anup Kumar Singh
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, India
| | - Sanatan Majhi
- Post Graduate Department of Biotechnology, Utkal University, Bhubaneswar 751004, Odisha, India
| | - Devraj Ganpat Mogare
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway.
| | - Atala Bihari Jena
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, India
| |
Collapse
|
2
|
Prosser AC, Klenerman P, Lucas M. Understanding Liver Transplantation Outcomes Through the Lens of Its Tissue-resident Immunobiome. Transplantation 2025:00007890-990000000-00973. [PMID: 39780303 DOI: 10.1097/tp.0000000000005303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Tissue-resident lymphocytes (TRLs) provide a front-line immunological defense mechanism uniquely placed to detect perturbations in tissue homeostasis. The heterogeneous TRL population spans the innate to adaptive immune continuum, with roles during normal physiology in homeostatic maintenance, tissue repair, pathogen detection, and rapid mounting of immune responses. TRLs are especially enriched in the liver, with every TRL subset represented, including liver-resident natural killer cells; tissue-resident memory B cells; conventional tissue-resident memory CD8, CD4, and regulatory T cells; and unconventional gamma-delta, natural killer, and mucosal-associated invariant T cells. The importance of donor- and recipient-derived TRLs after transplantation is becoming increasingly recognized, although it has not been examined in detail after liver transplantation. This review summarizes the evidence for the roles of TRLs in liver transplant immunology, focusing on their features, functions, and potential for their harnessing to improve transplant outcomes.
Collapse
Affiliation(s)
- Amy C Prosser
- Medical School, University of Western Australia, Perth, WA, Australia
| | - Paul Klenerman
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, United Kingdom
| | - Michaela Lucas
- Medical School, University of Western Australia, Perth, WA, Australia
- Department of Immunology, PathWest Laboratory Medicine, Perth, WA, Australia
- Department of Immunology, Sir Charles Gairdner Hospital, Perth, WA, Australia
- Department of Immunology, Perth Children's Hospital, Perth, WA, Australia
| |
Collapse
|
3
|
Higdon LE, Cooney LA, Serti E, Suwannasaen D, Muir VS, Wiedeman AE, Harris KM, Pardo J, Anderson MS, Speake C, Nepom GT, Linsley PS, Sanda S, Long SA. Early expansion of TIGIT+PD1+ effector memory CD4 T cells via agonistic effect of alefacept in new-onset type 1 diabetes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:12-22. [PMID: 40073269 PMCID: PMC11844141 DOI: 10.1093/jimmun/vkae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 10/28/2024] [Indexed: 03/14/2025]
Abstract
The CD2-depleting drug alefacept (LFA3-Ig) preserved beta cell function in new-onset type 1 diabetes (T1D) patients. The most promising biomarkers of response were late expansion of exhausted CD8 T cells and rare baseline inflammatory islet-reactive CD4 T cells, neither of which can be used to measure responses to drug in the weeks after treatment. Thus, we investigated whether early changes in T cell immunophenotypes could serve as biomarkers of drug activity. We characterized T cell responses by flow cytometry and identified an exhausted-like population of CD2low CD4 effector memory T cells coexpressing TIGIT and PD1 that expanded by 11 wk after the start of treatment. This population was not entirely spared from alefacept-mediated depletion in vivo or in vitro but recovered through homeostatic proliferation of CD2low cells in vivo. Proliferation of TIGIT+PD1+ effector memory CD4 T cells increased with treatment, with a concomitant reduction of proinflammatory cytokine production. The persistent increase of TIGIT+PD1+ effector memory CD4 T cells was specific to alefacept treatment; 2 other T cell depleting therapies, teplizumab and anti-thymocyte globulin, induced only a transient increase in this CD4 population. Our data suggest that the expanding TIGIT+PD1+ effector memory CD4 T cell population represents a promising biomarker of early treatment effects of alefacept. The nondepleting effects on proliferation and cytokine production also suggest agonistic activity by this CD2 targeted therapy.
Collapse
Affiliation(s)
- Lauren E Higdon
- Biomarker and Discovery Research, Immune Tolerance Network, San Francisco, CA, United States
| | - Laura A Cooney
- Biomarker and Discovery Research, Immune Tolerance Network, San Francisco, CA, United States
| | - Elisavet Serti
- Biomarker and Discovery Research, Immune Tolerance Network, San Francisco, CA, United States
| | - Duangchan Suwannasaen
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA, United States
| | - Virginia S Muir
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA, United States
| | - Alice E Wiedeman
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA, United States
| | - Kristina M Harris
- Biomarker and Discovery Research, Immune Tolerance Network, San Francisco, CA, United States
| | - Jorge Pardo
- Biomarker and Discovery Research, Immune Tolerance Network, San Francisco, CA, United States
| | - Mark S Anderson
- Biomarker and Discovery Research, Immune Tolerance Network, San Francisco, CA, United States
- Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
| | - Cate Speake
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA, United States
| | - Gerald T Nepom
- Biomarker and Discovery Research, Immune Tolerance Network, San Francisco, CA, United States
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA, United States
| | - Peter S Linsley
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA, United States
| | - Srinath Sanda
- Biomarker and Discovery Research, Immune Tolerance Network, San Francisco, CA, United States
| | - S Alice Long
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA, United States
| |
Collapse
|
4
|
Pratap Kashyap M, Mishra B, Sinha R, Jin L, Gou Y, Kumar N, Goliwas KF, Haque S, Deshane J, Berglund E, Berglund D, Elewski BE, Elmets CA, Athar M, Mukhtar MS, Raman C. CD2 expressing innate lymphoid and T cells are critical effectors of immunopathogenesis in hidradenitis suppurativa. Proc Natl Acad Sci U S A 2024; 121:e2409274121. [PMID: 39560648 PMCID: PMC11621750 DOI: 10.1073/pnas.2409274121] [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: 05/14/2024] [Accepted: 09/26/2024] [Indexed: 11/20/2024] Open
Abstract
Hidradenitis suppurativa (HS) is a chronic, debilitating inflammatory skin disease with a poorly understood immunopathogenesis. Here, we report that HS lesional skin is characterized by the expansion of innate lymphocytes and T cells expressing CD2, an essential activation receptor and adhesion molecule. Lymphocytes expressing elevated CD2 predominated with unique spatial distribution throughout the epidermis and hypodermis in the HS lesion. CD2+ cells were mainly innate lymphocytes expressing the NK cell marker, CD56, and CD4+ T cells. Importantly, these CD2+ cells interacted with CD58 (LFA3) expressing epidermal keratinocytes and fibroblasts in the hypodermis. Granzyme Abright NKT cells (CD2+CD3+CD56bright) clustered with α-SMA expressing fibroblasts juxtaposed to epithelialized tunnels and fibrotic regions of the hypodermis. Whereas NK cells (CD2+CD56dim) were perforin+, granzymes A+ and B+, and enriched adjacent to hyperplastic follicular epidermis and tunnels of HS showing presence of apoptotic cells. The cytokines IL-12, IL-15, and IL-18, which enhance NK cell maturation and function were significantly elevated in HS. Ex vivo HS skin explant cultures treated with CD2:CD58 interaction-blocking anti-CD2 monoclonal antibody attenuated secretion of inflammatory cytokines/chemokines and suppressed inflammatory gene signature. Additionally, CD2:CD58 blockade altered miRNAs involved in NK/NKT differentiation and/or function. In summary, we show that a cellular network of heterogenous NKT and NK cell populations drives inflammation and is critical in the pathobiology of HS, including tunnel formation and fibrosis. Finally, CD2 blockade is a viable immunotherapeutic approach for the effective management of HS.
Collapse
Affiliation(s)
- Mahendra Pratap Kashyap
- Center for Epigenomics and Translational Research in Inflammatory Skin Diseases, University of Alabama at Birmingham, Birmingham, AL35294
- Department of Dermatology at the University of Alabama at Birmingham, Birmingham, AL35294
| | - Bharat Mishra
- Department of Biology at the University of Alabama at Birmingham, Birmingham, AL35294
| | - Rajesh Sinha
- Center for Epigenomics and Translational Research in Inflammatory Skin Diseases, University of Alabama at Birmingham, Birmingham, AL35294
- Department of Dermatology at the University of Alabama at Birmingham, Birmingham, AL35294
| | - Lin Jin
- Center for Epigenomics and Translational Research in Inflammatory Skin Diseases, University of Alabama at Birmingham, Birmingham, AL35294
- Department of Dermatology at the University of Alabama at Birmingham, Birmingham, AL35294
| | - YiFei Gou
- Department of Biology at the University of Alabama at Birmingham, Birmingham, AL35294
| | - Nilesh Kumar
- Department of Biology at the University of Alabama at Birmingham, Birmingham, AL35294
| | - Kayla F. Goliwas
- Department of Medicine (Division of Pulmonary, Allergy and Critical Care Medicine) at the University of Alabama at Birmingham, Birmingham, AL35294
| | - Safiya Haque
- Department of Dermatology at the University of Alabama at Birmingham, Birmingham, AL35294
| | - Jessy Deshane
- Department of Medicine (Division of Pulmonary, Allergy and Critical Care Medicine) at the University of Alabama at Birmingham, Birmingham, AL35294
| | - Erik Berglund
- Department of Research and Development, ITB-MED AB, Stockholm113 68, Sweden
- Endocrine and Sarcoma Surgery Unit, Department of Molecular Medicine and Surgery, and Division of Transplantation Surgery, Karolinska Institute, Stockholm171 76, Sweden
- Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm171 77, Sweden
| | - David Berglund
- Department of Research and Development, ITB-MED AB, Stockholm113 68, Sweden
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala751 85, Sweden
| | - Boni E. Elewski
- Center for Epigenomics and Translational Research in Inflammatory Skin Diseases, University of Alabama at Birmingham, Birmingham, AL35294
- Department of Dermatology at the University of Alabama at Birmingham, Birmingham, AL35294
| | - Craig A. Elmets
- Center for Epigenomics and Translational Research in Inflammatory Skin Diseases, University of Alabama at Birmingham, Birmingham, AL35294
- Department of Dermatology at the University of Alabama at Birmingham, Birmingham, AL35294
| | - Mohammad Athar
- Center for Epigenomics and Translational Research in Inflammatory Skin Diseases, University of Alabama at Birmingham, Birmingham, AL35294
- Department of Dermatology at the University of Alabama at Birmingham, Birmingham, AL35294
| | - M. Shahid Mukhtar
- Department of Biology at the University of Alabama at Birmingham, Birmingham, AL35294
- Department of Genetics & Biochemistry at Clemson University, Clemson, SC29634
| | - Chander Raman
- Center for Epigenomics and Translational Research in Inflammatory Skin Diseases, University of Alabama at Birmingham, Birmingham, AL35294
- Department of Dermatology at the University of Alabama at Birmingham, Birmingham, AL35294
| |
Collapse
|
5
|
Burke GW, Mitrofanova A, Fontanella AM, Vendrame F, Ciancio G, Vianna RM, Roth D, Ruiz P, Abitbol CL, Chandar J, Merscher S, Pugliese A, Fornoni A. Transplantation: platform to study recurrence of disease. Front Immunol 2024; 15:1354101. [PMID: 38495894 PMCID: PMC10940352 DOI: 10.3389/fimmu.2024.1354101] [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: 12/11/2023] [Accepted: 01/29/2024] [Indexed: 03/19/2024] Open
Abstract
Beyond the direct benefit that a transplanted organ provides to an individual recipient, the study of the transplant process has the potential to create a better understanding of the pathogenesis, etiology, progression and possible therapy for recurrence of disease after transplantation while at the same time providing insight into the original disease. Specific examples of this include: 1) recurrence of focal segmental glomerulosclerosis (FSGS) after kidney transplantation, 2) recurrent autoimmunity after pancreas transplantation, and 3) recurrence of disease after orthotopic liver transplantation (OLT) for cirrhosis related to progressive steatosis secondary to jejuno-ileal bypass (JIB) surgery. Our team has been studying these phenomena and their immunologic underpinnings, and we suggest that expanding the concept to other pathologic processes and/or transplanted organs that harbor the risk for recurrent disease may provide novel insight into the pathogenesis of a host of other disease processes that lead to organ failure.
Collapse
Affiliation(s)
- George William Burke
- Division of Kidney-Pancreas Transplantation, Department of Surgery, Miami Transplant Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Alla Mitrofanova
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | | | - Francesco Vendrame
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Gaetano Ciancio
- Division of Kidney-Pancreas Transplantation, Department of Surgery, Miami Transplant Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Rodrigo M. Vianna
- Department of Surgery, Miami Transplant Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - David Roth
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Phillip Ruiz
- Transplant Pathology, Immunology and Histocompatibility Laboratory University of Miami Department of Surgery, Miami Transplant Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Carolyn L. Abitbol
- Pediatric Nephrology & Hypertension, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Jayanthi Chandar
- Pediatric Kidney Transplant, Miami Transplant Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
- Peggy and Harold Katz Family Drug Discovery Center, Department of Medicine, University of Miami - Miller School of Medicine, Miami, FL, United States
| | - Alberto Pugliese
- Department of Diabetes Immunology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, United States
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
- Peggy and Harold Katz Family Drug Discovery Center, Department of Medicine, University of Miami - Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
6
|
Abstract
Memory T cells that are specific for alloantigen can arise from a variety of stimuli, ranging from direct allogeneic sensitization from prior transplantation, blood transfusion, or pregnancy to the elicitation of pathogen-specific T cells that are cross-reactive with alloantigen. Regardless of the mechanism by which they arise, alloreactive memory T cells possess key metabolic, phenotypic, and functional properties that render them distinct from naive T cells. These properties affect the immune response to transplantation in 2 important ways: first, they can alter the speed, location, and effector mechanisms with which alloreactive T cells mediate allograft rejection, and second, they can alter T-cell susceptibility to immunosuppression. In this review, we discuss recent developments in understanding these properties of memory T cells and their implications for transplantation.
Collapse
Affiliation(s)
| | - Mandy L. Ford
- Emory Transplant Center, Emory University, Atlanta, GA
| |
Collapse
|
7
|
Mengrelis K, Muckenhuber M, Wekerle T. Chimerism-based Tolerance Induction in Clinical Transplantation: Its Foundations and Mechanisms. Transplantation 2023; 107:2473-2485. [PMID: 37046378 DOI: 10.1097/tp.0000000000004589] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Hematopoietic chimerism remains the most promising strategy to bring transplantation tolerance into clinical routine. The concept of chimerism-based tolerance aims to extend the recipient's mechanisms of self-tolerance (ie, clonal deletion, anergy, and regulation) to include the tolerization of donor antigens that are introduced through the cotransplantation of donor hematopoietic cells. For this to be successful, donor hematopoietic cells need to engraft in the recipient at least temporarily. Three pioneering clinical trials inducing chimerism-based tolerance in kidney transplantation have been published to date. Within this review, we discuss the mechanisms of tolerance that are associated with the specific therapeutic protocols of each trial. Recent data highlight the importance of regulation as a mechanism that maintains tolerance. Insufficient regulatory mechanisms are also a likely explanation for situations of tolerance failure despite persisting donor chimerism. After decades of preclinical development of chimerism protocols, mechanistic data from clinical trials have recently become increasingly important. Better understanding of the required mechanisms for tolerance to be induced in humans will be a key to design more reliable and less invasive chimerism protocols in the future.
Collapse
Affiliation(s)
- Konstantinos Mengrelis
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | | | | |
Collapse
|
8
|
Cvetkovski F, Razavi R, Sellberg F, Berglund E, Berglund D. Siplizumab combination therapy with belatacept or abatacept broadly inhibits human T cell alloreactivity in vitro. Am J Transplant 2023; 23:1603-1611. [PMID: 37270108 DOI: 10.1016/j.ajt.2023.05.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/09/2023] [Accepted: 05/25/2023] [Indexed: 06/05/2023]
Abstract
Combined antigen-specific T cell receptor stimulation and costimulation are needed for complete T cell activation. Belatacept and abatacept are nondepleting fusion proteins blocking CD28/B7 costimulation, whereas siplizumab is a depleting antiCD2 immunoglobulin G1 monoclonal antibody targeting CD2/CD58 costimulation. Herein, the effect of siplizumab combination therapy with abatacept or belatacept on T cell alloreactivity in mixed lymphocyte reactions was investigated. In contrast to monotherapy, the combination of siplizumab with belatacept or abatacept induced near-complete suppression of T cell proliferation and increased the potency of siplizumab-mediated T cell inhibition. Furthermore, dual targeting of CD2 and CD28 costimulation enhanced the selective depletion of memory T cells compared with monotherapy. Although siplizumab monotherapy leads to significant regulatory T cell enrichment, high doses of cytotoxic T-lymphocyte-associated antigen 4 and a human IgG1 Fc fragment in the combination therapy reduced this effect. These results support the clinical evaluation of dual costimulation blockade, combining siplizumab with abatacept or belatacept, for the prophylaxis of organ transplant rejection and improvement of long-term outcomes following transplantation. Ongoing investigative research will elucidate when other forms of siplizumab-based dual costimulatory blockade may be able to induce similarly strong inhibition of T cell activation although still allowing for enrichment of regulatory T cells.
Collapse
Affiliation(s)
- Filip Cvetkovski
- Research and Development, ITB-MED AB, Stockholm, Sweden; Endocrine and Sarcoma Surgery Unit, Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Ronia Razavi
- Research and Development, ITB-MED AB, Stockholm, Sweden
| | - Felix Sellberg
- Research and Development, ITB-MED AB, Stockholm, Sweden; Department of Immunology, Genetics and Pathology, Section of Clinical Immunology, Uppsala University, Sweden
| | - Erik Berglund
- Research and Development, ITB-MED AB, Stockholm, Sweden; Endocrine and Sarcoma Surgery Unit, Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden; Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden
| | - David Berglund
- Research and Development, ITB-MED AB, Stockholm, Sweden; Department of Immunology, Genetics and Pathology, Section of Clinical Immunology, Uppsala University, Sweden.
| |
Collapse
|
9
|
Zhang Q, Liu J, Yao D, Shi JX, Liu YJ, Wei YG, Guo S. Comprehensive Analysis to Identify Rh Family C Glycoprotein ( RHCG) as the Causative Gene for Psoriasis and Search for Alternative Treatment Modalities. Drug Des Devel Ther 2023; 17:2593-2611. [PMID: 37664450 PMCID: PMC10473404 DOI: 10.2147/dddt.s421300] [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: 05/30/2023] [Accepted: 08/17/2023] [Indexed: 09/05/2023] Open
Abstract
Background Psoriasis is a complex autoimmune disease. Frequent interactions between epidermal and immune cells are likely to be responsible for the strong heterogeneity of psoriasis. Therefore, our work aims to build on current knowledge and further search for new molecular mechanisms related to psoriasis pathogenesis in order to develop new targeted drugs. Methods Data from psoriasis samples were obtained from the Gene Expression Omnibus (GEO) database, and batch effects were corrected using the "Combat" algorithm in the "SVA" package. Functional annotation of differential genes in psoriasis was performed by Gene set enrichment analysis (GSEA). Core functional modules were identified using the Multiscale Embedded Gene Co-Expression Network Analysis (MEGENA) algorithm for selection from the differential gene interaction network. The expression and potential function of Rh Family C Glycoprotein (RHCG) was predicted in single cell data by the "Seurat" package and validated in psoriasis samples by multiplex immunofluorescence. In addition, the regulatory function of HOP Homeobox (HOPX) on RHCG in keratinocytes was confirmed using RNA interference. Using immune infiltration analysis, RHCG and DC cells were analyzed for their association. Finally, the molecular mechanisms of treatment of psoriasis using Tripterygii Radix (TR) and Cinnamomi Ramulus (CR) were explored through network pharmacology and experimental validation. Results Immune response (represented by C1_2) and collagen matrix formation (represented by C1_3) were identified as two important pathogenic factors in psoriasis and helped to define new biological subtypes of psoriasis. One important psoriasis hub gene, RHCG, was obtained and found to be closely associated with keratinocyte differentiation as well as DC cell maturation. And RHCG was regulated by HOPX in keratinocytes. In addition, the mechanism of action of CR and TR in the treatment of psoriasis was tentatively confirmed to be related to TRPV3, NFKB2, and YAP1. Conclusions Our study identifies a new causal disease gene (RHCG) and offers potential alternatives for the treatment of psoriasis.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Dermatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, People’s Republic of China
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
| | - Jia Liu
- Department of Dermatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, People’s Republic of China
| | - Dan Yao
- Department of Dermatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, People’s Republic of China
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
| | - Jian-Xin Shi
- Department of Dermatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, People’s Republic of China
| | - Yuan-Jie Liu
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, People’s Republic of China
- Key Laboratory of Tumor System Biology of Traditional Chinese Medicine, Nanjing, Jiangsu, 210029, People’s Republic of China
| | - Yue-Gang Wei
- Department of Dermatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, People’s Republic of China
| | - Shun Guo
- Department of Dermatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, People’s Republic of China
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
| |
Collapse
|
10
|
Regulatory T cell homeostasis: Requisite signals and implications for clinical development of biologics. Clin Immunol 2023; 246:109201. [PMID: 36470337 DOI: 10.1016/j.clim.2022.109201] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/28/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
Novel biologics are currently being tested in clinical trials for the treatment of autoimmune diseases and the prevention of transplant allograft rejection. Their premise is to deliver highly efficient immunosuppression while minimizing side-effects, as they specifically target inflammatory mediators involved in the dysregulation of the immune system. However, the pleiotropism of soluble mediators and cell-to-cell interactions with potential to exert both proinflammatory and regulatory influences on the outcome of the immune response can lead to unpredictable results. Predicting responses to biologic drugs requires mechanistic understanding of the cell type-specific effect of immune mediators. Elucidation of the central role of regulatory T cells (Treg), a small subset of T cells dedicated to immune homeostasis, in preventing the development of auto- and allo-immunity has provided a deeper understanding of the signaling pathways that govern immune tolerance. This review focuses on the requisite signals that promote Treg homeostasis and discusses the anticipated outcomes of biologics targeting these signals. Our goal is to inform and facilitate the design of cell-specific biologics that thwart T effector cells (Teff) while promoting Treg function for the treatment of autoimmune diseases and the prevention of transplant rejection.
Collapse
|
11
|
Li B, Lu Y, Zhong MC, Qian J, Li R, Davidson D, Tang Z, Zhu K, Argenty J, de Peredo AG, Malissen B, Roncagalli R, Veillette A. Cis interactions between CD2 and its ligands on T cells are required for T cell activation. Sci Immunol 2022; 7:eabn6373. [PMID: 35930657 DOI: 10.1126/sciimmunol.abn6373] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
CD2 is largely described to promote T cell activation when engaged by its ligands, CD48 in mice and CD58 in humans, that are present on antigen-presenting cells (APCs). However, both CD48 and CD58 are also expressed on T cells. By generating new knockout mouse strains lacking CD2 or CD48 in the C57BL/6 background, we determined that whereas CD2 was necessary on T cells for T cell activation, its ligand CD48 was not required on APCs. Rather, CD48 was also needed on T cells. One exception was during cytotoxicity, which required CD48 on T cells and APCs. Fluorescence resonance energy transfer (FRET) studies in nonimmune cells provided evidence that cis interactions between CD2 and CD48 existed within individual cells. CD2-CD48 interactions on T cells enabled more robust T cell receptor (TCR) signals, including protein tyrosine phosphorylation. Using T cells from a CD2 knock-in mouse in which a tag was inserted at the carboxyl terminus of CD2, mass spectrometry analyses revealed that the role of CD2 in T cell activation correlated with its ability to interact with components of the TCR complex and the protein tyrosine kinase Lck. CD2-CD58 provided a similar function in human T cells. Thus, our data imply that T cell-intrinsic cis interactions of CD2 with its ligands are required for TCR signaling and T cell activation. Interactions with ligands on APCs contribute during cytotoxicity.
Collapse
Affiliation(s)
- Bin Li
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada.,Molecular Biology Program, University of Montréal, Montréal, Québec H3T 1J4, Canada
| | - Yan Lu
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Ming-Chao Zhong
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Jin Qian
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Rui Li
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada.,Department of Medicine, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Dominique Davidson
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Zhenghai Tang
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Kaiwen Zhu
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada.,Department of Medicine, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Jérémy Argenty
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, 13288 Marseille, France
| | - Anne Gonzalez de Peredo
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS UPS, Toulouse, France
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, 13288 Marseille, France.,Centre d'Immunophénomique, Aix Marseille Université, INSERM, CNRS, 13288 Marseille, France
| | - Romain Roncagalli
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, 13288 Marseille, France
| | - André Veillette
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada.,Molecular Biology Program, University of Montréal, Montréal, Québec H3T 1J4, Canada.,Department of Medicine, McGill University, Montréal, Québec H3G 1Y6, Canada
| |
Collapse
|
12
|
Podestà MA, Sykes M. Chimerism-Based Tolerance to Kidney Allografts in Humans: Novel Insights and Future Perspectives. Front Immunol 2022; 12:791725. [PMID: 35069574 PMCID: PMC8767096 DOI: 10.3389/fimmu.2021.791725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/15/2021] [Indexed: 11/18/2022] Open
Abstract
Chronic rejection and immunosuppression-related toxicity severely affect long-term outcomes of kidney transplantation. The induction of transplantation tolerance – the lack of destructive immune responses to a transplanted organ in the absence of immunosuppression – could potentially overcome these limitations. Immune tolerance to kidney allografts from living donors has been successfully achieved in humans through clinical protocols based on chimerism induction with hematopoietic cell transplantation after non-myeloablative conditioning. Notably, two of these protocols have led to immune tolerance in a significant fraction of HLA-mismatched donor-recipient combinations, which represent the large majority of cases in clinical practice. Studies in mice and large animals have been critical in dissecting tolerance mechanisms and in selecting the most promising approaches for human translation. However, there are several key differences in tolerance induction between these models and humans, including the rate of success and stability of donor chimerism, as well as the relative contribution of different mechanisms in inducing donor-specific unresponsiveness. Kidney allograft tolerance achieved through durable full-donor chimerism may be due to central deletion of graft-reactive donor T cells, even though mechanistic data from patient series are lacking. On the other hand, immune tolerance attained with transient mixed chimerism-based protocols initially relies on Treg-mediated suppression, followed by peripheral deletion of donor-reactive recipient T-cell clones under antigenic pressure from the graft. These conclusions were supported by data deriving from novel high-throughput T-cell receptor sequencing approaches that allowed tracking of alloreactive repertoires over time. In this review, we summarize the most important mechanistic studies on tolerance induction with combined kidney-bone marrow transplantation in humans, discussing open issues that still need to be addressed and focusing on techniques developed in recent years to efficiently monitor the alloresponse in tolerance trials. These cutting-edge methods will be instrumental for the development of immune tolerance protocols with improved efficacy and to identify patients amenable to safe immunosuppression withdrawal.
Collapse
Affiliation(s)
- Manuel Alfredo Podestà
- Renal Division, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milano, Italy
| | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Department of Surgery, Department of Microbiology and Immunology, Columbia University, New York, NY, United States
| |
Collapse
|
13
|
Immunosuppression in the Age of Precision Medicine. Semin Nephrol 2022; 42:86-98. [DOI: 10.1016/j.semnephrol.2022.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
14
|
Fernandez Lahore G, Förster M, Johannesson M, Sabatier P, Lönnblom E, Aoun M, He Y, Nandakumar KS, Zubarev RA, Holmdahl R. Polymorphic estrogen receptor binding site causes Cd2-dependent sex bias in the susceptibility to autoimmune diseases. Nat Commun 2021; 12:5565. [PMID: 34552089 PMCID: PMC8458462 DOI: 10.1038/s41467-021-25828-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/20/2021] [Indexed: 01/22/2023] Open
Abstract
Complex autoimmune diseases are sexually dimorphic. An interplay between predisposing genetics and sex-related factors probably controls the sex discrepancy in the immune response, but the underlying mechanisms are unclear. Here we positionally identify a polymorphic estrogen receptor binding site that regulates Cd2 expression, leading to female-specific differences in T cell-dependent mouse models of autoimmunity. Female mice with reduced Cd2 expression have impaired autoreactive T cell responses. T cells lacking Cd2 costimulation upregulate inhibitory Lag-3. These findings help explain sexual dimorphism in human autoimmunity, as we find that CD2 polymorphisms are associated with rheumatoid arthritis and 17-β-estradiol-regulation of CD2 is conserved in human T cells. Hormonal regulation of CD2 might have implications for CD2-targeted therapy, as anti-Cd2 treatment more potently affects T cells in female mice. These results demonstrate the relevance of sex-genotype interactions, providing strong evidence for CD2 as a sex-sensitive predisposing factor in autoimmunity.
Collapse
Affiliation(s)
- Gonzalo Fernandez Lahore
- Division Medical Inflammation Research, Dept. Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Michael Förster
- Division Medical Inflammation Research, Dept. Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Martina Johannesson
- Division Medical Inflammation Research, Dept. Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institute, Karolinska University Hospital, SE-171 76, Stockholm, Sweden
| | - Pierre Sabatier
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Erik Lönnblom
- Division Medical Inflammation Research, Dept. Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Mike Aoun
- Division Medical Inflammation Research, Dept. Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Yibo He
- Division Medical Inflammation Research, Dept. Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Kutty Selva Nandakumar
- Division Medical Inflammation Research, Dept. Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
- SMU-KI United Medical Inflammation Centre, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Roman A Zubarev
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
- Department of Pharmacological & Technological Chemistry, I.M. Sechenov First Moscow State Medical University, Moscow, 119146, Russia
| | - Rikard Holmdahl
- Division Medical Inflammation Research, Dept. Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden.
- The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), 710004, Xi'an, China.
| |
Collapse
|
15
|
Strategies for Liver Transplantation Tolerance. Int J Mol Sci 2021; 22:ijms22052253. [PMID: 33668238 PMCID: PMC7956766 DOI: 10.3390/ijms22052253] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/19/2021] [Accepted: 02/21/2021] [Indexed: 12/13/2022] Open
Abstract
Liver transplant (LT) recipients require life-long immunosuppression (IS) therapy to preserve allograft function. The risks of chronic IS include an increased frequency of malignancy, infection, renal impairment, and other systemic toxicities. Despite advances in IS, long-term LT outcomes have not been improved over the past three decades. Standard-of-care (SoC) therapy can, in rare cases, lead to development of operational tolerance that permits safe withdrawal of maintenance IS. However, successful IS withdrawal cannot be reliably predicted and, in current prospective studies, is attempted several years after the transplant procedure, after considerable exposure to the cumulative burden of maintenance therapy. A recent pilot clinical trial in liver tolerance induction demonstrated that peri-transplant immunomodulation, using a regulatory T-cell (Treg) approach, can reduce donor-specific alloreactivity and allow early IS withdrawal. Herein we review protocols for active tolerance induction in liver transplantation, with a focus on identifying tolerogenic cell populations, as well as barriers to tolerance. In addition, we propose the use of novel IS agents to promote immunomodulatory mechanisms favoring tolerance. With numerous IS withdrawal trials underway, improved monitoring and use of novel immunomodulatory strategies will help provide the necessary knowledge to establish an active liver tolerance induction protocol for widespread use.
Collapse
|
16
|
Binder C, Sellberg F, Cvetkovski F, Berg S, Berglund E, Berglund D. Siplizumab Induces NK Cell Fratricide Through Antibody-Dependent Cell-Mediated Cytotoxicity. Front Immunol 2021; 12:599526. [PMID: 33643309 PMCID: PMC7904868 DOI: 10.3389/fimmu.2021.599526] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 01/05/2021] [Indexed: 12/17/2022] Open
Abstract
The glycoprotein CD2 is expressed on T and NK cells and contributes to cell-cell conjugation, agonistic signaling and actin cytoskeleton rearrangement. CD2 has previously been shown to have an important function in natural NK cell cytotoxicity but to be expendable in antibody-mediated cytotoxicity. Siplizumab is a monoclonal anti-CD2 IgG1 antibody that is currently undergoing clinical trials in the field of transplantation. This study investigated the effect of CD2 binding and Fc γ receptor binding by siplizumab (Fc-active) and Fc-silent anti-CD2 monoclonal antibodies in allogeneic mixed lymphocyte reaction and autologous lymphocyte culture. Further, induction of NK cell fratricide and inhibition of natural cytotoxicity as well as antibody-dependent cytotoxicity by these agents were assessed. Blockade of CD2 via monoclonal antibodies in the absence of Fc γ receptor binding inhibited NK cell activation in allogeneic mixed lymphocyte reaction. In contrast, siplizumab increased NK cell activation in both mixed lymphocyte reaction and autologous lymphocyte culture due to FcγRIIIA binding. However, experiments using purified NK cells did not show an inhibitory effect of CD2 blockade on natural cytotoxicity or antibody-dependent cytotoxicity. Lastly, it was shown that siplizumab induces NK cell fratricide. Concluding, siplizumab is a promising biopharmaceutical drug candidate for depletion of T and NK cells with minimal off-target effects.
Collapse
Affiliation(s)
- Christian Binder
- Department of Immunology, Genetics and Pathology, Section of Clinical Immunology, Uppsala University, Uppsala, Sweden
- Research and Development, ITB-Med AB, Stockholm, Sweden
| | - Felix Sellberg
- Department of Immunology, Genetics and Pathology, Section of Clinical Immunology, Uppsala University, Uppsala, Sweden
- Research and Development, ITB-Med AB, Stockholm, Sweden
| | | | - Stefan Berg
- Research and Development, ITB-Med AB, Stockholm, Sweden
| | - Erik Berglund
- Research and Development, ITB-Med AB, Stockholm, Sweden
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - David Berglund
- Department of Immunology, Genetics and Pathology, Section of Clinical Immunology, Uppsala University, Uppsala, Sweden
- Research and Development, ITB-Med AB, Stockholm, Sweden
| |
Collapse
|