1
|
Zhong J, Zhang W, Zhang L, Li J, Kang L, Li X. CircFLNA/miR-214 modulates regulatory T cells by regulating PD-1 in acute lung injury induced by sepsis. Autoimmunity 2023; 56:2259131. [PMID: 37724530 DOI: 10.1080/08916934.2023.2259131] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 09/10/2023] [Indexed: 09/21/2023]
Abstract
Sepsis-induced acute respiratory distress syndrome (ARDS) remains a major complication of death from bacterial infection. Regulatory T cells (Tregs) are important regulators in addressing lung injury. Considering the extensive research of circular RNAs (circRNAs), the role of circRNA in Treg modulation during ARDS remains unclear. In this study, patients with sepsis-induced ARDS along with non-ARDS controls were obtained, and bronchoalveolar lavage fluid (BALF) was collected as clinical samples. Additionally, cecal ligation and puncture (CLP) was performed to construct a septic ARDS model, and lung tissues as well as peripheral blood were collected. mRNA expressions were measured by RT-qPCR. ELISA was carried out to measure the concentration of inflammatory factors. A combination of online bioinformatics, dual-luciferase reporter, and RND pull-down assays was performed to verify interactions between microRNA (miRNA) and circRNA/mRNA. Tregs were measured by flow cytometry. Our data suggested that circFLNA was aberrantly elevated in ARDS, and depletion of circFLNA upregulated CD4+CD25+Foxp3+ Tregs and decreased inflammatory response. Additionally, miR-214-5p which binds with circFLNA, reversed circFLNA-induced effects in ARDS. Programmed cell death protein 1 (PD-1) is a downstream target gene of miR-214-5p, and abrogated the effects of miR-214-5p on regulating CD4+CD25+Foxp3+ Tregs and inflammatory response. In a word, circFLNA/miR-214-5p/PD-1 signaling is a novel pathway that modulates Tregs in ARDS.
Collapse
Affiliation(s)
- Jian Zhong
- Department of Emergency, Dongguan Tungwah Hospital, Dongguan, Guangdong, China
| | - Wei Zhang
- Department of Emergency, Dongguan Tungwah Hospital, Dongguan, Guangdong, China
| | - Leiyun Zhang
- Department of Emergency, Dongguan Tungwah Hospital, Dongguan, Guangdong, China
| | - Jieying Li
- Department of Emergency, Dongguan Tungwah Hospital, Dongguan, Guangdong, China
| | - Lingkai Kang
- Department of Critical Care Medicine, The Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, China
| | - Xiaoyue Li
- Department of Critical Care Medicine, The Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, China
| |
Collapse
|
2
|
Wei C, Sun Y, Zeng F, Chen X, Ma L, Liu X, Qi X, Shi W, Gao H. Exosomal miR-181d-5p Derived from Rapamycin-Conditioned MDSC Alleviated Allograft Rejection by Targeting KLF6. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304922. [PMID: 37870185 PMCID: PMC10700181 DOI: 10.1002/advs.202304922] [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: 07/19/2023] [Revised: 09/18/2023] [Indexed: 10/24/2023]
Abstract
Immune rejection and side effects of long-term administration of immunosuppressants are the two major obstacles to allograft acceptance and tolerance. The immunosuppressive extracellular vesicles (EVs)-based approach has been proven to be effective in treating autoimmune/inflammatory disorders. Herein, the anti-rejection advantage of exosomes (Rapa-Exo) from rapamycin-conditioned myeloid-derived suppressor cells (MDSCs) over exosomes (Exo-Nor) from the untreated MDSCs is shown. The exosomal small RNA sequencing and loss-of-function assays reveal that the anti-rejection effect of Rapa-Exo functionally relies on miR-181d-5p. Through target prediction and double-luciferase reporter assay, Kruppel-like factor (KLF) 6 is identified as a direct target of miR-181d-5p. Finally, KLF6 knockdown markedly resolves inflammation and prolongs the survival of corneal allografts. Taken together, these findings support that Rapa-Exo executes an anti-rejection effect, highlighting the immunosuppressive EVs-based treatment as a promising approach in organ transplantation.
Collapse
Affiliation(s)
- Chao Wei
- State Key Laboratory Cultivation BaseShandong Provincial Key Laboratory of OphthalmologyEye Institute of Shandong First Medical UniversityQingdao266071China
| | - Yaru Sun
- State Key Laboratory Cultivation BaseShandong Provincial Key Laboratory of OphthalmologyEye Institute of Shandong First Medical UniversityQingdao266071China
| | - Fanxing Zeng
- State Key Laboratory Cultivation BaseShandong Provincial Key Laboratory of OphthalmologyEye Institute of Shandong First Medical UniversityQingdao266071China
| | - Xiunian Chen
- State Key Laboratory Cultivation BaseShandong Provincial Key Laboratory of OphthalmologyEye Institute of Shandong First Medical UniversityQingdao266071China
| | - Li Ma
- State Key Laboratory Cultivation BaseShandong Provincial Key Laboratory of OphthalmologyEye Institute of Shandong First Medical UniversityQingdao266071China
| | - Xiaoxue Liu
- State Key Laboratory Cultivation BaseShandong Provincial Key Laboratory of OphthalmologyEye Institute of Shandong First Medical UniversityQingdao266071China
| | - Xiaolin Qi
- State Key Laboratory Cultivation BaseShandong Provincial Key Laboratory of OphthalmologyEye Institute of Shandong First Medical UniversityQingdao266071China
- Eye Hospital of Shandong First Medical University (Shandong Eye Hospital)Jinan250117China
- School of OphthalmologyShandong First Medical University & Shandong Academy of Medical ScienceJinan250117China
| | - Weiyun Shi
- State Key Laboratory Cultivation BaseShandong Provincial Key Laboratory of OphthalmologyEye Institute of Shandong First Medical UniversityQingdao266071China
- Eye Hospital of Shandong First Medical University (Shandong Eye Hospital)Jinan250117China
- School of OphthalmologyShandong First Medical University & Shandong Academy of Medical ScienceJinan250117China
| | - Hua Gao
- State Key Laboratory Cultivation BaseShandong Provincial Key Laboratory of OphthalmologyEye Institute of Shandong First Medical UniversityQingdao266071China
- Eye Hospital of Shandong First Medical University (Shandong Eye Hospital)Jinan250117China
- School of OphthalmologyShandong First Medical University & Shandong Academy of Medical ScienceJinan250117China
| |
Collapse
|
3
|
Azim S, Zubair H, Rousselle T, McDaniels JM, Shetty AC, Kuscu C, Kuscu C, Talwar M, Eason JD, Maluf DG, Mas VR. Single-cell RNA sequencing reveals peripheral blood mononuclear immune cell landscape associated with operational tolerance in a kidney transplant recipient. Am J Transplant 2023; 23:1434-1445. [PMID: 37201755 PMCID: PMC10527369 DOI: 10.1016/j.ajt.2023.04.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/29/2023] [Accepted: 04/15/2023] [Indexed: 05/20/2023]
Abstract
Operational tolerance (OT) after kidney transplantation is defined as stable graft acceptance without the need for immunosuppression therapy. However, it is not clear which cellular and molecular pathways are driving tolerance in these patients. In this first-of-its-kind pilot study, we assessed the immune landscape associated with OT using single-cell analyses. Peripheral mononuclear cells from a kidney transplant recipient with OT (Tol), 2 healthy individuals (HC), and a kidney transplant recipient with normal kidney function on standard-of-care immunosuppression (SOC) were evaluated. The immune landscape of the Tol was drastically different from that of SOC and emerged closer to the profile of HC. TCL1A+ naive B cells and LSGAL1+ regulatory T cells (Tregs) were in higher proportions in Tol. We were unable to identify the Treg subcluster in SOC. The ligand-receptor analysis in HC and Tol identified interactions between B cells, and Tregs that enhance the proliferation and suppressive function of Tregs. SOC reported the highest proportion of activated B cells with more cells in the G2M phase. Our single-cell RNA sequencing study identified the mediators of tolerance; however, it emphasizes the requirement of similar investigations on a larger cohort to reaffirm the role of immune cells in tolerance.
Collapse
Affiliation(s)
- Shafquat Azim
- Surgical Sciences Division, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Haseeb Zubair
- Surgical Sciences Division, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Thomas Rousselle
- Surgical Sciences Division, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jennifer M McDaniels
- Surgical Sciences Division, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Amol C Shetty
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Cem Kuscu
- Department of Surgery, Transplant Research Institute, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Canan Kuscu
- Department of Surgery, Transplant Research Institute, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Manish Talwar
- James D. Eason Transplant Institute, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - James D Eason
- James D. Eason Transplant Institute, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Daniel G Maluf
- Program in Transplantation, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Valeria R Mas
- Surgical Sciences Division, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
4
|
Kim S, Shukla RK, Yu H, Baek A, Cressman SG, Golconda S, Lee GE, Choi H, Reneau JC, Wang Z, Huang CA, Liyanage NPM, Kim S. CD3e-immunotoxin spares CD62L lo Tregs and reshapes organ-specific T-cell composition by preferentially depleting CD3e hi T cells. Front Immunol 2022; 13:1011190. [PMID: 36389741 PMCID: PMC9643874 DOI: 10.3389/fimmu.2022.1011190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/04/2022] [Indexed: 02/03/2023] Open
Abstract
CD3-epsilon(CD3e) immunotoxins (IT), a promising precision reagent for various clinical conditions requiring effective depletion of T cells, often shows limited treatment efficacy for largely unknown reasons. Tissue-resident T cells that persist in peripheral tissues have been shown to play pivotal roles in local and systemic immunity, as well as transplant rejection, autoimmunity and cancers. The impact of CD3e-IT treatment on these local cells, however, remains poorly understood. Here, using a new murine testing model, we demonstrate a substantial enrichment of tissue-resident Foxp3+ Tregs following CD3e-IT treatment. Differential surface expression of CD3e among T-cell subsets appears to be a main driver of Treg enrichment in CD3e-IT treatment. The surviving Tregs in CD3e-IT-treated mice were mostly the CD3edimCD62Llo effector phenotype, but the levels of this phenotype markedly varied among different lymphoid and nonlymphoid organs. We also found notable variations in surface CD3e levels among tissue-resident T cells of different organs, and these variations drive CD3e-IT to uniquely reshape T-cell compositions in local organs. The functions of organs and anatomic locations (lymph nodes) also affected the efficacy of CD3e-IT. The multi-organ pharmacodynamics of CD3e-IT and potential treatment resistance mechanisms identified in this study may generate new opportunities to further improve this promising treatment.
Collapse
Affiliation(s)
- Shihyoung Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Rajni Kant Shukla
- Department of Microbial Immunity and Infection, The Ohio State University, Columbus, OH, United States
| | - Hannah Yu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Alice Baek
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Sophie G. Cressman
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Sarah Golconda
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Ga-Eun Lee
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Hyewon Choi
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - John C. Reneau
- Division of Hematology, The Ohio State University, Columbus, OH, United States
| | - Zhirui Wang
- Department of Surgery, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Christene A. Huang
- Department of Surgery, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Namal P. M. Liyanage
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States,Department of Microbial Immunity and Infection, The Ohio State University, Columbus, OH, United States,Infectious Disease Institute, The Ohio State University, Columbus, OH, United States,*Correspondence: Namal P. M. Liyanage, ; Sanggu Kim,
| | - Sanggu Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States,Infectious Disease Institute, The Ohio State University, Columbus, OH, United States,*Correspondence: Namal P. M. Liyanage, ; Sanggu Kim,
| |
Collapse
|
5
|
Barjon C, Dahlqvist G, Ghazal K, Saliba F, Durand F, Duvoux C, Aoudjehane L, Conti F. Influence of everolimus-based treatment on circulating regulatory T cells after liver transplantation: Comparative study with tacrolimus-based therapy. Clin Res Hepatol Gastroenterol 2021; 45:101559. [PMID: 33191181 DOI: 10.1016/j.clinre.2020.10.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 02/04/2023]
Abstract
Liver transplantation remains the only treatment for terminal liver diseases. However, immunosuppressive drugs required for allograft acceptance are toxic and may be responsible for severe side effects. Modulating the immune system to induce tolerance is a promising approach to reduce immunosuppressive regimen. More particularly, promoting natural CD4+ CD25+ FoxP3+ Tregs could be crucial in achieving tolerance. Contrary to calcineurin inhibitors, reports indicate that mTOR inhibitors may have a positive impact on Tregs. Here we present the first randomized prospective clinical study where Tregs levels from liver transplanted patients receiving either tacrolimus or everolimus were monitored for 6 months, starting from the day of transplantation. A total of 30 patients from four centers were monitored. Blood samples were obtained at day 0, day 14, one month, three months and six months post-transplantation. Flow-cytometry immunophenotyping of Tregs (CD4+ CD25+ CD127- FoxP3+) and functional assays with Tregs were performed to assess their immunosuppressive capacity. Levels of Tregs were significantly reduced after one month of standard tacrolimus-based immunosuppressive regimen (p<0.05). Four months after conversion, levels of Tregs from patients treated with everolimus was significantly higher than patients under tacrolimus (p<0.02). Functional assays demonstrated that Tregs conserved their capacity to suppress the proliferation of activated PBMC.
Collapse
Affiliation(s)
- Clément Barjon
- UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France; De Duve Institute, UCLouvain, 1200 Brussels, Belgium
| | - Géraldine Dahlqvist
- UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France; Hepatogastroenterology Unit, Cliniques Universitaires Saint-Luc, Brussels, Belgium.
| | | | - Faouzi Saliba
- Centre Hépato-Biliaire, Hôpital Paul Brousse, AP-HP, Villejuif, France
| | - François Durand
- Liver Intensive Care Unit, Hôpital Beaujon, Assistance Publique-Hopitaux de Paris and University of Paris, Paris, France
| | - Christophe Duvoux
- Department of Hepatology, Hôpital Henri Mondor, AP-HP, Créteil, France
| | | | - Filomena Conti
- UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France; Liver Transplantation Unit, AP-HP, Hôpital de la Pitié-Salpêtrière, 75012 Paris, France
| |
Collapse
|
6
|
Otsuka S, Melis N, Gaida MM, Dutta D, Weigert R, Ashwell JD. Calcineurin inhibitors suppress acute graft-versus-host disease via NFAT-independent inhibition of T cell receptor signaling. J Clin Invest 2021; 131:147683. [PMID: 33822776 DOI: 10.1172/jci147683] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/31/2021] [Indexed: 01/31/2023] Open
Abstract
Inhibitors of calcineurin phosphatase activity (CNIs) such as cyclosporin A (CsA) are widely used to treat tissue transplant rejection and acute graft-versus-host disease (aGVHD), for which inhibition of gene expression dependent on nuclear factor of activated T cells (NFAT) is the mechanistic paradigm. We recently reported that CNIs inhibit TCR-proximal signaling by preventing calcineurin-mediated dephosphorylation of LckS59, an inhibitory modification, raising the possibility of another mechanism by which CNIs suppress immune responses. Here we used T cells from mice that express LckS59A, which cannot accept a phosphate at residue 59, to initiate aGVHD. Although CsA inhibited NFAT-dependent gene upregulation in allo-aggressive T cells expressing either LckWT or LckS59A, it was ineffective in treating disease when the T cells expressed LckS59A. Two important NFAT-independent T cell functions were found to be CsA-resistant in LckS59A T cells: upregulation of the cytolytic protein perforin in tissue-infiltrating CD8+ T cells and antigen-specific T/DC adhesion and clustering in lymph nodes. These results demonstrate that effective treatment of aGVHD by CsA requires NFAT-independent inhibition of TCR signaling. Given that NFATs are widely expressed and off-target effects are a major limitation in CNI use, it is possible that targeting TCR-associated calcineurin directly may provide effective therapies with less toxicity.
Collapse
Affiliation(s)
| | - Nicolas Melis
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Matthias M Gaida
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | | | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
7
|
Janikashvili N, Gérard C, Thébault M, Brazdova A, Boibessot C, Cladière C, Ciudad M, Greigert H, Ouandji S, Ghesquière T, Samson M, Audia S, Saas P, Bonnotte B. Efficiency of human monocyte-derived suppressor cell-based treatment in graft-versus-host disease prevention while preserving graft-versus-leukemia effect. Oncoimmunology 2021; 10:1880046. [PMID: 33659098 PMCID: PMC7899641 DOI: 10.1080/2162402x.2021.1880046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background Immunosuppressive cell-based therapy is a recent strategy for controlling Graft-versus-Host Disease (GvHD). Such cells ought to maintain their suppressive function in inflammatory conditions and in the presence of immunosuppressive agents currently used in allogeneic hematopoietic cell transplantation (allo-HCT). Moreover, these therapies should not diminish the benefits of allo-HCT, the Graft-versus-Leukemia (GvL) effect. We have previously reported on a novel subset of human monocyte-derived suppressor cells (HuMoSC) as a prospective approach for controlling GvHD.Objective The objective of this study was to explore the therapeutic relevance of the HuMoSC in clinical conditions. Methods Immune regulatory functions of HuMoSC were assessed in inflammatory conditions and in the presence of immunosuppressants. The therapeutic efficiency of the association of HuMoSC with immunosuppressants was evaluated in an experimental model of GvHD induced by human PBMC in NOD/SCID/IL2-Rγc−/− (NSG) mice. Interestingly, the inhibitory functions of HuMoSC against T lymphocytes and their ability to polarize Treg are preserved, in vitro, in inflammatory environments and are not affected by immunosuppressive agents. In vivo, the association of HuMoSC-based treatment with an immunosuppressive drug showed a synergistic effect for controlling GvHD. Furthermore, HuMoSC control GvHD while preserving GvL effect in a xeno-GvHD conditioned mouse model with cell neoplasm (CAL-1). HuMoSC are generated according to good manufacturing practices (GMP) and we demonstrated that these cells tolerate long-term preservation with unaltered phenotype and function.Conclusion HuMoSC-based therapy represents a promising approach for controlling GvHD and could be quickly implemented in clinical practice.
Collapse
Affiliation(s)
- Nona Janikashvili
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire Et Génique, Dijon, France.,Department of Immunology, Faculty of Medicine, Tbilisi State Medical University (TSMU), Tbilisi, Georgia
| | - Claire Gérard
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire Et Génique, Dijon, France.,Department of Internal Medicine, University Hospital, Dijon, France
| | - Marine Thébault
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire Et Génique, Dijon, France
| | - Andrea Brazdova
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire Et Génique, Dijon, France.,Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Clovis Boibessot
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire Et Génique, Dijon, France
| | - Claudie Cladière
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire Et Génique, Dijon, France
| | - Marion Ciudad
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire Et Génique, Dijon, France
| | - Hélène Greigert
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire Et Génique, Dijon, France
| | - Séthi Ouandji
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire Et Génique, Dijon, France
| | - Thibault Ghesquière
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire Et Génique, Dijon, France.,Department of Internal Medicine, University Hospital, Dijon, France
| | - Maxime Samson
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire Et Génique, Dijon, France.,Department of Internal Medicine, University Hospital, Dijon, France
| | - Sylvain Audia
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire Et Génique, Dijon, France.,Department of Internal Medicine, University Hospital, Dijon, France
| | - Philippe Saas
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire Et Génique, Besançon, France
| | - Bernard Bonnotte
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire Et Génique, Dijon, France.,Department of Internal Medicine, University Hospital, Dijon, France
| |
Collapse
|
8
|
Malone K, Diaz Diaz AC, Shearer JA, Moore AC, Waeber C. The effect of fingolimod on regulatory T cells in a mouse model of brain ischaemia. J Neuroinflammation 2021; 18:37. [PMID: 33516262 PMCID: PMC7847573 DOI: 10.1186/s12974-021-02083-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/14/2021] [Indexed: 11/30/2022] Open
Abstract
Background The role of the immune system in stroke is well-recognised. Fingolimod, an immunomodulatory agent licensed for the management of relapsing-remitting multiple sclerosis, has been shown to provide benefit in rodent models of stroke. Its mechanism of action, however, remains unclear. We hypothesised fingolimod increases the number and/or function of regulatory T cells (Treg), a lymphocyte population which promotes stroke recovery. The primary aim of this study was to rigorously investigate the effect of fingolimod on Tregs in a mouse model of brain ischaemia. The effect of fingolimod in mice with common stroke-related comorbidities (ageing and hypercholesteremia) was also investigated. Methods Young (15–17 weeks), aged C57BL/6 mice (72–73 weeks), and ApoE−/− mice fed a high-fat diet (20–21 weeks) underwent permanent electrocoagulation of the left middle cerebral artery. Mice received either saline or fingolimod (0.5 mg/kg or 1 mg/kg) at 2, 24, and 48 h post-ischaemia via intraperitoneal injection. Another cohort of young mice (8–9, 17–19 weeks) received short-term (5 days) or long-term (10 days) fingolimod (0.5 mg/kg) treatment. Flow cytometry was used to quantify Tregs in blood, spleen, and lymph nodes. Immunohistochemistry was used to quantify FoxP3+ cell infiltration into the ischaemic brain. Results Fingolimod significantly increased the frequency of Tregs within the CD4+ T cell population in blood and spleen post-ischaemia in all three mouse cohorts compared to untreated ischemic mice. The highest splenic Treg frequency in fingolimod-treated mice was observed in ApoE−/− mice (9.32 ± 1.73% vs. 7.8 ± 3.01% in young, 6.09 ± 1.64% in aged mice). The highest circulating Treg frequency was also noted in ApoE−/− mice (8.39 ± 3.26% vs. 5.43 ± 2.74% in young, 4.56 ± 1.60% in aged mice). Fingolimod significantly increased the number of FoxP3+ cells in the infarct core of all mice. The most pronounced effects were seen when mice were treated for 10 days post-ischaemia. Conclusions Fingolimod increases Treg frequency in spleen and blood post-ischaemia and enhances the number of FoxP3+ cells in the ischaemic brain. The effect of fingolimod on this regulatory cell population may underlie its neuroprotective activity and could be exploited as part of future stroke therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02083-5.
Collapse
Affiliation(s)
- Kyle Malone
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland.,School of Pharmacy, University College Cork, Cork, Ireland
| | - Andrea C Diaz Diaz
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland.,School of Pharmacy, University College Cork, Cork, Ireland
| | - Jennifer A Shearer
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland.,School of Pharmacy, University College Cork, Cork, Ireland
| | - Anne C Moore
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Christian Waeber
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland. .,School of Pharmacy, University College Cork, Cork, Ireland.
| |
Collapse
|
9
|
Li L, Liu S, Yu J. Autoimmune thyroid disease and type 1 diabetes mellitus: same pathogenesis; new perspective? Ther Adv Endocrinol Metab 2020; 11:2042018820958329. [PMID: 32973994 PMCID: PMC7493255 DOI: 10.1177/2042018820958329] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 08/20/2020] [Indexed: 12/14/2022] Open
Abstract
Autoimmune thyroid disease (AITD) and type 1 diabetes mellitus (T1DM) are two common autoimmune diseases that can occur concomitantly. In general, patients with diabetes have a high risk of AITD. It has been proposed that a complex genetic basis together with multiple nongenetic factors make a variable contribution to the pathogenesis of T1DM and AITD. In this paper, we summarize current knowledge in the field regarding potential pathogenic factors of T1DM and AITD, including human leukocyte antigen, autoimmune regulator, lymphoid protein tyrosine phosphatase, forkhead box protein P3, cytotoxic T lymphocyte-associated antigen, infection, vitamin D deficiency, and chemokine (C-X-C motif) ligand. These findings offer an insight into future immunotherapy for autoimmune diseases.
Collapse
Affiliation(s)
- Liyan Li
- Department of Endocrinology, First People’s Hospital of Jinan, Jinan, People’s Republic of China
| | - Shudong Liu
- Department of Endocrinology, Shandong Rongjun General Hospital, Jinan, People’s Republic of China
| | - Junxia Yu
- Department of Endocrinology, Tengzhou Central People’s Hospital, 181 Xingtan Road, Tengzhou, Shandong Province, 277500, People’s Republic of China
| |
Collapse
|
10
|
Zhao LL. TIPE2 suppresses progression and tumorigenesis of the oral tongue squamous cell carcinoma by regulating FoxP3 + regulatory T cells. J Bioenerg Biomembr 2020; 52:279-289. [PMID: 32594290 DOI: 10.1007/s10863-020-09840-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/01/2020] [Indexed: 12/21/2022]
Abstract
To discover the effect of tumor necrosis factor-alpha-induced protein 8-like 2 (TIPE2) on the oral tongue squamous cell carcinoma (OTSCC) via affecting FoxP3+ regulatory T (Treg) cells. Immunohistochemistry was conducted to examine TIPE2 and FoxP3 expressions in OTSCC tumor tissues and corresponding oral mucosa. Tca8113 cells were transfected with TIPE2/control lentiviral activation particles followed by the detection with qRT-PCR, Western blotting, MTT assay, Wound healing, Transwell assay and Annexin V-FITC/PI staining. In vivo experiment was carried out on the nude mice xenografts of OTSCC with TIPE2 overexpression to observe the tumor volume and survival, and the CD4+ T cell subgroups were detected by flow cytometry. TIPE2 was lower in the OTSCC tissues than the corresponding oral mucosa, which was correlated with T stage, N stage, TNM stage, and differentiation of patients. Patients with TIPE2-positive expression had worse prognosis and lower expression of FoxP3+ Treg cells than the negative ones. Furthermore, TIPE2 overexpression curbed proliferation, invasion and migration of Tca8113 cells, while cell apoptosis was increased. Besides, TIPE2 suppressed the tumor growth and extended the survival of OTSCC mice, with the decreased proportion of FoxP3+ Treg cells in the spleen and tumor-infiltrated lymphocytes (TILs). The clinical results showed the down-regulation of TIPE2 in OTSCC tissues. Meanwhile TIPE2 overexpression affected OTSCC cells biological behavior in vitro, as well as exhibited strong tumor-growth suppressive effects in vivo, which may be a potential therapeutic target in OTSCC via regulating FoxP3+ Treg cells.
Collapse
Affiliation(s)
- Li-Li Zhao
- Department of Stomatology, Caoxian people's Hospital, Heze City, 274400, Shandong Province, China.
| |
Collapse
|
11
|
Slight-Webb S, Guthridge JM, Chakravarty EF, Chen H, Lu R, Macwana S, Bean K, Maecker HT, Utz PJ, James JA. Mycophenolate mofetil reduces STAT3 phosphorylation in systemic lupus erythematosus patients. JCI Insight 2019; 4:124575. [PMID: 30674728 DOI: 10.1172/jci.insight.124575] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/11/2018] [Indexed: 11/17/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a highly variable autoimmune disease that can involve severe organ-threatening symptoms, such as lupus nephritis. Certain drugs, such as mycophenolate mofetil (MMF), are effective at reducing morbidity associated with nephritis; however, the immune pathways associated with disease suppression are poorly defined. Here, we provide evidence that MMF inhibits phosphorylation of STAT3 and other associated immune pathways. Using mass cytometry and bead-based or ELISA assays, the systemic phenotype of SLE patients not taking (MMF-) or taking (MMF+) MMF were studied. MMF+ SLE patients had significant reductions in total numbers of transitional B cells, plasmablasts, and T cells, specifically CD4+ Th17-type and CD4+ Treg-type cells, compared with MMF- patients. Plasma soluble mediators were decreased in MMF+ patients including chemokines (MIG/CXCL9 and SDF-1α/CXCL12) and growth factors (VEGF-A and PDGF-BB). Soluble mediators and cell subsets grouped by functional properties revealed significant modifications associated with STAT3 and B cell pathways. Further, healthy PBMCs treated with IL-6 revealed a reduction in p-STAT3 following the addition of mycophenolic acid (the active metabolite of MMF). In conclusion, the inhibition of STAT3 phosphorylation by MMF may explain the effectiveness of this treatment in SLE patients, since increased levels of p-STAT3 are associated with disease pathology.
Collapse
Affiliation(s)
- Samantha Slight-Webb
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Joel M Guthridge
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA.,Departments of Medicine and Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Eliza F Chakravarty
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Hua Chen
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Rufei Lu
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA.,Departments of Medicine and Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Susan Macwana
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Krista Bean
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | | | - Paul J Utz
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
| | - Judith A James
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA.,Departments of Medicine and Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
12
|
Martin-Moreno PL, Tripathi S, Chandraker A. Regulatory T Cells and Kidney Transplantation. Clin J Am Soc Nephrol 2018; 13:1760-1764. [PMID: 29789350 PMCID: PMC6237070 DOI: 10.2215/cjn.01750218] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The ability of the immune system to differentiate self from nonself is critical in determining the immune response to antigens expressed on transplanted tissue. Even with conventional immunosuppression, acceptance of the allograft is an active process often determined by the presence of regulatory T cells (Tregs). Tregs classically are CD4+ cells that constitutively express high levels of the IL-2 receptor α chain CD25, along with the transcription factor Foxp3. The use of Tregs in the field of solid organ transplantation is related specifically to the objective of achieving tolerance, with the goal of reducing or eliminating immunosuppressive drugs as well as maintaining tissue repair and managing acute rejection. A key issue in clinical use of Tregs is how to effectively expand the number of Tregs, either through increasing numbers of endogenous Tregs or by the direct infusion of exogenously expanded Tregs. In order to realize the benefits of Treg therapy in solid organ transplantation, a number of outstanding challenges need to be overcome, including assuring an effective expansion of Tregs, improving long-term Treg stability and reduction of risk-related to off-target, nonspecific, immunosuppressive effects related specially to cancer.
Collapse
Affiliation(s)
- Paloma Leticia Martin-Moreno
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts; and
- Nephrology Department, Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Sudipta Tripathi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Anil Chandraker
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts; and
| |
Collapse
|
13
|
Kusano J, Tanaka S, Matsuda H, Hara Y, Fujii Y, Suzuki S, Sekiyama M, Ando E, Sugiyama K, Hirano T. Vitamin K1 and Vitamin K2 immunopharmacological effects on the peripheral lymphocytes of healthy subjects and dialysis patients, as estimated by the lymphocyte immunosuppressant sensitivity test. J Clin Pharm Ther 2018; 43:895-902. [DOI: 10.1111/jcpt.12747] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 06/25/2018] [Indexed: 11/30/2022]
Affiliation(s)
- Junichi Kusano
- Division of Pharmacy; Kanagawa-ken Keiyukai Keiyu Hospital; Kanagawa Japan
| | - Sachiko Tanaka
- Department of Clinical Pharmacology; School of Pharmacy; Tokyo University of Pharmacy and Life Sciences; Tokyo Japan
| | - Hiroto Matsuda
- Division of Nephrology; Kanagawa-ken Keiyukai Keiyu Hospital; Kanagawa Japan
| | - Yoshikazu Hara
- Division of Nephrology; Kanagawa-ken Keiyukai Keiyu Hospital; Kanagawa Japan
| | - Yoshiaki Fujii
- Division of Nephrology; Kanagawa-ken Keiyukai Keiyu Hospital; Kanagawa Japan
| | - Shinya Suzuki
- Division of Pharmacy; Kanagawa-ken Keiyukai Keiyu Hospital; Kanagawa Japan
| | - Masao Sekiyama
- Division of Pharmacy; Kanagawa-ken Keiyukai Keiyu Hospital; Kanagawa Japan
| | - Eiki Ando
- Division of Pharmacy; Kanagawa-ken Keiyukai Keiyu Hospital; Kanagawa Japan
| | - Kentaro Sugiyama
- Department of Clinical Pharmacology; School of Pharmacy; Tokyo University of Pharmacy and Life Sciences; Tokyo Japan
| | - Toshihiko Hirano
- Department of Clinical Pharmacology; School of Pharmacy; Tokyo University of Pharmacy and Life Sciences; Tokyo Japan
| |
Collapse
|
14
|
FOXP3 rs3761549 polymorphism predicts long-term renal allograft function in patients receiving cyclosporine-based immunosuppressive regimen. Gene 2018; 644:93-100. [DOI: 10.1016/j.gene.2017.10.081] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 09/04/2017] [Accepted: 10/30/2017] [Indexed: 01/20/2023]
|
15
|
Yan JJ, Lee JG, Jang JY, Koo TY, Ahn C, Yang J. IL-2/anti-IL-2 complexes ameliorate lupus nephritis by expansion of CD4+CD25+Foxp3+ regulatory T cells. Kidney Int 2017; 91:603-615. [DOI: 10.1016/j.kint.2016.09.022] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 09/08/2016] [Accepted: 09/08/2016] [Indexed: 12/23/2022]
|
16
|
Norero B, Serrano CA, Sanchez-Fueyo A, Duarte I, Torres J, Ocquetau M, Barrera F, Arrese M, Soza A, Benítez C. Conversion to mycophenolate mofetil monotherapy in liver recipients: Calcineurin inhibitor levels are key. Ann Hepatol 2017; 16:94-106. [PMID: 28051798 DOI: 10.5604/16652681.1226820] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The use of calcineurin inhibitors (CNI) after liver transplantation is associated with post-transplant nephrotoxicity. Conversion to mycophenolate mofetil (MMF) monotherapy improves renal function, but is related to graft rejection in some recipients. Our aim was to identify variables associated with rejection after conversion to MMF monotherapy. Conversion was attempted in 40 liver transplant recipients. Clinical variables were determined and peripheral mononuclear blood cells were immunophenotyped during a 12-month follow-up. Conversion was classified as successful (SC) if rejection did not occur during the follow-up. MMF conversion was successful with 28 patients (70%) and was associated with higher glomerular filtration rates at the end of study. It also correlated with increased time elapsed since transplantation, low baseline CNI levels (Tacrolimus ≤ 6.5 ng/mL or Cyclosporine ≤ 635 ng/mL) and lower frequency of tacrolimus use. The only clinical variable independently related to SC in multivariate analysis was low baseline CNI levels (p = 0.02, OR: 6.93, 95%, CI: 1.3-29.7). Mean baseline fluorescent intensity of FOXP3+ T cells was significantly higher among recipients with SC. In conclusion, this study suggests that baseline CNI levels can be used to identify recipients with higher probability of SC to MMF monotherapy. Clinicaltrials.gov identification: NCT01321112.
Collapse
Affiliation(s)
- Blanca Norero
- Departamentos de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile
| | - Carolina A Serrano
- Departamentos de Gastroenterología y Nutrición Pediátrica, Escuela de Medicina, Pontificia Universidad Católica de Chile
| | - Alberto Sanchez-Fueyo
- Institute of Liver Studies, MRC Centre for Transplantation, King's College London, London SE5 9RS, United Kingdom
| | - Ignacio Duarte
- Patología y Escuela de Medicina, Pontificia Universidad Católica de Chile
| | - Javiera Torres
- Patología y Escuela de Medicina, Pontificia Universidad Católica de Chile
| | - Mauricio Ocquetau
- Hemato-Oncología, Escuela de Medicina, Pontificia Universidad Católica de Chile
| | | | - Marco Arrese
- Departamentos de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile
| | - Alejandro Soza
- Departamentos de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile
| | - Carlos Benítez
- Departamentos de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile
| |
Collapse
|
17
|
Northrup L, Sullivan BP, Hartwell BL, Garza A, Berkland C. Screening Immunomodulators To Skew the Antigen-Specific Autoimmune Response. Mol Pharm 2016; 14:66-80. [PMID: 28043135 DOI: 10.1021/acs.molpharmaceut.6b00725] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Current therapies to treat autoimmune diseases often result in side effects such as nonspecific immunosuppression. Therapies that can induce antigen-specific immune tolerance provide an opportunity to reverse autoimmunity and mitigate the risks associated with global immunosuppression. In an effort to induce antigen-specific immune tolerance, co-administration of immunomodulators with autoantigens has been investigated in an effort to reprogram autoimmunity. To date, identifying immunomodulators that may skew the antigen-specific immune response has been ad hoc at best. To address this need, we utilized splenocytes obtained from mice with experimental autoimmune encephalomyelitis (EAE) in order to determine if certain immunomodulators may induce markers of immune tolerance following antigen rechallenge. Of the immunomodulatory compounds investigated, only dexamethasone modified the antigen-specific immune response by skewing the cytokine response and decreasing T-cell populations at a concentration corresponding to a relevant in vivo dose. Thus, antigen-educated EAE splenocytes provide an ex vivo screen for investigating compounds capable of skewing the antigen-specific immune response, and this approach could be extrapolated to antigen-educated cells from other diseases or human tissues.
Collapse
Affiliation(s)
- Laura Northrup
- Department of Pharmaceutical Chemistry, University of Kansas , Lawrence, Kansas 66047, United States
| | - Bradley P Sullivan
- Department of Pharmaceutical Chemistry, University of Kansas , Lawrence, Kansas 66047, United States
| | - Brittany L Hartwell
- Bioengineering Graduate Program, University of Kansas , Lawrence, Kansas 66045, United States
| | - Aaron Garza
- Department of Chemical and Petroleum Engineering, University of Kansas , Lawrence, Kansas 66045, United States
| | - Cory Berkland
- Department of Pharmaceutical Chemistry, University of Kansas , Lawrence, Kansas 66047, United States.,Bioengineering Graduate Program, University of Kansas , Lawrence, Kansas 66045, United States.,Department of Chemical and Petroleum Engineering, University of Kansas , Lawrence, Kansas 66045, United States
| |
Collapse
|
18
|
Han C, Wu T, Na N, Zhao Y, Li W, Zhao Y. The effect of immunosuppressive drug cyclosporine A on myeloid-derived suppressor cells in transplanted mice. Inflamm Res 2016; 65:679-88. [PMID: 27147271 DOI: 10.1007/s00011-016-0949-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 04/14/2016] [Accepted: 04/25/2016] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVE Myeloid-derived suppressor cells (MDSCs) play important roles in preventing graft rejection. Immunosuppressive drug cyclosporine A (CsA) is widely used in clinics to treat patients with allografts and autoimmune diseases. However, the effect of CsA on CD11b(+)Gr1(+) MDSCs has not been studied. SUBJECTS The subjects of the study include BALB/c skin-grafted C57BL/6 mice and the in vitro MDSCs induction system. TREATMENT Skin-grafted mice were treated with CsA (30 mg/kg, i.p.) or control buffer daily. 0.01 μg/ml CsA was added during MDSC induction. METHODS Flow cytometry was used to check cell phenotypes and proliferation. Real-time PCR was used for gene expressions. Inducible nitric oxide synthase iNOS-knockout mice were used for the role of iNOS in the immunosuppression of MDSCs. RESULTS CsA in MDSC-induction system significantly increased the number of CD11b(+)Gr1(+)MDSCs without detectable effects on the expressions of CD31, CD115 and CD274. However, GM-CSF + CsA-induced MDSCs express higher iNOS than control MDSCs. Blocking iNOS activity by inhibitor or gene deletion significantly reversed the inhibitory effects of GM-CSF + CsA-induced MDSCs on T cell proliferation. Importantly, CsA treatment significantly increased the number and the immunosuppressive ability of CD11b(+)Gr1(+)MDSCs in allogeneic skin-grafted mice. CONCLUSIONS CsA promotes MDSC induction and immunosuppressive function, which might be of clinical importance in treating graft rejection and autoimmune diseases.
Collapse
Affiliation(s)
- Chenlu Han
- College of Life Science, Henan Normal University, Xinxiang, Henan, China.,Transplantation Biology Research Division, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road 1-5, Chaoyang District, 100101, Beijing, China
| | - Tingting Wu
- College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Ning Na
- Department of Kidney Transplantation, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yang Zhao
- College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Weiguo Li
- Transplantation Biology Research Division, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road 1-5, Chaoyang District, 100101, Beijing, China.
| | - Yong Zhao
- College of Life Science, Henan Normal University, Xinxiang, Henan, China.
| |
Collapse
|
19
|
Lee KW, Seo YD, Oh SC, Suh SW, Jeong J, Kim H, Yi NJ, Suh KS. What is the best immunosuppressant combination in terms of antitumor effect in hepatocellular carcinoma? Hepatol Res 2016; 46:593-600. [PMID: 26348114 DOI: 10.1111/hepr.12588] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 08/21/2015] [Accepted: 08/31/2015] [Indexed: 12/09/2022]
Abstract
AIM Despite its known anticancer benefits, monotherapy with sirolimus is not sufficient to achieve optimal immunosuppression to prevent rejection. However, there is no published prospective study to compare the anticancer effect between various immunosuppressive combinations. Therefore, we analyzed the anticancer effects of various immunosuppressive regimens in order to provide experimental evidence for selecting an optimal immunosuppressive regimen after liver transplantation for hepatocellular carcinoma (HCC). METHODS The Huh7 cell line was used as a model for HCC in both in vitro and in vivo mouse experiments. The immunosuppressant regimens tested were: tacrolimus, sirolimus, MMF, sirolimus plus tacrolimus, and sirolimus plus MMF. 3-(4 5-Dimethylthiazol-2-yl)-2 5-diphenyltetrazolium bromide assays showed that the sirolimus plus MMF combination appeared to be synergistic in its cell suppressive effects, achieving statistically significant lowest cell viability. RESULTS In vitro western blot analysis showed that there were lower levels of expression of phosphorylated mammalian target of rapamycin, p70S6K and p4EBP1, transforming growth factor-β and pSmad3 expression in the cells treated with sirolimus, MMF and sirolimus plus MMF. Finally, in the mouse model of tumorigenesis, the sirolimus plus MMF and sirolimus plus tacrolimus showed the most suppressive effect in terms of tumor volume. CONCLUSION Throughout both the in vitro and in vivo experiments, the sirolimus and MMF combination had the most consistent and greatest antiproliferative effects.
Collapse
Affiliation(s)
- Kwang-Woong Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Yongwoo David Seo
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Seung Cheol Oh
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Suk-Won Suh
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Jaehong Jeong
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyeyoung Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Nam-Joon Yi
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Kyung-Suk Suh
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
20
|
Northrup L, Christopher MA, Sullivan BP, Berkland C. Combining antigen and immunomodulators: Emerging trends in antigen-specific immunotherapy for autoimmunity. Adv Drug Deliv Rev 2016; 98:86-98. [PMID: 26546466 DOI: 10.1016/j.addr.2015.10.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 10/23/2015] [Accepted: 10/26/2015] [Indexed: 01/05/2023]
Abstract
A majority of current therapies for autoimmune diseases are general immunosuppressants, which can compromise patient response to opportunistic infection and lead to adverse events. Using antigen-specific immunotherapy (ASIT) to selectively disarm autoimmune diseases, without suppressing the global immune response, would be a transformative therapy for patients. ASIT has been used historically in allergy hyposensitization therapy to induce tolerance to an allergen. Similar strategies to induce immune tolerance toward autoantigens responsible for autoimmune disease have been attempted but have yielded limited clinical success. Recent studies of ASIT for autoimmunity have explored combination therapy, combining the disease-causing autoantigen with an immunomodulatory compound. ASIT combination therapy may direct the immune response in an antigen-specific manner, potentially reversing the root cause of autoimmunity while limiting side effects. This review analyzes recent advances in ASIT applied to autoimmune diseases, emphasizing current combination therapies and future strategies.
Collapse
Affiliation(s)
- Laura Northrup
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA
| | - Matthew A Christopher
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA
| | - Bradley P Sullivan
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA
| | - Cory Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA; Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, KS 66045, USA.
| |
Collapse
|
21
|
Lu QB, Zhu S. Modulation of an aqueous extract of Chinese medicine prescription Anzi Heji () on ratio of CD4 +CD25 +FOXP3 + regulatory T cells in anticardiolipin antibody-positive patients with threatened abortion. Chin J Integr Med 2016:10.1007/s11655-015-2444-3. [PMID: 26919832 DOI: 10.1007/s11655-015-2444-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Indexed: 10/22/2022]
Abstract
OBJECTIVE To evaluate Chinese medicine prescription, Anzi Heji (, AZHJ), on immune regulation of CD4+CD25+FOXP3+ regulatory T cells (Tregs) in anticardiolipin antibody (ACA)-positive patients with threatened abortion. METHODS Twenty-seven ACA-positive female patients with threatened abortion in the study group were treated with an aqueous extract of AZHJ 125 mL, twice daily for 4 consecutive weeks. The results were compared with control group composed by 15 healthy pregnant women. The ratio of CD4+CD25+FOXP3+ Treg in peripheral blood was identified by flow cytometry. The indicators of ACA were detected by enzyme-linked immunosorbent assay, and embryo development was checked by B-ultrasound. RESULTS Compared with the control group, the ratio of CD4+CD25+FOXP3+ Treg cells in the study group was significantly lower before AZHJ treatment (P<0.01) and significantly increased after AZHJ treatment (P<0.01). After treatment, 20 of 27 patients (85%) showed that ACA indicators turned into negative, and 7 cases of quantitative indicators of ACA titers were significantly decreased (P<0.01). Total efficiency of treating miscarriage by AZHJ was 92.59%. CONCLUSION AZHJ can regulate the immune function of pregnant women by increasing number of CD4+CD25+FOXP3+ Tregs.
Collapse
Affiliation(s)
- Qi-Bin Lu
- Department of Gynaecology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, 210029, China.
| | - Shu Zhu
- The First Clinical Medicine College, Nanjing University of Chinese Medicine, Nanjing, 210029, China
| |
Collapse
|
22
|
Wu T, Zhao Y, Wang H, Li Y, Shao L, Wang R, Lu J, Yang Z, Wang J, Zhao Y. mTOR masters monocytic myeloid-derived suppressor cells in mice with allografts or tumors. Sci Rep 2016; 6:20250. [PMID: 26833095 PMCID: PMC4735296 DOI: 10.1038/srep20250] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 12/30/2015] [Indexed: 01/06/2023] Open
Abstract
CD11b(+) Gr1(+) myeloid-derived suppressor cells (MDSCs) play critical roles in controlling the processes of tumors, infections, autoimmunity and graft rejection. Immunosuppressive drug rapamycin (RPM), targeting on the key cellular metabolism molecule mTOR, is currently used in clinics to treat patients with allo-grafts, autoimmune diseases and tumors. However, the effect of RPM on MDSCs has not been studied. RPM significantly decreases the cell number and the immunosuppressive ability on T cells of CD11b(+) Ly6C(high) monocytic MDSCs (M-MDSCs) in both allo-grafts-transplanted and tumor-bearing mice respectively. Mice with a myeloid-specific deletion of mTOR have poor M-MDSCs after grafting with allo-skin tissue or a tumor. Grafting of allo-skin or tumors significantly activates glycolysis pathways in myeloid precursor cells in bone marrow, which is inhibited by RPM or mTOR deletion. 2-deoxyglucose (2-DG), an inhibitor of the glycolytic pathway, inhibits M-MDSC differentiation from precursors, while enhancing glycolysis by metformin significantly rescues the RPM-caused deficiency of M-MDSCs. Therefore, we offer evidence supporting that mTOR is an intrinsic factor essential for the differentiation and immunosuppressive function of M-MDSCs and that these metabolism-relevant medicines may impact MDSCs-mediated immunosuppression or immune tolerance induction, which is of considerable clinical importance in treating graft rejection, autoimmune diseases and cancers.
Collapse
Affiliation(s)
- Tingting Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yang Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Hao Wang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | - Yang Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Lijuan Shao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Department of Oncology, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Ruoyu Wang
- Department of Oncology, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Jun Lu
- Hepatology and Cancer Biotherapy Ward, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Zhongzhou Yang
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Junjie Wang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
23
|
Mazzola MA, Raheja R, Murugaiyan G, Rajabi H, Kumar D, Pertel T, Regev K, Griffin R, Aly L, Kivisakk P, Nejad P, Patel B, Gwanyalla N, Hei H, Glanz B, Chitnis T, Weiner HL, Gandhi R. Identification of a novel mechanism of action of fingolimod (FTY720) on human effector T cell function through TCF-1 upregulation. J Neuroinflammation 2015; 12:245. [PMID: 26714756 PMCID: PMC4696082 DOI: 10.1186/s12974-015-0460-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 12/15/2015] [Indexed: 12/21/2022] Open
Abstract
Background Fingolimod (FTY720), the first oral treatment for multiple sclerosis (MS), blocks immune cell trafficking and prevents disease relapses by downregulation of sphingosine-1-phosphate receptor. We determined the effect of FTY720 on human T cell activation and effector function. Methods T cells from MS patients and healthy controls were isolated to measure gene expression profiles in the presence or absence of FTY720 using nanostring and quantitative real-time polymerase chain reaction (qPCR). Cytokine protein expression was measured using luminex assay and flow cytometry analysis. Lentivirus vector carrying short hairpin RNA (shRNA) was used to knock down the expression of specific genes in CD4+ T cells. Chromatin immunoprecipitation was performed to assess T cell factor 1 (TCF-1) binding to promoter regions. Luciferase assays were performed to test the direct regulation of interferon gamma (IFN-γ) and granzyme B (GZMB) by TCF-1. Western blot analysis was used to assess the phosphorylation status of Akt and GSK3β. Results We showed that FTY720 treatment not only affects T cell trafficking but also T cell activation. Patients treated with FTY720 showed a significant reduction in circulating CD4 T cells. Activation of T cells in presence of FTY720 showed a less inflammatory phenotype with reduced production of IFN-γ and GZMB. This decreased effector phenotype of FTY720-treated T cells was dependent on the upregulation of TCF-1. FTY720-induced TCF-1 downregulated the pathogenic cytokines IFN-γ and GZMB by binding to their promoter/enhancer regions and mediating epigenetic modifications. Furthermore, we observed that TCF-1 expression was lower in T cells from multiple sclerosis patients than in those from healthy individuals, and FTY720 treatment increased TCF-1 expression in multiple sclerosis patients. Conclusions These results reveal a previously unknown mechanism of the effect of FTY720 on human CD4+ T cell modulation in multiple sclerosis and demonstrate the role of TCF-1 in human T cell activation and effector function. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0460-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maria Antonietta Mazzola
- Brigham and Women's Hospital, Ann Romney Center for Neurologic Diseases, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA.
| | - Radhika Raheja
- Brigham and Women's Hospital, Ann Romney Center for Neurologic Diseases, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA.
| | - Gopal Murugaiyan
- Brigham and Women's Hospital, Ann Romney Center for Neurologic Diseases, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA.
| | - Hasan Rajabi
- Dana Farber Cancer Institute, Boston, MA, 02115, USA.
| | - Deepak Kumar
- Department of Biochemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA.
| | - Thomas Pertel
- Brigham and Women's Hospital, Ann Romney Center for Neurologic Diseases, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA.
| | - Keren Regev
- Brigham and Women's Hospital, Ann Romney Center for Neurologic Diseases, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA.
| | - Russell Griffin
- Brigham and Women's Hospital, Ann Romney Center for Neurologic Diseases, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA.
| | - Lilian Aly
- Brigham and Women's Hospital, Ann Romney Center for Neurologic Diseases, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA.
| | - Pia Kivisakk
- Brigham and Women's Hospital, Ann Romney Center for Neurologic Diseases, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA.
| | - Parham Nejad
- Brigham and Women's Hospital, Ann Romney Center for Neurologic Diseases, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA.
| | - Bonny Patel
- Brigham and Women's Hospital, Ann Romney Center for Neurologic Diseases, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA.
| | - Nguendab Gwanyalla
- Brigham and Women's Hospital, Ann Romney Center for Neurologic Diseases, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA.
| | - Hillary Hei
- Brigham and Women's Hospital, Ann Romney Center for Neurologic Diseases, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA.
| | - Bonnie Glanz
- Partners MS Center, Brigham and Women's Hospital, 1 Brookline Place, Brookline, MA, 02445, USA.
| | - Tanuja Chitnis
- Partners MS Center, Brigham and Women's Hospital, 1 Brookline Place, Brookline, MA, 02445, USA.
| | - Howard L Weiner
- Brigham and Women's Hospital, Ann Romney Center for Neurologic Diseases, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA. .,Partners MS Center, Brigham and Women's Hospital, 1 Brookline Place, Brookline, MA, 02445, USA.
| | - Roopali Gandhi
- Brigham and Women's Hospital, Ann Romney Center for Neurologic Diseases, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA.
| |
Collapse
|
24
|
Zhang A, Ning B, Sun N, Wei J, Ju X. Indirubin Increases CD4+CD25+Foxp3+ Regulatory T Cells to Prevent Immune Thrombocytopenia in Mice. PLoS One 2015; 10:e0142634. [PMID: 26571298 PMCID: PMC4646632 DOI: 10.1371/journal.pone.0142634] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 10/23/2015] [Indexed: 01/10/2023] Open
Abstract
Indirubin, a traditional Chinese medicine, is used to treat autoimmune diseases in clinics. However, the effects of indirubin on the immunosuppressive CD4+CD25+Foxp3+ regulatory T cells (Treg) have not been addressed. Thus, we aimed to investigate the effects of indirubin on CD4+CD25+Treg cells in immune thrombocytopenia (ITP) CBA mice, which were established by immunization with Wistar rat platelets. 50 mg/kg indirubin treatment daily for 4 weeks significantly decreased anti-platelet antibody production and prevented the decrease of platelets caused by immunization in ITP mice. Consistently, indirubin significantly enhanced the percentage and cell number of CD4+CD25+Foxp3+Treg cells in the peripheral blood, spleen and lymph nodes. We also observed a significant increase of the frequency and cell number of CD4+CD25+Foxp3+Treg cells in the thymus upon indirubin treatment. Furthermore, CD4+CD25+Treg cells from indirubin-treated mice showed similar immunosuppression on T effector cells as compared to those from control mice. Altogether, indirubin ameliorates ITP by enhancing CD4+CD25+Foxp3+Treg cell level with preserving immunosuppressive function.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/therapeutic use
- Antibodies, Monoclonal/chemistry
- Blood Platelets/metabolism
- Disease Models, Animal
- Female
- Flow Cytometry
- Forkhead Transcription Factors/metabolism
- Immune Tolerance
- Immunosuppressive Agents/chemistry
- Indoles/therapeutic use
- Interleukin-2 Receptor alpha Subunit/metabolism
- Lymph Nodes/metabolism
- Mice
- Mice, Inbred CBA
- Microscopy, Fluorescence
- Purpura, Thrombocytopenic, Idiopathic/metabolism
- Purpura, Thrombocytopenic, Idiopathic/prevention & control
- Rats
- Rats, Wistar
- Spleen/cytology
- Spleen/metabolism
- T-Lymphocytes, Regulatory/cytology
Collapse
Affiliation(s)
- Aijun Zhang
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, China
| | - Bin Ning
- Department of Orthopaedic, Jinan Central Hospital, Shandong University, Jinan, China
| | - Nianzheng Sun
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, China
| | - Jianlu Wei
- Department of Orthopaedic, Jinan Central Hospital, Shandong University, Jinan, China
| | - Xiuli Ju
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, China
- * E-mail:
| |
Collapse
|
25
|
Advances on Non-CD4 + Foxp3+ T Regulatory Cells: CD8+, Type 1, and Double Negative T Regulatory Cells in Organ Transplantation. Transplantation 2015; 99:1553-9. [PMID: 26193065 DOI: 10.1097/tp.0000000000000813] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The overwhelming body of research on T regulatory cells (Treg) has focused on CD4 + CD25 + Foxp3+ T cells. However, recent years have witnessed a resurgence in interest in CD4 - CD8+, CD4 - CD8- (double negative [DN]), and CD4 + Foxp3- type 1 Treg (Tr1) Treg and their role in controlling autoimmune diseases and in promoting the survival of organ allografts and xenografts. CD8+ and DN Treg can arise spontaneously (natural Treg) or can be induced in situ. Both CD8+ and DN Treg have been shown to enhance the survival of organ allografts and xenografts. Additionally, both can suppress alloimmune responses by contact-dependent mechanisms by either inducing apoptosis or mediating direct cytolysis of effector T cells. CD8+, DN, and Tr1 Treg can also act in a contact-independent manner by elaborating soluble immunosuppressive factors, such as TGF-β and IL-10. Applying CD8+, DN, and Tr1 Treg for enhancing the survival of organ allografts and xenografts is still in its infancy but holds significant potential. Furthermore, there is a need for a more comprehensive understanding of how current immunosuppressive therapies applied to organ transplantations affect the wide array of Treg populations.
Collapse
|
26
|
Lee CF, Lo YC, Cheng CH, Furtmüller GJ, Oh B, Andrade-Oliveira V, Thomas AG, Bowman CE, Slusher BS, Wolfgang MJ, Brandacher G, Powell JD. Preventing Allograft Rejection by Targeting Immune Metabolism. Cell Rep 2015; 13:760-770. [PMID: 26489460 DOI: 10.1016/j.celrep.2015.09.036] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 08/04/2015] [Accepted: 09/11/2015] [Indexed: 12/13/2022] Open
Abstract
Upon antigen recognition and co-stimulation, T lymphocytes upregulate the metabolic machinery necessary to proliferate and sustain effector function. This metabolic reprogramming in T cells regulates T cell activation and differentiation but is not just a consequence of antigen recognition. Although such metabolic reprogramming promotes the differentiation and function of T effector cells, the differentiation of regulatory T cells employs different metabolic reprogramming. Therefore, we hypothesized that inhibition of glycolysis and glutamine metabolism might prevent graft rejection by inhibiting effector generation and function and promoting regulatory T cell generation. We devised an anti-rejection regimen involving the glycolytic inhibitor 2-deoxyglucose (2-DG), the anti-type II diabetes drug metformin, and the inhibitor of glutamine metabolism 6-diazo-5-oxo-L-norleucine (DON). Using this triple-drug regimen, we were able to prevent or delay graft rejection in fully mismatched skin and heart allograft transplantation models.
Collapse
Affiliation(s)
- Chen-Fang Lee
- Sidney-Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Chang-Gung Transplantation Institute, Department of Liver and Transplantation Surgery, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Ying-Chun Lo
- Sidney-Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Chih-Hsien Cheng
- Sidney-Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Chang-Gung Transplantation Institute, Department of Liver and Transplantation Surgery, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Georg J Furtmüller
- Vascularized Composite Allotransplantation Laboratory, Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Byoungchol Oh
- Vascularized Composite Allotransplantation Laboratory, Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Vinicius Andrade-Oliveira
- Sidney-Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Ajit G Thomas
- Department of Neurology and Brain Science Institute, NeuroTranslational Drug Discovery Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Caitlyn E Bowman
- Department of Biological Chemistry, Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Barbara S Slusher
- Department of Neurology and Brain Science Institute, NeuroTranslational Drug Discovery Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael J Wolfgang
- Department of Biological Chemistry, Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Gerald Brandacher
- Vascularized Composite Allotransplantation Laboratory, Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jonathan D Powell
- Sidney-Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
27
|
Scottà C, Fanelli G, Hoong SJ, Romano M, Lamperti EN, Sukthankar M, Guggino G, Fazekasova H, Ratnasothy K, Becker PD, Afzali B, Lechler RI, Lombardi G. Impact of immunosuppressive drugs on the therapeutic efficacy of ex vivo expanded human regulatory T cells. Haematologica 2015; 101:91-100. [PMID: 26471483 DOI: 10.3324/haematol.2015.128934] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 10/09/2015] [Indexed: 12/21/2022] Open
Abstract
Immunosuppressive drugs in clinical transplantation are necessary to inhibit the immune response to donor antigens. Although they are effective in controlling acute rejection, they do not prevent long-term transplant loss from chronic rejection. In addition, immunosuppressive drugs have adverse side effects, including increased rate of infections and malignancies. Adoptive cell therapy with human Tregs represents a promising strategy for the induction of transplantation tolerance. Phase I/II clinical trials in transplanted patients are already underway, involving the infusion of Tregs alongside concurrent immunosuppressive drugs. However, it remains to be determined whether the presence of immunosuppressive drugs negatively impacts Treg function and stability. We tested in vitro and in vivo the effects of tacrolimus, mycophenolate and methylprednisolone (major ISDs used in transplantation) on ex vivo expanded, rapamycin-treated human Tregs. The in vitro results showed that these drugs had no effect on phenotype, function and stability of Tregs, although tacrolimus affected the expression of chemokine receptors and IL-10 production. However, viability and proliferative capacity were reduced in a dose-dependent manner by all the three drugs. The in vivo experiments using a humanized mouse model confirmed the in vitro results. However, treatment of mice with only rapamycin maintained the viability, function and proliferative ability of adoptively transferred Tregs. Taken together, our results suggest that the key functions of ex vivo expanded Tregs are not affected by a concurrent immunosuppressive therapy. However, the choice of the drug combination and their timing and dosing should be considered as an essential component to induce and maintain tolerance by Treg.
Collapse
Affiliation(s)
- Cristiano Scottà
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Giorgia Fanelli
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Sec Julie Hoong
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Marco Romano
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. & A. Seràgnoli", University of Bologna, Italy
| | - Estefania Nova Lamperti
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Mitalee Sukthankar
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Giuliana Guggino
- Dipartimento di Biopatologia e Biotecnologie Mediche, University of Palermo, Italy
| | - Henrieta Fazekasova
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Kulachelvy Ratnasothy
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Pablo D Becker
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Behdad Afzali
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institutes of Arthritis, and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Robert I Lechler
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Giovanna Lombardi
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| |
Collapse
|
28
|
|
29
|
Regulatory T Cells Resist Cyclosporine-Induced Cell Death via CD44-Mediated Signaling Pathways. Int J Cell Biol 2015; 2015:614297. [PMID: 26448755 PMCID: PMC4581548 DOI: 10.1155/2015/614297] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 01/19/2015] [Accepted: 01/19/2015] [Indexed: 12/20/2022] Open
Abstract
Cyclosporine A (CSA) is an immunosuppressive agent that specifically targets T cells and also increases the percentage of pro-tolerogenic CD4+Foxp3+ regulatory T cells (Treg) through unknown mechanisms. We previously reported that CD44, a receptor for the extracellular matrix glycosaminoglycan hyaluronan (HA), promotes Treg stability in IL-2-low environments. Here, we asked whether CD44 signaling also promotes Treg resistance to CSA. We found that CD44 cross-linking promoted Foxp3 expression and Treg viability in the setting of CSA treatment. This effect was IL-2 independent but could be suppressed using sc-355979, an inhibitor of Stat5-phosphorylation. Moreover, we found that inhibition of HA synthesis impairs Treg homeostasis but that this effect could be overcome with exogenous IL-2 or CD44-cross-linking. Together, these data support a model whereby CD44 cross-linking by HA promotes IL-2-independent Foxp3 expression and Treg survival in the face of CSA.
Collapse
|
30
|
Safinia N, Scotta C, Vaikunthanathan T, Lechler RI, Lombardi G. Regulatory T Cells: Serious Contenders in the Promise for Immunological Tolerance in Transplantation. Front Immunol 2015; 6:438. [PMID: 26379673 PMCID: PMC4553385 DOI: 10.3389/fimmu.2015.00438] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 08/12/2015] [Indexed: 01/12/2023] Open
Abstract
Regulatory T cells (Tregs) play an important role in immunoregulation and have been shown in animal models to promote transplantation tolerance and curb autoimmunity following their adoptive transfer. The safety and potential therapeutic efficacy of these cells has already been reported in Phase I trials of bone-marrow transplantation and type I diabetes, the success of which has motivated the broadened application of these cells in solid-organ transplantation. Despite major advances in the clinical translation of these cells, there are still key questions to be addressed to ensure that Tregs attest their reputation as ideal candidates for tolerance induction. In this review, we will discuss the unique traits of Tregs that have attracted such fame in the arena of tolerance induction. We will outline the protocols used for their ex vivo expansion and discuss the future directions of Treg cell therapy. In this regard, we will review the concept of Treg heterogeneity, the desire to isolate and expand a functionally superior Treg population and report on the effect of differing culture conditions. The relevance of Treg migratory capacity will also be discussed together with methods of in vivo visualization of the infused cells. Moreover, we will highlight key advances in the identification and expansion of antigen-specific Tregs and discuss their significance for cell therapy application. We will also summarize the clinical parameters that are of importance, alongside cell manufacture, from the choice of immunosuppression regimens to the number of injections in order to direct the success of future efficacy trials of Treg cell therapy. Years of research in the field of tolerance have seen an accumulation of knowledge and expertise in the field of Treg biology. This perpetual progression has been the driving force behind the many successes to date and has put us now within touching distance of our ultimate success, immunological tolerance.
Collapse
Affiliation(s)
- Niloufar Safinia
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, Faculty of Life Sciences and Medicine, King's College London , London , UK
| | - Cristiano Scotta
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, Faculty of Life Sciences and Medicine, King's College London , London , UK
| | - Trishan Vaikunthanathan
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, Faculty of Life Sciences and Medicine, King's College London , London , UK
| | - Robert I Lechler
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, Faculty of Life Sciences and Medicine, King's College London , London , UK
| | - Giovanna Lombardi
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, Faculty of Life Sciences and Medicine, King's College London , London , UK
| |
Collapse
|
31
|
Sabbatini M, Ruggiero G, Palatucci AT, Rubino V, Federico S, Giovazzino A, Apicella L, Santopaolo M, Matarese G, Galgani M, Terrazzano G. Oscillatory mTOR inhibition and Treg increase in kidney transplantation. Clin Exp Immunol 2015; 182:230-40. [PMID: 26077103 DOI: 10.1111/cei.12669] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2015] [Indexed: 01/13/2023] Open
Abstract
Intracellular metabolic pathways dependent upon the mammalian target of rapamycin (mTOR) play a key role in immune-tolerance control. In this study, we focused on long-term mTOR-dependent immune-modulating effects in kidney transplant recipients undergoing conversion from calcineurin inhibitors (CNI) to mTOR inhibitors (everolimus) in a 1-year follow-up. The conversion to everolimus is associated with a decrease of neutrophils and of CD8(+) T cells. In addition, we observed a reduced production of interferon (IFN)-γ by CD8(+) T cells and of interleukin (IL)-17 by CD4(+) T lymphocytes. An increase in CD4(+) CD25(+) forkhead box protein 3 (FoxP3)(+) [regulatory T cell [(Treg)] numbers was also seen. Treg increase correlated with a higher proliferation rate of this regulatory subpopulation when compared with the CD4(+) FoxP3(-) effector counterpart. Basal phosphorylation level of S6 kinase, a major mTOR-dependent molecular target, was substantially maintained in patients treated with everolimus. Moreover, oscillations in serum concentration of everolimus were associated with changes in basal and activation-dependent S6 kinase phosphorylation of CD4(+) and CD8(+) T cells. Indeed, T cell receptor (TCR) triggering was observed to induce significantly higher S6 kinase phosphorylation in the presence of lower everolimus serum concentrations. These results unveil the complex mTOR-dependent immune-metabolic network leading to long-term immune-modulation and might have relevance for novel therapeutic settings in kidney transplants.
Collapse
Affiliation(s)
- M Sabbatini
- Dipartimento di Sanità Pubblica, DH di Nefrologia e Trapianto di Rene, Università di Napoli 'Federico II', Napoli, Italy
| | - G Ruggiero
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli 'Federico II', Napoli, Italy
| | - A T Palatucci
- Dottorato di Scienze.,Dipartimento di Scienze, Università della Basilicata, Potenza, Italy
| | - V Rubino
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli 'Federico II', Napoli, Italy
| | - S Federico
- Dipartimento di Sanità Pubblica, DH di Nefrologia e Trapianto di Rene, Università di Napoli 'Federico II', Napoli, Italy
| | - A Giovazzino
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli 'Federico II', Napoli, Italy.,Dipartimento di Scienze, Università della Basilicata, Potenza, Italy
| | - L Apicella
- Dipartimento di Sanità Pubblica, DH di Nefrologia e Trapianto di Rene, Università di Napoli 'Federico II', Napoli, Italy
| | - M Santopaolo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli 'Federico II', Napoli Italy
| | - G Matarese
- Dipartimento di Medicina e Chirurgia, Università di Salerno, Salerno, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milano, Italy
| | - M Galgani
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
| | - G Terrazzano
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli 'Federico II', Napoli, Italy.,Dipartimento di Scienze, Università della Basilicata, Potenza, Italy
| |
Collapse
|
32
|
Montano-Loza AJ, Czaja AJ. Cell mediators of autoimmune hepatitis and their therapeutic implications. Dig Dis Sci 2015; 60:1528-42. [PMID: 25487192 DOI: 10.1007/s10620-014-3473-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 11/27/2014] [Indexed: 12/12/2022]
Abstract
Autoimmune hepatitis is associated with interactive cell populations of the innate and adaptive immune systems, and these populations are amenable to therapeutic manipulation. The goals of this review are to describe the key cell populations implicated in autoimmune hepatitis and to identify investigational opportunities to develop cell-directed therapies for this disease. Studies cited in PubMed from 1972 to 2014 for autoimmune hepatitis, innate and adaptive immune systems, and therapeutic interventions were examined. Dendritic cells can promote immune tolerance to self-antigens, present neo-antigens that enhance the immune response, and expand the regulatory T cell population. Natural killer cells can secrete pro-inflammatory and anti-inflammatory cytokines and modulate the activity of dendritic cells and antigen-specific T lymphocytes. T helper 2 lymphocytes can inhibit the cytotoxic activities of T helper 1 lymphocytes and limit the expansion of T helper 17 lymphocytes. T helper 17 lymphocytes can promote inflammatory activity, and they can also up-regulate genes that protect against oxidative stress and hepatocyte apoptosis. Natural killer T cells can expand the regulatory T cell population; gamma delta lymphocytes can secrete interleukin-10, stimulate hepatic regeneration, and induce the apoptosis of hepatic stellate cells; and antigen-specific regulatory T cells can dampen immune cell proliferation and function. Pharmacological agents, neutralizing antibodies, and especially the adoptive transfer of antigen-specific regulatory T cells that have been freshly generated ex vivo are evolving as management strategies. The cells within the innate and adaptive immune systems are key contributors to the occurrence of autoimmune hepatitis, and they are attractive therapeutic targets.
Collapse
Affiliation(s)
- Aldo J Montano-Loza
- Division of Gastroenterology and Liver Unit, University of Alberta Hospital, Edmonton, AB, Canada
| | | |
Collapse
|
33
|
Kim KW, Chung BH, Kim BM, Cho ML, Yang CW. The effect of mammalian target of rapamycin inhibition on T helper type 17 and regulatory T cell differentiation in vitro and in vivo in kidney transplant recipients. Immunology 2015; 144:68-78. [PMID: 24974886 DOI: 10.1111/imm.12351] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 06/03/2014] [Accepted: 06/24/2014] [Indexed: 12/20/2022] Open
Abstract
Sirolimus (SRL) is a promising alternative to calcineurin inhibitors, such as tacrolimus (TAC), in kidney transplant recipients (KTRs), but the immunological benefits of conversion from calcineurin inhibitors to SRL are not fully investigated. In the present study, we evaluated the effect of conversion from TAC to SRL on the T helper type 17/regulatory T (Th17/Treg) axis in three separate studies. First, the effect of SRL on the Th17/Treg axis was evaluated in vitro using peripheral blood mononuclear cells (PBMCs). Second, the effect of conversion from TAC to SRL on the Th17/Treg axis was studied in KTRs. Finally, the effect of SRL on CD8(+) Treg cells was evaluated. In vitro analysis of PBMCs isolated from KTRs showed that SRL suppressed Th17 cell differentiation but TAC did not. Conversion from TAC to SRL markedly decreased the number of effector memory CD8(+) T cells and significantly increased the proportion of CD4(+) and CD8(+) Treg cells compared with TAC in KTRs. SRL treatment induced the CD8(+) Treg cells, and these cells inhibited the proliferation of allogeneic CD4(+) T cells and Th17 cells. In conclusion, conversion from TAC to SRL favourably regulates Th17 and Treg cell differentiation in KTRs. These findings provide a rationale for conversion from TAC to SRL in KTRs.
Collapse
Affiliation(s)
- Kyoung Woon Kim
- Convergent Research Consortium for Immunologic Disease, Seoul St Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Seocho-gu, South Korea
| | | | | | | | | |
Collapse
|
34
|
Anderson KM, Olson KE, Estes KA, Flanagan K, Gendelman HE, Mosley RL. Dual destructive and protective roles of adaptive immunity in neurodegenerative disorders. Transl Neurodegener 2014; 3:25. [PMID: 25671101 PMCID: PMC4323229 DOI: 10.1186/2047-9158-3-25] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/28/2014] [Indexed: 12/14/2022] Open
Abstract
Inappropriate T cell responses in the central nervous system (CNS) affect the pathogenesis of a broad range of neuroinflammatory and neurodegenerative disorders that include, but are not limited to, multiple sclerosis, amyotrophic lateral sclerosis, Alzheimer’s disease and Parkinson’s disease. On the one hand immune responses can exacerbate neurotoxic responses; while on the other hand, they can lead to neuroprotective outcomes. The temporal and spatial mechanisms by which these immune responses occur and are regulated in the setting of active disease have gained significant recent attention. Spatially, immune responses that affect neurodegeneration may occur within or outside the CNS. Migration of antigen-specific CD4+ T cells from the periphery to the CNS and consequent immune cell interactions with resident glial cells affect neuroinflammation and neuronal survival. The destructive or protective mechanisms of these interactions are linked to the relative numerical and functional dominance of effector or regulatory T cells. Temporally, immune responses at disease onset or during progression may exhibit a differential balance of immune responses in the periphery and within the CNS. Immune responses with predominate T cell subtypes may differentially manifest migratory, regulatory and effector functions when triggered by endogenous misfolded and aggregated proteins and cell-specific stimuli. The final result is altered glial and neuronal behaviors that influence the disease course. Thus, discovery of neurodestructive and neuroprotective immune mechanisms will permit potential new therapeutic pathways that affect neuronal survival and slow disease progression.
Collapse
Affiliation(s)
- Kristi M Anderson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, The University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Katherine E Olson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, The University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Katherine A Estes
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, The University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Ken Flanagan
- Prothena Biosciences, South San Francisco, 650 Gateway Boulevard, CA 94080 USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, The University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - R Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, The University of Nebraska Medical Center, Omaha, NE 68198 USA
| |
Collapse
|
35
|
Abstract
INTRODUCTION Corticosteroids alone or in combination with azathioprine are the mainstay therapies of autoimmune hepatitis. Suboptimal responses (treatment failure, partial response, drug toxicity), frequent relapse after drug withdrawal, and the emergence of alternative immunosuppressive medications have fueled the pursuit of new treatments. The goals of this review are to present current management strategies and evolving interventions. AREAS COVERED PubMed searches from 1970 - 2014 provide the bases for this review. Corticosteroid regimens should be administered until resolution of symptoms, laboratory tests, and liver tissue abnormalities. Treatment failure warrants high doses of the original regimen, and relapse warrants re-treatment followed by long-term maintenance with azathioprine. The calcineurin inhibitors, budesonide, and mycophenolate mofetil are evolving as frontline therapies, and they may be considered as salvage therapies with the exception of budesonide. Rapamycin, rituximab, and infliximab have also rescued refractory patients but experiences are limited. Anti-oxidants, recombinant molecules, mAbs, and modulators of critical cell populations are key prospects. EXPERT OPINION Autoimmune hepatitis must be managed by multiple medications that supplement or supplant current regimens depending on the clinical situation. Rescue therapies will emerge as adjunctive interventions to minimize tissue damage (prevent fibrosis and hepatocyte apoptosis) and improve immune tolerance (regulatory T cell manipulations).
Collapse
Affiliation(s)
- Albert J Czaja
- Mayo Clinic College of Medicine, From the Division of Gastroenterology and Hepatology , 200 First Street S.W, Rochester, MN 55905 , USA +1 507 284 2691 ; +1 507 284 0538 ;
| |
Collapse
|
36
|
Cheng M, Xu H, Wang Y, Chen H, He B, Gao X, Li Y, Han J, Zhang Z. Glycyrrhetinic acid-modified chitosan nanoparticles enhanced the effect of 5-fluorouracil in murine liver cancer model via regulatory T-cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2013; 7:1287-99. [PMID: 24187487 PMCID: PMC3810199 DOI: 10.2147/dddt.s52809] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Modified chitosan nanoparticles are a promising platform for drug, such as 5-fluorouracil (5-FU), gene, and vaccine delivery. Here, we used chitosan and hepatoma cell-specific binding molecule glycyrrhetinic acid (GA) to synthesize glycyrrhetinic acid-modified chitosan (GA-CTS). The synthetic product was confirmed by infrared spectroscopy and hydrogen nuclear magnetic resonance. By combining GA-CTS and 5-FU, we obtained a GA-CTS/5-FU nanoparticle, with a particle size of 193.7 nm, drug loading of 1.56%, and a polydispersity index of 0.003. The GA-CTS/5-FU nanoparticle provided a sustained-release system comprising three distinct phases of quick, steady, and slow release. In vitro data indicated that it had a dose- and time-dependent anticancer effect. The effective drug exposure time against hepatic cancer cells was increased in comparison with that observed with 5-FU. In vivo studies on an orthotropic liver cancer mouse model demonstrated that GA-CTS/5-FU significantly inhibited cancer cell proliferation, resulting in increased survival time. The antitumor mechanisms for GA-CTS/5-FU nanoparticle were possibly associated with an increased expression of regulatory T-cells, decreased expression of cytotoxic T-cell and natural killer cells, and reduced levels of interleukin-2 and interferon gamma.
Collapse
Affiliation(s)
- Mingrong Cheng
- Department of General Surgery, Pudong New Area District Zhoupu Hospital, Shanghai, People's Republic of China ; Department of Endoscopy, Pudong New Area District Zhoupu Hospital, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Zachou K, Muratori P, Koukoulis GK, Granito A, Gatselis N, Fabbri A, Dalekos GN, Muratori L. Review article: autoimmune hepatitis -- current management and challenges. Aliment Pharmacol Ther 2013; 38:887-913. [PMID: 24010812 DOI: 10.1111/apt.12470] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 03/22/2013] [Accepted: 08/12/2013] [Indexed: 12/14/2022]
Abstract
BACKGROUND Autoimmune hepatitis (AIH) is a disease of unknown aetiology characterised by interface hepatitis, hypergammaglobulinaemia, circulating autoantibodies and a favourable response to immunosuppression. AIM To review recent advancements in understanding aetiopathogenesis, clinical, serological and histological features, diagnostic criteria and treatment strategies of AIH. METHODS Published studies on AIH extracted mainly from PubMed during the last 15 years. RESULTS Autoimmune hepatitis has a global distribution affecting any age, both sexes and all ethnic groups. Clinical manifestations are variable ranging from no symptoms to severe acute hepatitis and only seldom to fulminant hepatic failure. Autoimmune attack is perpetuated, possibly via molecular mimicry mechanisms, and favoured by the impaired control of regulatory T-cells. A typical laboratory finding is hypergammaglobulinaemia with selective elevation of IgG, although in 15-25% of patients - particularly children, elderly and acute cases - IgG levels are normal. Liver histology and autoantibodies, although not pathognomonic, still remain the hallmark for diagnosis. Immunosuppressive treatment is mandatory and life-saving; however, to meet strict response criteria, the conventional therapy with prednisolone with or without azathioprine is far from ideal. CONCLUSIONS Autoimmune hepatitis remains a major diagnostic and therapeutic challenge. The clinician, the hepato-pathologist and the laboratory personnel need to become more familiar with different expressions of the disease, interpretation of liver histology and autoimmune serology. According to the strict definition of treatment response issued by the 2010 AASLD guidelines, many patients are nonresponders to conventional treatment. Newer immunosuppressive agents targeting pathogenetic mechanisms can improve patient management, which needs to be tailored on a case-by-case basis.
Collapse
Affiliation(s)
- K Zachou
- Department of Medicine and Research Laboratory of Internal Medicine, Medical School, Thessaly University, Larissa, Greece
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Keijzer C, van der Zee R, van Eden W, Broere F. Treg inducing adjuvants for therapeutic vaccination against chronic inflammatory diseases. Front Immunol 2013; 4:245. [PMID: 23970886 PMCID: PMC3747555 DOI: 10.3389/fimmu.2013.00245] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Accepted: 08/06/2013] [Indexed: 01/21/2023] Open
Abstract
Many existing therapies in autoimmune diseases are based on systemic suppression of inflammation and the observed side effects of these therapies illustrate the pressing need for more specific interventions. Regulatory T-cells (Treg) are pivotal controllers of (auto-aggressive) immune responses and inflammation, and decreased Treg numbers and/or functioning have been associated with autoimmune disease. Therefore, Treg became frequently studied targets for more specific immunotherapy. Especially antigen-specific targeting of Treg would enable local and tailor made interventions, while obviating the negative side effect of general immuno-suppression. Self-antigens that participate in inflammation, irrespective of the etiology of the different autoimmune diseases, are held to be candidate antigens for antigen-specific interventions. Rather than tolerance induction to disease inciting self-antigens, which are frequently unknown, general self-antigens expressed at sites of inflammation would allow targeting of disease independent, but inflammatory-site specific, regulatory mechanisms. Preferably, such self-antigens should be abundantly expressed and up-regulated at the inflammatory-site. In this perspective heat shock proteins (Hsp) have several characteristics that make them highly attractive targets for antigen-specific Treg inducing therapy. The development of an antigen-specific Treg inducing vaccine is a major novel goal in the field of immunotherapy in autoimmune diseases. However, progress is hampered not only by the lack of effective antigens, but also by the fact that other factors such as dose, route, and the presence or absence of an adjuvant, turned out to be critical unknowns, with respect to the effective induction of Treg. In addition, the use of a Treg inducing adjuvant might be required to achieve an effective regulatory response, in the case of ongoing inflammation. Future goals in clinical trials will be the optimization of natural Treg expansion (or the induction of adaptive Treg) without loss of their suppressive function or the concomitant induction of non-regulatory T-cells. Here, we will discuss the potential use of protein/peptide-based vaccines combined with Treg inducing adjuvants for the development of therapeutic vaccines against chronic inflammatory conditions.
Collapse
Affiliation(s)
- Chantal Keijzer
- Immunology, Infectious Diseases and Immunology, Faculty Veterinary Medicine, University Utrecht , Utrecht , Netherlands
| | | | | | | |
Collapse
|
39
|
Wang XJ, Leveson-Gower D, Golab K, Wang LJ, Marek-Trzonkowska N, Krzystyniak A, Wardowska A, Millis JM, Trzonkowski P, Witkowski P. Influence of pharmacological immunomodulatory agents on CD4(+)CD25(high)FoxP3(+) T regulatory cells in humans. Int Immunopharmacol 2013; 16:364-70. [PMID: 23499512 DOI: 10.1016/j.intimp.2013.02.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Accepted: 02/18/2013] [Indexed: 12/24/2022]
Abstract
T regulatory cells (Tregs) play a critical role in the immunologic tolerance to the graft in transplantation. Thus, due to their immunosuppressive capability, ex vivo expanded Tregs may be used as a cellular therapy and an attractive novel strategy to control chronic rejection and eliminate need for lifelong pharmacological immunosuppression. Since Treg therapy is still in its infancy, initially Tregs still need to be applied in combination with pharmacological agents to prevent rejection. Fortunately, some of the medications have been shown to enhance the function and number of Tregs. In the clinic, different immunosuppressive regimens are used for individual patients for different types of organ transplantation. In this review, we present the most commonly used pharmacological agents for immunosuppression and discuss how they affect the Treg population. It is extremely difficult to dissect the effect of single agent on Tregs population in clinical settings since usually the combination of several medications is applied at the same time for graft protection. Nevertheless, experimental and clinical data indicate that thymoglobulin as immunosuppressive induction and mTOR inhibitors as immunosuppressive maintenance agents have the most beneficial effect on Treg population in the blood. Among supplemental agents promoting Tregs, anti-TNFα preparations have been in clinical use (in autoimmune diseases) for many years, so they are optimal candidates for testing in transplant settings in combination with Treg based cellular therapy.
Collapse
Affiliation(s)
- Xiao-Jun Wang
- Department of Surgery, Section of Transplantation, University of Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|