1
|
Short S, Lewik G, Issa F. An Immune Atlas of T Cells in Transplant Rejection: Pathways and Therapeutic Opportunities. Transplantation 2023; 107:2341-2352. [PMID: 37026708 PMCID: PMC10593150 DOI: 10.1097/tp.0000000000004572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 01/10/2023] [Accepted: 01/28/2023] [Indexed: 04/08/2023]
Abstract
Short-term outcomes in allotransplantation are excellent due to technical and pharmacological advances; however, improvement in long-term outcomes has been limited. Recurrent episodes of acute cellular rejection, a primarily T cell-mediated response to transplanted tissue, have been implicated in the development of chronic allograft dysfunction and loss. Although it is well established that acute cellular rejection is primarily a CD4 + and CD8 + T cell mediated response, significant heterogeneity exists within these cell compartments. During immune responses, naïve CD4 + T cells are activated and subsequently differentiate into specific T helper subsets under the influence of the local cytokine milieu. These subsets have distinct phenotypic and functional characteristics, with reported differences in their contribution to rejection responses specifically. Of particular relevance are the regulatory subsets and their potential to promote tolerance of allografts. Unraveling the specific contributions of these cell subsets in the context of transplantation is complex, but may reveal new avenues of therapeutic intervention for the prevention of rejection.
Collapse
Affiliation(s)
- Sarah Short
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, Oxfordshire, United Kingdom
| | - Guido Lewik
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, Oxfordshire, United Kingdom
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, Oxfordshire, United Kingdom
| |
Collapse
|
2
|
Ibrahim R, Saleh K, Chahine C, Khoury R, Khalife N, Cesne AL. LAG-3 Inhibitors: Novel Immune Checkpoint Inhibitors Changing the Landscape of Immunotherapy. Biomedicines 2023; 11:1878. [PMID: 37509517 PMCID: PMC10377063 DOI: 10.3390/biomedicines11071878] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/25/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023] Open
Abstract
One of the most important steps forward in the management of cancer was the discovery of immunotherapy. It has become an essential pillar in the treatment paradigm of cancer patients. Unfortunately, despite the various options presented with immune checkpoint inhibitors (ICIs), the benefit is still limited to select patients and the vast majority of these patients gain either minimal benefit or eventually progress, leaving an unmet need for the development of novel therapeutic agents and strategies. Lymphocyte activation gene-3 (LAG-3), an immune checkpoint receptor protein, is a molecule found on the surface of activated T-cells. It plays a major role in negatively regulating T-cell function thereby providing tumors with an immune escape in the tumor microenvironment (TME). Given its importance in regulating the immune system, LAG-3 has been considered as a promising target in oncology and precision medicine. To date, two LAG-3-directed agents (eftilagimod alpha and relatlimab) have been approved in combination with programmed death-1 (PD-1) inhibitors in the setting of advanced solid tumors. In this review, we discuss the structure of LAG-3, its mechanism of action, and its interaction with its ligands. We also shed light on the emerging treatments targeting LAG-3 for the treatment of solid tumors.
Collapse
Affiliation(s)
- Rebecca Ibrahim
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Khalil Saleh
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Claude Chahine
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Rita Khoury
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Nadine Khalife
- Department of head and neck Oncology, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Axel Le Cesne
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| |
Collapse
|
3
|
Liu G, Zhang Z, Wu Y, Feng J, Lan Y, Dong D, Liu Y, Yuan H, Tai G, Li S, Ni W. Anti-PD-L1 antibody reverses the immune tolerance induced by multiple MUC1-MBP vaccine immunizations by increasing the CD80/PD-L1 ratio, resulting in DC maturation, and decreasing Treg activity in B16-MUC1 melanoma-bearing mice. Int Immunopharmacol 2023; 121:110487. [PMID: 37364328 DOI: 10.1016/j.intimp.2023.110487] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/28/2023] [Accepted: 06/09/2023] [Indexed: 06/28/2023]
Abstract
In this study, we explored the possible mechanism of tumor tolerance induced by multiple repeated immunizations with a tumor vaccine (MUC1-MBP fusion protein plus CpG2006). We first analyzed the mechanism of tolerance by immunizing tumor-bearing mice 2, 5, or 8 times and found that compared with five immunizations with the M-M vaccine, eight immunizations increased tumor volume and weight and Treg levels, while the proportions of Th1 and Tc1 cells in the spleen and lymph nodes were decreased. In particular, the M-M vaccine induced PD-L1 expression in CD11c + DCs and decreased their CD80/PD-L1 ratio. Therefore, the mechanism of tolerance induction by multiple immunizations with the M-M vaccine was investigated by focusing on the CD80/PD-L1 ratio, and an anti-PD-L1 antibody (αPD-L1) and the M-M vaccine were used in combination to treat melanoma. The results showed that αPD-L1 increased the CD80/PD-L1 ratio and enhanced the maturation of cDC1s by blocking PD-L1 on DCs, which potentially increased the activity of Th1 and Tc1 cells. Furthermore, the combination of the M-M vaccine with αPD-L1 decreased the activity and proportion of Tregs, which reversed the immune tolerance induced by eight immunizations with the vaccine. This study reveals the mechanism of the combination of M-M and αPD-L1 and provides a new combination strategy for improving the therapeutic effect of the M-M vaccine, laying a theoretical basis for the clinical application of the vaccine.
Collapse
Affiliation(s)
- Guomu Liu
- Department of Dermatology and Venereology, The First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Zenan Zhang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yixuan Wu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Jingyue Feng
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yue Lan
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Dai Dong
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yu Liu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Hongyan Yuan
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Guixiang Tai
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Shanshan Li
- Department of Dermatology and Venereology, The First Hospital of Jilin University, Changchun 130021, Jilin, China.
| | - Weihua Ni
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
4
|
Lajiness JD, Cook-Mills JM. Catching Our Breath: Updates on the Role of Dendritic Cell Subsets in Asthma. Adv Biol (Weinh) 2023; 7:e2200296. [PMID: 36755197 PMCID: PMC10293089 DOI: 10.1002/adbi.202200296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/04/2023] [Indexed: 02/10/2023]
Abstract
Dendritic cells (DCs), as potent antigen presenting cells, are known to play a central role in the pathophysiology of asthma. The understanding of DC biology has evolved over the years to include multiple subsets of DCs with distinct functions in the initiation and maintenance of asthma. Furthermore, asthma is increasingly recognized as a heterogeneous disease with potentially diverse underlying mechanisms. The goal of this review is to summarize the role of DCs and the various subsets therein in the pathophysiology of asthma and highlight some of the crucial animal models shaping the field today. Potential future avenues of investigation to address existing gaps in knowledge are discussed.
Collapse
Affiliation(s)
- Jacquelyn D Lajiness
- Department of Pediatrics, Division of Neonatology, Indiana University School of Medicine, 1030 West Michigan Street, Suite C 4600, Indianapolis, IN, 46202-5201, USA
| | - Joan M Cook-Mills
- Department of Pediatrics, Department of Microbiology and Immunology, Pediatric Pulmonary, Asthma, and Allergy Basic Research Program, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W. Walnut Street, R4-202A, Indianapolis, IN, 46202, USA
| |
Collapse
|
5
|
Mutascio S, Mota T, Franchitti L, Sharma AA, Willemse A, Bergstresser SN, Wang H, Statzu M, Tharp GK, Weiler J, Sékaly RP, Bosinger SE, Paiardini M, Silvestri G, Jones RB, Kulpa DA. CD8 + T cells promote HIV latency by remodeling CD4 + T cell metabolism to enhance their survival, quiescence, and stemness. Immunity 2023; 56:1132-1147.e6. [PMID: 37030290 PMCID: PMC10880039 DOI: 10.1016/j.immuni.2023.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 11/16/2022] [Accepted: 03/15/2023] [Indexed: 04/10/2023]
Abstract
HIV infection persists during antiretroviral therapy (ART) due to a reservoir of latently infected cells that harbor replication-competent virus and evade immunity. Previous ex vivo studies suggested that CD8+ T cells from people with HIV may suppress HIV expression via non-cytolytic mechanisms, but the mechanisms responsible for this effect remain unclear. Here, we used a primary cell-based in vitro latency model and demonstrated that co-culture of autologous activated CD8+ T cells with HIV-infected memory CD4+ T cells promoted specific changes in metabolic and/or signaling pathways resulting in increased CD4+ T cell survival, quiescence, and stemness. Collectively, these pathways negatively regulated HIV expression and ultimately promoted the establishment of latency. As shown previously, we observed that macrophages, but not B cells, promoted latency in CD4+ T cells. The identification of CD8-specific mechanisms of pro-latency activity may favor the development of approaches to eliminate the viral reservoir in people with HIV.
Collapse
Affiliation(s)
- Simona Mutascio
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Talia Mota
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lavinia Franchitti
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Ashish A Sharma
- Department of Pathology & Laboratory Medicine, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Abigail Willemse
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | | | - Hong Wang
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Maura Statzu
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Gregory K Tharp
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Jared Weiler
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Rafick-Pierre Sékaly
- Department of Pathology & Laboratory Medicine, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Steven E Bosinger
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Pathology & Laboratory Medicine, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Mirko Paiardini
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Pathology & Laboratory Medicine, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Guido Silvestri
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Pathology & Laboratory Medicine, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - R Brad Jones
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Deanna A Kulpa
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Pathology & Laboratory Medicine, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
6
|
Morita N, Hoshi M, Tezuka H, Ando T, Yoshida S, Sato F, Yokoi H, Ito H, Saito K. CD8+ Regulatory T Cells Induced by Lipopolysaccharide Improve Mouse Endotoxin Shock. Immunohorizons 2023; 7:353-363. [PMID: 37212786 PMCID: PMC10579971 DOI: 10.4049/immunohorizons.2200074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 05/03/2023] [Indexed: 05/23/2023] Open
Abstract
Sepsis is a systemic inflammatory disease caused by a bacterial infection that leads to severe mortality, especially in elderly patients, because of an excessive immune response and impaired regulatory functions. Antibiotic treatment is widely accepted as the first-line therapy for sepsis; however, its excessive use has led to the emergence of multidrug-resistant bacteria in patients with sepsis. Therefore, immunotherapy may be effective in treating sepsis. Although CD8+ regulatory T cells (Tregs) are known to have immunomodulatory effects in various inflammatory diseases, their role during sepsis remains unclear. In this study, we investigated the role of CD8+ Tregs in an LPS-induced endotoxic shock model in young (8-12 wk old) and aged (18-20 mo old) mice. The adoptive transfer of CD8+ Tregs into LPS-treated young mice improved the survival rate of LPS-induced endotoxic shock. Moreover, the number of CD8+ Tregs in LPS-treated young mice increased through the induction of IL-15 produced by CD11c+ cells. In contrast, LPS-treated aged mice showed a reduced induction of CD8+ Tregs owing to the limited production of IL-15. Furthermore, CD8+ Tregs induced by treatment with the rIL-15/IL-15Rα complex prevented LPS-induced body wight loss and tissue injury in aged mice. In this study, to our knowledge, the induction of CD8+ Tregs as novel immunotherapy or adjuvant therapy for endotoxic shock might reduce the uncontrolled immune response and ultimately improve the outcomes of endotoxic shock.
Collapse
Affiliation(s)
- Nanaka Morita
- Department of Disease Control and Prevention, Fujita Health University, Toyoake, Aichi, Japan
| | - Masato Hoshi
- Department of Disease Control and Prevention, Fujita Health University, Toyoake, Aichi, Japan
- Department of Informative Clinical Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Hiroyuki Tezuka
- Cellular Function Analysis, Research Promotion Headquarters, Fujita Health University, Toyoake, Aichi, Japan
| | - Tatsuya Ando
- Joint Research Laboratory of Clinical Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Sayaka Yoshida
- Department of Informative Clinical Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Fumiaki Sato
- Department of Informative Clinical Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Hiroyuki Yokoi
- Department of Informative Clinical Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Hiroyasu Ito
- Cellular Function Analysis, Research Promotion Headquarters, Fujita Health University, Toyoake, Aichi, Japan
| | - Kuniaki Saito
- Department of Disease Control and Prevention, Fujita Health University, Toyoake, Aichi, Japan
| |
Collapse
|
7
|
Sharma RK, Sharma J, Kumar R, Badal D, Pattekar A, Sehgal S, Gupta A, Jain P, Sachdeva N. TLR9 signalling activation via direct ligation and its functional consequences in CD4 + T cells. Scand J Immunol 2022; 96:e13214. [PMID: 37406035 PMCID: PMC9788197 DOI: 10.1111/sji.13214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 08/11/2022] [Accepted: 08/23/2022] [Indexed: 11/29/2022]
Abstract
CpG Oligodeoxynucleotides (ODNs) are established TLR9 ligands; however, their functional responses in CD4+ T cells are believed to be independent of TLR9 and MyD88. We studied ligand-receptor interactions of ODN 2216 and TLR9 in human CD4+ T cells and assessed their consequences in terms of TLR9 signalling and cell phenotype. We demonstrated that the uptake of ODN 2216, a synthetic TLR9 agonist, is controlled by TLR9 signalling molecules and results in an increase in the expression of TLR9 signalling molecules, regulated via a feedback mechanism. Next, the uptake of ODN 2216 resulted in TLR9 signalling dependent but MyD88 independent increase in expression of TGF-β. Finally, ODN 2216 treated CD4+ T cells showed an anti-inflammatory phenotype that was similar to Th3 type of regulatory T cells. These Th3-like cells were able to suppress the proliferation of untreated CD4+ T cells. Collectively, our results demonstrate a direct and interdependent relationship between ODN 2216 uptake and TLR9 signalling in CD4+ T cells. Our findings thus pave the way for future research to explore direct modulation of adaptive immune cells, using innate immune ligands, to subvert exaggerated inflammatory responses.
Collapse
Affiliation(s)
- Ravi Kumar Sharma
- Advanced Eye CentrePost Graduate Institute of Medical Education and Research (PGIMER)ChandigarhIndia
- Department of Microbiology and Immunology and the Institute for Molecular Medicine and Infectious DiseaseDrexel University College of MedicinePhiladelphiaPennsylvaniaUSA
- Division of Rheumatology, Department of MedicineKarolinska InstitutetSolnaSweden
| | - Jyoti Sharma
- Advanced Eye CentrePost Graduate Institute of Medical Education and Research (PGIMER)ChandigarhIndia
| | - Rajendra Kumar
- Division of Biological SciencesIndian Institute of Science Education and ResearchMohaliPunjabIndia
- Department of OncologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Darshan Badal
- Department of EndocrinologyPost Graduate Institute of Medical Education and Research (PGIMER)ChandigarhIndia
| | - Ajinkya Pattekar
- Department of Microbiology and Immunology and the Institute for Molecular Medicine and Infectious DiseaseDrexel University College of MedicinePhiladelphiaPennsylvaniaUSA
| | - Shobha Sehgal
- Department of ImmunopathologyPost Graduate Institute of Medical Education and Research (PGIMER)ChandigarhIndia
| | - Amod Gupta
- Advanced Eye CentrePost Graduate Institute of Medical Education and Research (PGIMER)ChandigarhIndia
| | - Pooja Jain
- Department of Microbiology and Immunology and the Institute for Molecular Medicine and Infectious DiseaseDrexel University College of MedicinePhiladelphiaPennsylvaniaUSA
| | - Naresh Sachdeva
- Department of EndocrinologyPost Graduate Institute of Medical Education and Research (PGIMER)ChandigarhIndia
| |
Collapse
|
8
|
Yero A, Shi T, Routy JP, Tremblay C, Durand M, Costiniuk CT, Jenabian MA. FoxP3+ CD8 T-cells in acute HIV infection and following early antiretroviral therapy initiation. Front Immunol 2022; 13:962912. [PMID: 35967314 PMCID: PMC9372390 DOI: 10.3389/fimmu.2022.962912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 06/29/2022] [Indexed: 12/21/2022] Open
Abstract
ObjectivesBesides CD4 regulatory T-cells (Tregs), immunosuppressor FoxP3+ CD8 T-cells are emerging as an important subset of Tregs, which contribute to immune dysfunction and disease progression in HIV infection. However, FoxP3+ CD8 T-cell dynamics in acute HIV infection and following early antiretroviral therapy (ART) initiation remain understudied.MethodsSubsets of FoxP3+ CD8 T-cells were characterized both prospectively and cross-sectionally in PBMCs from untreated acute (n=26) and chronic (n=10) HIV-infected individuals, early ART-treated in acute infection (n=10, median of ART initiation: 5.5 months post-infection), ART-treated in chronic infection (n=10), elite controllers (n=18), and HIV-uninfected controls (n=21).ResultsAcute and chronic infection were associated with increased total, effector memory, and terminally differentiated FoxP3+ CD8 T-cells, while early ART normalized only the frequencies of total FoxP3+ CD8 T-cells. We observed an increase in FoxP3+ CD8 T-cell immune activation (HLADR+/CD38+), senescence (CD57+/CD28-), and PD-1 expression during acute and chronic infection, which were not normalized by early ART. FoxP3+ CD8 T-cells in untreated participants expressed higher levels of immunosuppressive LAP(TGF-β1) and CD39 than uninfected controls, whereas early ART did not affect their expression. The expression of gut-homing markers CCR9 and Integrin-β7 by total FoxP3+ CD8 T-cells and CD39+ and LAP(TGF-β1)+ FoxP3+ CD8 T-cells increased in untreated individuals and remained higher than in uninfected controls despite early ART. Elite controllers share most of the FoxP3+ CD8 T-cell characteristics in uninfected individuals.ConclusionsAlthough early ART normalized total FoxP3+ CD8 T-cells frequencies, it did not affect the persistent elevation of the gut-homing potential of CD39+ and LAP(TGF-β1)+ FoxP3+ CD8 T-cell, which may contribute to immune dysfunction.
Collapse
Affiliation(s)
- Alexis Yero
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC, Canada
| | - Tao Shi
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC, Canada
| | - Jean-Pierre Routy
- Research Institute of McGill University Health Centre, Montreal, QC, Canada
- Chronic Viral Illness Service, Department of Medicine, Glen Site, McGill University Health Centre, Montreal, QC, Canada
| | - Cécile Tremblay
- Centre hospitalier de l'Université de Montréal (CHUM) Research Centre, Montreal, QC, Canada
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Madeleine Durand
- Centre hospitalier de l'Université de Montréal (CHUM) Research Centre, Montreal, QC, Canada
| | - Cecilia T. Costiniuk
- Research Institute of McGill University Health Centre, Montreal, QC, Canada
- Chronic Viral Illness Service, Department of Medicine, Glen Site, McGill University Health Centre, Montreal, QC, Canada
| | - Mohammad-Ali Jenabian
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC, Canada
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- *Correspondence: Mohammad-Ali Jenabian,
| |
Collapse
|
9
|
Bolivar-Wagers S, Larson JH, Jin S, Blazar BR. Cytolytic CD4 + and CD8 + Regulatory T-Cells and Implications for Developing Immunotherapies to Combat Graft-Versus-Host Disease. Front Immunol 2022; 13:864748. [PMID: 35493508 PMCID: PMC9040077 DOI: 10.3389/fimmu.2022.864748] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/16/2022] [Indexed: 02/03/2023] Open
Abstract
Regulatory T-cells (Treg) are critical for the maintenance of immune homeostasis and tolerance induction. While the immunosuppressive mechanisms of Treg have been extensively investigated for decades, the mechanisms responsible for Treg cytotoxicity and their therapeutic potential in regulating immune responses have been incompletely explored and exploited. Conventional cytotoxic T effector cells (Teffs) are known to be important for adaptive immune responses, particularly in the settings of viral infections and cancer. CD4+ and CD8+ Treg subsets may also share similar cytotoxic properties with conventional Teffs. Cytotoxic effector Treg (cyTreg) are a heterogeneous population in the periphery that retain the capacity to suppress T-cell proliferation and activation, induce cellular apoptosis, and migrate to tissues to ensure immune homeostasis. The latter can occur through several cytolytic mechanisms, including the Granzyme/Perforin and Fas/FasL signaling pathways. This review focuses on the current knowledge and recent advances in our understanding of cyTreg and their potential application in the treatment of human disease, particularly Graft-versus-Host Disease (GVHD).
Collapse
Affiliation(s)
| | | | | | - Bruce R. Blazar
- Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
10
|
Fu J, Lehmann CHK, Wang X, Wahlbuhl M, Allabauer I, Wilde B, Amon L, Dolff S, Cesnjevar R, Kribben A, Woelfle J, Rascher W, Hoyer PF, Dudziak D, Witzke O, Hoerning A. CXCR4 blockade reduces the severity of murine heart allograft rejection by plasmacytoid dendritic cell-mediated immune regulation. Sci Rep 2021; 11:23815. [PMID: 34893663 PMCID: PMC8664946 DOI: 10.1038/s41598-021-03115-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 11/08/2021] [Indexed: 12/20/2022] Open
Abstract
Allograft-specific regulatory T cells (Treg cells) are crucial for long-term graft acceptance after transplantation. Although adoptive Treg cell transfer has been proposed, major challenges include graft-specificity and stability. Thus, there is an unmet need for the direct induction of graft-specific Treg cells. We hypothesized a synergism of the immunotolerogenic effects of rapamycin (mTOR inhibition) and plerixafor (CXCR4 antagonist) for Treg cell induction. Thus, we performed fully-mismatched heart transplantations and found combination treatment to result in prolonged allograft survival. Moreover, fibrosis and myocyte lesions were reduced. Although less CD3+ T cell infiltrated, higher Treg cell numbers were observed. Noteworthy, this was accompanied by a plerixafor-dependent plasmacytoid dendritic cells-(pDCs)-mobilization. Furthermore, in vivo pDC-depletion abrogated the plerixafor-mediated Treg cell number increase and reduced allograft survival. Our pharmacological approach allowed to increase Treg cell numbers due to pDC-mediated immune regulation. Therefore pDCs can be an attractive immunotherapeutic target in addition to plerixafor treatment.
Collapse
Affiliation(s)
- Jian Fu
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany.,Department for Pediatric and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Loschgestrasse 15, 91054, Erlangen, Germany.,The Emergency and Trauma Center, The First Affiliated Hospital of Hai Nan Medical University, Haikou, China
| | - Christian H K Lehmann
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Research Module II, Hartmannstr. 14, 91052, Erlangen, Germany. .,Medical Immunology Campus and German Centre for Immuntherapy (Deutsches Zentrum für Immuntherapie-DZI) Erlangen, FAU Erlangen-Nürnberg, 91054, Erlangen, Germany.
| | - Xinning Wang
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany.,The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Mandy Wahlbuhl
- Department for Pediatric and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Loschgestrasse 15, 91054, Erlangen, Germany
| | - Ida Allabauer
- Department for Pediatric and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Loschgestrasse 15, 91054, Erlangen, Germany
| | - Benjamin Wilde
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Lukas Amon
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Research Module II, Hartmannstr. 14, 91052, Erlangen, Germany
| | - Sebastian Dolff
- Department of Infectious Diseases, West German Centre of Infectious Diseases, Universitätsmedizin Essen, University Duisburg-Essen, Essen, Germany
| | - Robert Cesnjevar
- Department of Pediatric Cardiac Surgery, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany.,Department of Cardiac Surgery, Universitäts-Kinderspital Zürich, Zurich, Switzerland
| | - Andreas Kribben
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Joachim Woelfle
- Department for Pediatric and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Loschgestrasse 15, 91054, Erlangen, Germany
| | - Wolfgang Rascher
- Department for Pediatric and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Loschgestrasse 15, 91054, Erlangen, Germany
| | - Peter F Hoyer
- Department of Pediatrics II, Pediatric Nephrology, Gastroenterology, Endocrinology and Transplant Medicine, Children's Hospital Essen, University Duisburg-Essen, Duisburg, Germany
| | - Diana Dudziak
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Research Module II, Hartmannstr. 14, 91052, Erlangen, Germany.,Medical Immunology Campus and German Centre for Immuntherapy (Deutsches Zentrum für Immuntherapie-DZI) Erlangen, FAU Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Oliver Witzke
- Department of Infectious Diseases, West German Centre of Infectious Diseases, Universitätsmedizin Essen, University Duisburg-Essen, Essen, Germany
| | - André Hoerning
- Department for Pediatric and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Loschgestrasse 15, 91054, Erlangen, Germany. .,Department of Pediatrics II, Pediatric Nephrology, Gastroenterology, Endocrinology and Transplant Medicine, Children's Hospital Essen, University Duisburg-Essen, Duisburg, Germany.
| |
Collapse
|
11
|
Capitani N, Patrussi L, Baldari CT. Nature vs. Nurture: The Two Opposing Behaviors of Cytotoxic T Lymphocytes in the Tumor Microenvironment. Int J Mol Sci 2021; 22:ijms222011221. [PMID: 34681881 PMCID: PMC8540886 DOI: 10.3390/ijms222011221] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/14/2021] [Accepted: 10/16/2021] [Indexed: 11/16/2022] Open
Abstract
Similar to Janus, the two-faced god of Roman mythology, the tumor microenvironment operates two opposing and often conflicting activities, on the one hand fighting against tumor cells, while on the other hand, favoring their proliferation, survival and migration to other sites to establish metastases. In the tumor microenvironment, cytotoxic T cells-the specialized tumor-cell killers-also show this dual nature, operating their tumor-cell directed killing activities until they become exhausted and dysfunctional, a process promoted by cancer cells themselves. Here, we discuss the opposing activities of immune cells populating the tumor microenvironment in both cancer progression and anti-cancer responses, with a focus on cytotoxic T cells and on the molecular mechanisms responsible for the efficient suppression of their killing activities as a paradigm of the power of cancer cells to shape the microenvironment for their own survival and expansion.
Collapse
|
12
|
Pellegrino B, Hlavata Z, Migali C, De Silva P, Aiello M, Willard-Gallo K, Musolino A, Solinas C. Luminal Breast Cancer: Risk of Recurrence and Tumor-Associated Immune Suppression. Mol Diagn Ther 2021; 25:409-424. [PMID: 33974235 PMCID: PMC8249273 DOI: 10.1007/s40291-021-00525-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2021] [Indexed: 12/24/2022]
Abstract
Hormone-receptor positive (HR+) breast cancer (BC) (including the luminal A and the luminal B subtypes) is the most common type of tumor in women diagnosed with early-stage BC (EBC). It represents a highly heterogeneous subgroup that is characterized by different risks of relapse. The aim of this review is to discuss the possible role played by the immune response in predicting this risk, along with the most common clinical and pathological factors and molecular tools that have been developed and are already in use. As opposed to what has previously been observed in the most aggressive human epidermal growth factor receptor 2 (HER2)-positive and triple-negative breast cancer (TNBC) subtypes, a high proportion of tumor-infiltrating lymphocytes (TILs)-reflecting a spontaneous and pre-existing immune response to the tumor-has been linked to a worse prognosis in HR+ EBC. This work provides some immune biological rationale explaining these findings and provides the basics to understand the principal clinical trials that are testing immunotherapy in HR+ (luminal) BC.
Collapse
Affiliation(s)
- Benedetta Pellegrino
- Department of Medicine and Surgery, University of Parma, Str. dell’Università, 12, 43121 Parma, PR Italy
- Medical Oncology and Breast Unit, University Hospital of Parma, Parma, Italy
| | - Zuzana Hlavata
- Department of Medical Oncology, CHR Mons-Hainaut, Avenue Baudouin de Constantinople, n. 5, Mons, Hainaut Belgium
| | | | - Pushpamali De Silva
- Wellman Center for Photomedicine, Department of Dermatology, Harvard Medical School and Massachusetts General Hospital, Boston, MA USA
| | - Marco Aiello
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria Policlinico San Marco, Catania, Italy
| | | | - Antonino Musolino
- Department of Medicine and Surgery, University of Parma, Str. dell’Università, 12, 43121 Parma, PR Italy
- Medical Oncology and Breast Unit, University Hospital of Parma, Parma, Italy
| | - Cinzia Solinas
- Azienda Tutela della Salute Sardegna, Ospedale A. Segni, Ozieri, Italy
| |
Collapse
|
13
|
Montamat G, Leonard C, Poli A, Klimek L, Ollert M. CpG Adjuvant in Allergen-Specific Immunotherapy: Finding the Sweet Spot for the Induction of Immune Tolerance. Front Immunol 2021; 12:590054. [PMID: 33708195 PMCID: PMC7940844 DOI: 10.3389/fimmu.2021.590054] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/04/2021] [Indexed: 01/16/2023] Open
Abstract
Prevalence and incidence of IgE-mediated allergic diseases have increased over the past years in developed and developing countries. Allergen-specific immunotherapy (AIT) is currently the only curative treatment available for allergic diseases that has long-term efficacy. Although AIT has been proven successful as an immunomodulatory therapy since its beginnings, it still faces several unmet needs and challenges today. For instance, some patients can experience severe side effects, others are non-responders, and prolonged treatment schedules can lead to lack of patient adherence and therapy discontinuation. A common strategy to improve AIT relies on the use of adjuvants and immune modulators to boost its effects and improve its safety. Among the adjuvants tested for their clinical efficacy, CpG oligodeoxynucleotide (CpG-ODN) was investigated with limited success and without reaching phase III trials for clinical allergy treatment. However, recently discovered immune tolerance-promoting properties of CpG-ODN place this adjuvant again in a prominent position as an immune modulator for the treatment of allergic diseases. Indeed, it has been shown that the CpG-ODN dose and concentration are crucial in promoting immune regulation through the recruitment of pDCs. While low doses induce an inflammatory response, high doses of CpG-ODN trigger a tolerogenic response that can reverse a pre-established allergic milieu. Consistently, CpG-ODN has also been found to stimulate IL-10 producing B cells, so-called B regulatory cells (Bregs). Accordingly, CpG-ODN has shown its capacity to prevent and revert allergic reactions in several animal models showing its potential as both preventive and active treatment for IgE-mediated allergy. In this review, we describe how CpG-ODN-based therapies for allergic diseases, despite having shown limited success in the past, can still be exploited further as an adjuvant or immune modulator in the context of AIT and deserves additional attention. Here, we discuss the past and current knowledge, which highlights CpG-ODN as a potential adjuvant to be reevaluated for the enhancement of AIT when used in appropriate conditions and formulations.
Collapse
Affiliation(s)
- Guillem Montamat
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Cathy Leonard
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Aurélie Poli
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Ludger Klimek
- Centre for Rhinology and Allergology, Wiesbaden, Germany
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Department of Dermatology and Allergy Centre, Odense University Hospital, Odense, Denmark
| |
Collapse
|
14
|
De Silva P, Aiello M, Gu-Trantien C, Migliori E, Willard-Gallo K, Solinas C. Targeting CTLA-4 in cancer: Is it the ideal companion for PD-1 blockade immunotherapy combinations? Int J Cancer 2020; 149:31-41. [PMID: 33252786 DOI: 10.1002/ijc.33415] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 10/25/2020] [Accepted: 10/28/2020] [Indexed: 12/19/2022]
Abstract
Immunotherapy approaches boosting spontaneous and durable antitumor immune responses through immune checkpoint blockade are revolutionizing treatment and patient outcomes in solid tumors and hematological malignancies. Among the various inhibitory molecules employed by the immune system to regulate the adaptive immune responses, cytotoxic T lymphocyte antigen-4 (CTLA-4) is the first successfully targeted immune checkpoint molecule in the clinic, giving rise to significant but selective benefit either when targeted alone or in combination with anti-programmed cell death protein-1 (PD-1) antibodies (Abs). However, the use of anti-CTLA-4 Abs was associated with the incidence of autoimmune-like adverse events (AEs), which were particularly frequent and severe with the use of combinational strategies. Nevertheless, the higher incidence of AEs is associated with an improved clinical benefit indicating treatment response. A prompt recognition of AEs followed by early and adequate treatment with immunosuppressive agents allows the management of these potentially serious AEs. This narrative review aims to summarize CTLA-4 biology, the rationale for the use as a companion for anti-PD-1 Abs in humans with results from the most relevant Phase III clinical trials including anti-CTLA-4 Abs in combination with anti-PD-1 Abs in solid tumors.
Collapse
Affiliation(s)
- Pushpamali De Silva
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Marco Aiello
- Medical Oncology Unit A.O.U. Policlinico, Vittorio Emanuele di Catania, Catania, Italy
| | - Chunyan Gu-Trantien
- Institute of Medical Immunology, Université Libre de Bruxelles, Brussels, Belgium
| | - Edoardo Migliori
- Columbia University Medical Center, Columbia Center for Translational Immunology, New York, New York, USA
| | | | - Cinzia Solinas
- Regional Hospital of Valle d'Aosta, Azienda U.S.L. Valle d'Aosta, Aosta, Italy
| |
Collapse
|
15
|
Flippe L, Bézie S, Anegon I, Guillonneau C. Future prospects for CD8 + regulatory T cells in immune tolerance. Immunol Rev 2019; 292:209-224. [PMID: 31593314 PMCID: PMC7027528 DOI: 10.1111/imr.12812] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CD8+ Tregs have been long described and significant progresses have been made about their phenotype, their functional mechanisms, and their suppressive ability compared to conventional CD4+ Tregs. They are now at the dawn of their clinical use. In this review, we will summarize their phenotypic characteristics, their mechanisms of action, the similarities, differences and synergies between CD8+ and CD4+ Tregs, and we will discuss the biology, development and induction of CD8+ Tregs, their manufacturing for clinical use, considering open questions/uncertainties and future technically accessible improvements notably through genetic modifications.
Collapse
Affiliation(s)
- Léa Flippe
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Séverine Bézie
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Ignacio Anegon
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Carole Guillonneau
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| |
Collapse
|
16
|
Abstract
Although cluster of differentiation (CD)8 regulatory T (Treg) cells have been in the last 20 years more studied since evidences of their role in tolerance as been demonstrated in transplantation, autoimmune diseases and cancer, their characteristics are still controversial. In this review, we will focus on recent advances on CD8 Treg cells and description of a role for CD8 Treg cells in tolerance in both solid organ transplantation and graft-versus-host disease and their potential for clinical trials.
Collapse
|
17
|
Boor PPC, Bosma BM, Tran KTC, van der Laan LJW, Hagenaars H, IJzermans JNM, Metselaar HJ, Kwekkeboom J. Characterization of Antigen-Presenting Cell Subsets in Human Liver-Draining Lymph Nodes. Front Immunol 2019; 10:441. [PMID: 30930897 PMCID: PMC6428028 DOI: 10.3389/fimmu.2019.00441] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 02/19/2019] [Indexed: 12/14/2022] Open
Abstract
T-cell immunity in the liver is tightly regulated to prevent chronic liver inflammation in response to antigens and toxins derived from food and intestinal bacterial flora. Since the main sites of T cell activation in response to foreign components entering solid tissues are the draining lymph nodes (LN), we aimed to study whether Antigen-Presenting Cell (APC) subsets in human liver lymph-draining LN show features that may contribute to the immunologically tolerant liver environment. Healthy liver LN, iliac LN, spleen and liver perfusates were obtained from multi-organ donors, while diseased liver LN were collected from explanted patient livers. Inguinal LN were obtained from kidney transplant recipients. Mononuclear cells were isolated from fresh tissues, and immunophenotypic and functional characteristics of APC subsets were studied using flowcytometry and in ex vivo cultures. Healthy liver-draining LN contained significantly lower relative numbers of CD1c+ conventional dendritic cells (cDC2), plasmacytoid DC (PDC), and CD14+CD163+DC-SIGN+ macrophages (MF) compared to inguinal LN. Compared to spleen, both types of LN contained low relative numbers of CD141hi cDC1. Both cDC subsets in liver LN showed a more activated/mature immunophenotype than those in inguinal LN, iliacal LN, spleen and liver tissue. Despite their more mature status, cDC2 isolated from hepatic LN displayed similar cytokine production capacity (IL-10, IL-12, and IL-6) and allogeneic T cell stimulatory capacity as their counterparts from spleen. Liver LN from patients with inflammatory liver diseases showed a further reduction of cDC1, but had increased relative numbers of PDC and MF. In steady state conditions human liver LN contain relatively low numbers of cDC2, PDC, and macrophages, and relative numbers of cDC1 in liver LN decline during liver inflammation. The paucity of cDC in liver LN may contribute to immune tolerance in the liver environment.
Collapse
Affiliation(s)
- Patrick P C Boor
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, Netherlands
| | - Brenda M Bosma
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, Netherlands
| | - Khe T C Tran
- Department of Surgery, Erasmus MC-University Medical Centre, Rotterdam, Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC-University Medical Centre, Rotterdam, Netherlands
| | - Hanneke Hagenaars
- Department of Surgery, Erasmus MC-University Medical Centre, Rotterdam, Netherlands
| | - Jan N M IJzermans
- Department of Surgery, Erasmus MC-University Medical Centre, Rotterdam, Netherlands
| | - Herold J Metselaar
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, Netherlands
| | - Jaap Kwekkeboom
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, Netherlands
| |
Collapse
|
18
|
Long L, Zhang X, Chen F, Pan Q, Phiphatwatchara P, Zeng Y, Chen H. The promising immune checkpoint LAG-3: from tumor microenvironment to cancer immunotherapy. Genes Cancer 2018; 9:176-189. [PMID: 30603054 PMCID: PMC6305110 DOI: 10.18632/genesandcancer.180] [Citation(s) in RCA: 253] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cancer immunotherapy and tumor microenvironment have been at the forefront of research over the past decades. Targeting immune checkpoints especially programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) has made a breakthrough in treating advanced malignancies. However, the low response rate brings a daunting challenge, changing the focus to dig deeply into the tumor microenvironment for alternative therapeutic targets. Strikingly, the inhibitory immune checkpoint lymphocyte activation gene-3 (LAG-3) holds considerable potential. LAG-3 suppresses T cells activation and cytokines secretion, thereby ensuring immune homeostasis. It exerts differential inhibitory impacts on various types of lymphocytes and shows a remarkable synergy with PD-1 to inhibit immune responses. Targeting LAG-3 immunotherapy is moving forward in active clinical trials, and combination immunotherapy of anti-LAG-3 and anti-PD-1 has shown exciting efficacy in fighting PD-1 resistance. Herein, we shed light on the significance of LAG-3 in the tumor microenvironment, highlight its role to regulate different lymphocytes, interplay with other immune checkpoints especially PD-1, and emphasize new advances in LAG-3-targeted immunotherapy.
Collapse
Affiliation(s)
- Long Long
- Department of Pathology, School of Basic Medical Science, Wuhan University, Wuhan, P. R. China.,Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, P. R. China
| | - Xue Zhang
- Department of Pathology, School of Basic Medical Science, Wuhan University, Wuhan, P. R. China
| | - Fuchun Chen
- Department of Thoracosurgery, Traditional Chinese Medical Hospital of Wenling, Wenling, Zhejiang, China
| | - Qi Pan
- Department of Thoracosurgery, Traditional Chinese Medical Hospital of Wenling, Wenling, Zhejiang, China
| | | | - Yuyang Zeng
- Department of Pathology, School of Basic Medical Science, Wuhan University, Wuhan, P. R. China
| | - Honglei Chen
- Department of Pathology, School of Basic Medical Science, Wuhan University, Wuhan, P. R. China
| |
Collapse
|
19
|
Description of CD8 + Regulatory T Lymphocytes and Their Specific Intervention in Graft-versus-Host and Infectious Diseases, Autoimmunity, and Cancer. J Immunol Res 2018; 2018:3758713. [PMID: 30155493 PMCID: PMC6098849 DOI: 10.1155/2018/3758713] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 05/09/2018] [Accepted: 06/06/2018] [Indexed: 12/13/2022] Open
Abstract
Gershon and Kondo described CD8+ Treg lymphocytes as the first ones with regulating activity due to their tolerance ability to foreign antigens and their capacity to inhibit the proliferation of other lymphocytes. Regardless, CD8+ Treg lymphocytes have not been fully described-unlike CD4+ Treg lymphocytes-because of their low numbers in blood and the lack of specific and accurate population markers. Still, these lymphocytes have been studied for the past 30 years, even after finding difficulties during investigations. As a result, studies have identified markers that define their subpopulations. This review is focused on the expression of cell membrane markers as CD25, CD122, CD103, CTLA-4, CD39, CD73, LAG-3, and FasL as well as soluble molecules such as FoxP3, IFN-γ, IL-10, TGF-β, IL-34, and IL-35, in addition to the lack of expression of cell activation markers such as CD28, CD127 CD45RC, and CD49d. This work also underlines the importance of identifying some of these markers in infections with several pathogens, autoimmunity, cancer, and graft-versus-host disease as a strategy in their prevention, monitoring, and cure.
Collapse
|
20
|
Zhou G, Noordam L, Sprengers D, Doukas M, Boor PPC, van Beek AA, Erkens R, Mancham S, Grünhagen D, Menon AG, Lange JF, Burger PJWA, Brandt A, Galjart B, Verhoef C, Kwekkeboom J, Bruno MJ. Blockade of LAG3 enhances responses of tumor-infiltrating T cells in mismatch repair-proficient liver metastases of colorectal cancer. Oncoimmunology 2018; 7:e1448332. [PMID: 29900067 PMCID: PMC5993483 DOI: 10.1080/2162402x.2018.1448332] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/26/2018] [Accepted: 02/28/2018] [Indexed: 02/06/2023] Open
Abstract
Purpose: Liver metastasis develops in >50% of patients with colorectal cancer (CRC), and is a leading cause of CRC-related mortality. We aimed to identify which inhibitory immune checkpoint pathways can be targeted to enhance functionality of intra-tumoral T-cells in mismatch repair-proficient liver metastases of colorectal cancer (LM-CRC). Methodology: Intra-tumoral expression of multiple inhibitory molecules was compared among mismatch repair-proficient LM-CRC, peritoneal metastases of colorectal cancer (PM-CRC) and primary CRC. Expression of inhibitory molecules was also analyzed on leukocytes isolated from paired resected metastatic liver tumors, tumor-free liver tissues, and blood of patients with mismatch repair-proficient LM-CRC. The effects of blocking inhibitory pathways on tumor-infiltrating T-cell responses were studied in ex vivo functional assays. Results: Mismatch repair-proficient LM-CRC showed higher expression of inhibitory receptors on intra-tumoral T-cells and contained higher proportions of CD8+ T-cells, dendritic cells and monocytes than mismatch repair-proficient primary CRC and/or PM-CRC. Inhibitory receptors LAG3, PD-1, TIM3 and CTLA4 were higher expressed on CD8+ T-cells, CD4+ T-helper and/or regulatory T-cells in LM-CRC tumors compared with tumor-free liver and blood. Antibody blockade of LAG3 or PD-L1 increased proliferation and effector cytokine production of intra-tumoral T-cells isolated from LM-CRC in response to both polyclonal and autologous tumor-specific stimulations. Higher LAG3 expression on intra-tumoral CD8+ T-cells associated with longer progression-free survival of LM-CRC patients. Conclusion: Mismatch repair-proficient LM-CRC may be more sensitive to immune checkpoint inhibitors than mismatch repair-proficient primary CRC. Blocking LAG3 enhances tumor-infiltrating T-cell responses of mismatch repair-proficient LM-CRC, and therefore may be a new promising immunotherapeutic target for LM-CRC.
Collapse
Affiliation(s)
- Guoying Zhou
- Departments of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Lisanne Noordam
- Departments of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Dave Sprengers
- Departments of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Michail Doukas
- Department of Pathology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Patrick P C Boor
- Departments of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Adriaan A van Beek
- Departments of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Remco Erkens
- Departments of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Shanta Mancham
- Departments of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Dirk Grünhagen
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Anand G Menon
- Department of Surgery, Havenziekenhuis and IJsselland Hospital, Rotterdam, the Netherlands
| | - Johan F Lange
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Pim J W A Burger
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Alexandra Brandt
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Boris Galjart
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Cornelis Verhoef
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Jaap Kwekkeboom
- Departments of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Marco J Bruno
- Departments of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
21
|
Zhou G, Sprengers D, Boor PPC, Doukas M, Schutz H, Mancham S, Pedroza-Gonzalez A, Polak WG, de Jonge J, Gaspersz M, Dong H, Thielemans K, Pan Q, IJzermans JNM, Bruno MJ, Kwekkeboom J. Antibodies Against Immune Checkpoint Molecules Restore Functions of Tumor-Infiltrating T Cells in Hepatocellular Carcinomas. Gastroenterology 2017. [PMID: 28648905 DOI: 10.1053/j.gastro.2017.06.017] [Citation(s) in RCA: 298] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Ligand binding to inhibitory receptors on immune cells, such as programmed cell death 1 (PD-1) and cytotoxic T-lymphocyte associated protein 4 (CTLA4), down-regulates the T-cell-mediated immune response (called immune checkpoints). Antibodies that block these receptors increase antitumor immunity in patients with melanoma, non-small-cell lung cancer, and renal cell cancer. Tumor-infiltrating CD4+ and CD8+ T cells in patients with hepatocellular carcinoma (HCC) have been found to be functionally compromised. We analyzed HCC samples from patients to determine if these inhibitory pathways prevent T-cell responses in HCCs and to find ways to restore their antitumor functions. METHODS We collected HCC samples from 59 patients who underwent surgical resection from November 2013 through May 2017, along with tumor-free liver tissues (control tissues) and peripheral blood samples. We isolated tumor-infiltrating lymphocytes (TIL) and intra-hepatic lymphocytes. We used flow cytometry to quantify expression of the inhibitory receptors PD-1, hepatitis A virus cellular receptor 2 (TIM3), lymphocyte activating 3 (LAG3), and CTLA4 on CD8+ and CD4+ T cells from tumor, control tissue, and blood; we studied the effects of antibodies that block these pathways in T-cell activation assays. RESULTS Expression of PD-1, TIM3, LAG3, and CTLA4 was significantly higher on CD8+ and CD4+ T cells isolated from HCC tissue than control tissue or blood. Dendritic cells, monocytes, and B cells in HCC tumors expressed ligands for these receptors. Expression of PD-1, TIM3, and LAG3 was higher on tumor-associated antigen (TAA)-specific CD8+ TIL, compared with other CD8+ TIL. Compared with TIL that did not express these inhibitory receptors, CD8+ and CD4+ TIL that did express these receptors had higher levels of markers of activation, but similar or decreased levels of granzyme B and effector cytokines. Antibodies against CD274 (PD-ligand1 [PD-L1]), TIM3, or LAG3 increased proliferation of CD8+ and CD4+ TIL and cytokine production in response to stimulation with polyclonal antigens or TAA. Importantly, combining antibody against PD-L1 with antibodies against TIM3, LAG3, or CTLA4 further increased TIL functions. CONCLUSIONS The immune checkpoint inhibitory molecules PD-1, TIM3, and LAG3 are up-regulated on TAA-specific T cells isolated from human HCC tissues, compared with T cells from tumor-free liver tissues or blood. Antibodies against PD-L1, TIM3, or LAG3 restore responses of HCC-derived T cells to tumor antigens, and combinations of the antibodies have additive effects. Strategies to block PD-L1, TIM3, and LAG3 might be developed for treatment of primary liver cancer.
Collapse
MESH Headings
- Antibodies, Monoclonal/pharmacology
- Antibodies, Neutralizing/pharmacology
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Antineoplastic Agents/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- CTLA-4 Antigen/antagonists & inhibitors
- CTLA-4 Antigen/immunology
- CTLA-4 Antigen/metabolism
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Proliferation/drug effects
- Cells, Cultured
- Coculture Techniques
- Cytokines/metabolism
- Hepatitis A Virus Cellular Receptor 2/antagonists & inhibitors
- Hepatitis A Virus Cellular Receptor 2/immunology
- Hepatitis A Virus Cellular Receptor 2/metabolism
- Humans
- Immunotherapy/methods
- Liver Neoplasms/drug therapy
- Liver Neoplasms/immunology
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Lymphocyte Activation/drug effects
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- Programmed Cell Death 1 Receptor/metabolism
- Signal Transduction/drug effects
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Escape/drug effects
- Tumor Microenvironment
- Up-Regulation
- Lymphocyte Activation Gene 3 Protein
Collapse
Affiliation(s)
- Guoying Zhou
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, the Netherlands
| | - Dave Sprengers
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, the Netherlands
| | - Patrick P C Boor
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, the Netherlands
| | - Michail Doukas
- Department of Pathology, Erasmus MC-University Medical Centre, Rotterdam, the Netherlands
| | - Hannah Schutz
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, the Netherlands
| | - Shanta Mancham
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, the Netherlands
| | | | - Wojciech G Polak
- Department of Surgery, Erasmus MC-University Medical Centre, Rotterdam, the Netherlands
| | - Jeroen de Jonge
- Department of Surgery, Erasmus MC-University Medical Centre, Rotterdam, the Netherlands
| | - Marcia Gaspersz
- Department of Surgery, Erasmus MC-University Medical Centre, Rotterdam, the Netherlands
| | - Haidong Dong
- Department of Urology and Immunology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Kris Thielemans
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit, Brussels, and eTheRNA immunotherapies NV, Niel, Belgium
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, the Netherlands
| | | | - Marco J Bruno
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, the Netherlands
| | - Jaap Kwekkeboom
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, the Netherlands.
| |
Collapse
|
22
|
Bézie S, Usal C, Guillonneau C. In Vitro and In Vivo Assessment of T, B and Myeloid Cells Suppressive Activity and Humoral Responses from Transplant Recipients. J Vis Exp 2017. [PMID: 28829428 DOI: 10.3791/55510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The main concern in transplantation is to achieve specific tolerance through induction of regulatory cells. The understanding of tolerance mechanisms requires reliable models. Here, we describe models of tolerance to cardiac allograft in rat, induced by blockade of costimulation signals or by upregulation of immunoregulatory molecules through gene transfer. Each of these models allowed in vivo generation of regulatory cells such as regulatory T cells (Tregs), regulatory B cells (Bregs) or regulatory myeloid cells (RegMCs). In this manuscript, we describe two complementary protocols that have been used to identify and define in vitro and in vivo regulatory cell activity to determine their responsibility in tolerance induction and maintenance. First, an in vitro suppressive assay allowed rapid identification of cells with suppressive capacity on effector immune responses in a dose dependent manner, and can be used for further analysis such as cytokine measurement or cytotoxicity. Second, the adoptive transfer of cells from a tolerant treated recipient to a newly irradiated grafted recipient, highlighted the tolerogenic properties of these cells in controlling graft directed immune responses and/or converting new regulatory cells (termed infectious tolerance). These methods are not restricted to cells with known phenotypic markers and can be extended to any cell population. Furthermore, donor directed allospecificity of regulatory cells (an important goal in the field) can be assessed by using third party donor cells or graft either in vitro or in vivo. Finally, to determine the specific tolerogenic capacity of these regulatory cells, we provide protocols to assess the humoral anti-donor antibody responses and the capacity of the recipient to develop humoral responses against new or former known antigens. The models of tolerance described can be used to further characterize regulatory cells, to identify new biomarkers, and immunoregulatory molecules, and are adaptable to other transplantation models or autoimmune diseases in rodent or human.
Collapse
Affiliation(s)
- Séverine Bézie
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes; Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes
| | - Claire Usal
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes; Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes
| | - Carole Guillonneau
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes; Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes;
| |
Collapse
|
23
|
Abstract
Regulatory T cells (Tregs) represent a cell type that promotes immune tolerance to autologous components and maintains immune system homeostasis. The abnormal function of Tregs is relevant to the pathogenesis of systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and other autoimmune diseases. Therefore, therapeutic modulation of Tregs could be a potent means of treating autoimmune diseases. Human Tregs are diverse, however, and not all of them have immunosuppressive effects. Forkhead box P3 (Foxp3), a pivotal transcription factor of Tregs that is crucial in maintaining Treg immunosuppressive function, can be expressed heterogeneously or unstably across Treg subpopulations. Insights into modulating Treg differentiation on the level of DNA transcription or protein modification may improve the success of Treg modifying immunotherapies. In this review, we will summarize three main prospects: the regulatory mechanism of Foxp3, the influence on Foxp3 and Tregs in autoimmune diseases, then finally, how Tregs can be used to treat autoimmune diseases.
Collapse
|
24
|
Wu M, Lou J, Zhang S, Chen X, Huang L, Sun R, Huang P, Pan S, Wang F. Gene expression profiling of CD8 + T cells induced by ovarian cancer cells suggests a possible mechanism for CD8 + Treg cell production. Cell Prolif 2016; 49:669-677. [PMID: 27641758 DOI: 10.1111/cpr.12294] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 07/30/2016] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES The aim of this study was to investigate a possible mechanism of CD8+ regulatory T-cell (Treg) production in an ovarian cancer (OC) microenvironment. MATERIALS AND METHODS Agilent microarray was used to detect changes in gene expression between CD8+ T cells cultured with and without the SKOV3 ovarian adenocarcinoma cell line. QRT-PCR was performed to determine glycolysis gene expression in CD8+ T cells from a transwell culturing system and OC patients. We also detected protein levels of glycolysis-related genes using Western blot analysis. RESULTS Comparing gene expression profiles revealed significant differences in expression levels of 1420 genes, of which 246 were up-regulated and 1174 were down-regulated. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis indicated that biological processes altered in CD8+ Treg are particularly associated with energy metabolism. CD8+ Treg cells induced by co-culture with SKOV3 had lower glycolysis gene expression compared to CD8+ T cells cultured alone. Glycolysis gene expression was also decreased in the CD8+ T cells of OC patients. CONCLUSIONS These findings provide a comprehensive bioinformatics analysis of DEGs in CD8+ T cells cultured with and without SKOV3 and suggests that metabolic processes may be a possible mechanism for CD8+ Treg induction.
Collapse
Affiliation(s)
- Meng Wu
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China
| | - Jianfang Lou
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China
| | - Shuping Zhang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China
| | - Xian Chen
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China
| | - Lei Huang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China
| | - Ruihong Sun
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China
| | - Peijun Huang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China
| | - Shiyang Pan
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China
| | - Fang Wang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China. .,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China.
| |
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
|
Lin K, Chen S, Chen G. Role of Memory T Cells and Perspectives for Intervention in Organ Transplantation. Front Immunol 2015; 6:473. [PMID: 26441978 PMCID: PMC4568416 DOI: 10.3389/fimmu.2015.00473] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 08/31/2015] [Indexed: 12/12/2022] Open
Abstract
Memory T cells are necessary for protective immunity against invading pathogens, especially under conditions of immunosuppression. However, their presence also threatens transplant survival, making transplantation a great challenge. Significant progress has been achieved in recent years in advancing our understanding of the role that memory T cells play in transplantation. This review focuses on the latest advances in our understanding of the involvement of memory T cells in graft rejection and transplant tolerance and discusses potential strategies for targeting memory T cells in order to minimize allograft rejection and optimize clinical outcomes.
Collapse
Affiliation(s)
- Kailin Lin
- Institute of Organ Transplantation, Tongji Hospital, Huazhong University of Science and Technology , Wuhan , China
| | - Song Chen
- Institute of Organ Transplantation, Tongji Hospital, Huazhong University of Science and Technology , Wuhan , China ; Key Laboratory of Organ Transplantation, Ministry of Education , Wuhan , China ; Key Laboratory of Organ Transplantation, Ministry of Public Health , Wuhan , China
| | - Gang Chen
- Institute of Organ Transplantation, Tongji Hospital, Huazhong University of Science and Technology , Wuhan , China ; Key Laboratory of Organ Transplantation, Ministry of Education , Wuhan , China ; Key Laboratory of Organ Transplantation, Ministry of Public Health , Wuhan , China
| |
Collapse
|
27
|
Lin PL, Flynn JL. CD8 T cells and Mycobacterium tuberculosis infection. Semin Immunopathol 2015; 37:239-49. [PMID: 25917388 PMCID: PMC4439333 DOI: 10.1007/s00281-015-0490-8] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 04/06/2015] [Indexed: 12/25/2022]
Abstract
Tuberculosis is primarily a respiratory disease that is caused by Mycobacterium tuberculosis. M. tuberculosis can persist and replicate in macrophages in vivo, usually in organized cellular structures called granulomas. There is substantial evidence for the importance of CD4 T cells in control of tuberculosis, but the evidence for a requirement for CD8 T cells in this infection has not been proven in humans. However, animal model data support a non-redundant role for CD8 T cells in control of M. tuberculosis infection. In humans, infection with this pathogen leads to generation of specific CD8 T cell responses. These responses include classical (MHC Class I restricted) and non-classical CD8 T cells. Here, we discuss the potential roles of CD8 T cells in defense against tuberculosis, and our current understanding of the wide range of CD8 T cell types seen in M. tuberculosis infection.
Collapse
Affiliation(s)
- Philana Ling Lin
- Department of Pediatrics, Division of Infectious Disease, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, Pittsburgh, PA, 15224, USA
| | | |
Collapse
|
28
|
Bobosha K, Wilson L, van Meijgaarden KE, Bekele Y, Zewdie M, van der Ploeg- van Schip JJ, Abebe M, Hussein J, Khadge S, Neupane KD, Hagge DA, Jordanova ES, Aseffa A, Ottenhoff THM, Geluk A. T-cell regulation in lepromatous leprosy. PLoS Negl Trop Dis 2014; 8:e2773. [PMID: 24722473 PMCID: PMC3983090 DOI: 10.1371/journal.pntd.0002773] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 02/19/2014] [Indexed: 12/12/2022] Open
Abstract
Regulatory T (Treg) cells are known for their role in maintaining self-tolerance and balancing immune reactions in autoimmune diseases and chronic infections. However, regulatory mechanisms can also lead to prolonged survival of pathogens in chronic infections like leprosy and tuberculosis (TB). Despite high humoral responses against Mycobacterium leprae (M. leprae), lepromatous leprosy (LL) patients have the characteristic inability to generate T helper 1 (Th1) responses against the bacterium. In this study, we investigated the unresponsiveness to M. leprae in peripheral blood mononuclear cells (PBMC) of LL patients by analysis of IFN-γ responses to M. leprae before and after depletion of CD25+ cells, by cell subsets analysis of PBMC and by immunohistochemistry of patients' skin lesions. Depletion of CD25+ cells from total PBMC identified two groups of LL patients: 7/18 (38.8%) gained in vitro responsiveness towards M. leprae after depletion of CD25+ cells, which was reversed to M. leprae-specific T-cell unresponsiveness by addition of autologous CD25+ cells. In contrast, 11/18 (61.1%) remained anergic in the absence of CD25+ T-cells. For both groups mitogen-induced IFN-γ was, however, not affected by depletion of CD25+ cells. In M. leprae responding healthy controls, treated lepromatous leprosy (LL) and borderline tuberculoid leprosy (BT) patients, depletion of CD25+ cells only slightly increased the IFN-γ response. Furthermore, cell subset analysis showed significantly higher (p = 0.02) numbers of FoxP3+ CD8+CD25+ T-cells in LL compared to BT patients, whereas confocal microscopy of skin biopsies revealed increased numbers of CD68+CD163+ as well as FoxP3+ cells in lesions of LL compared to tuberculoid and borderline tuberculoid leprosy (TT/BT) lesions. Thus, these data show that CD25+ Treg cells play a role in M. leprae-Th1 unresponsiveness in LL. Leprosy is a curable infectious disease caused by Mycobacterium leprae (M. leprae) that affects the skin and peripheral nerves. It is manifested in different forms ranging from self-healing, tuberculoid leprosy (TT) with low bacillary load and high cellular immunity against M. leprae, to lepromatous leprosy (LL) with high bacillary load and high antibody titers to M. leprae antigens. However, LL patients have poor cell mediated response against M. leprae leading to delayed clearance of the bacilli. A possible explanation for this bacterial persistence could lie in the presence of more regulatory cells at infection sites and in peripheral blood. This study shows the recovery of the cell mediated response by depletion of CD25+ cells in a subset of LL patients, while another patient subset was not affected similarly. Moreover, an increased frequency of FoxP3+ T cells together with anti-inflammatory macrophages was observed in LL patients' skin biopsies. Thus, these data show that CD25+ Treg cells play a role in M. leprae-unresponsiveness in leprosy patients.
Collapse
Affiliation(s)
- Kidist Bobosha
- The Dept. of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
- Armauer Hansen Research Institute and ALERT hospital, Addis Ababa, Ethiopia
- * E-mail:
| | - Louis Wilson
- The Dept. of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Yonas Bekele
- Armauer Hansen Research Institute and ALERT hospital, Addis Ababa, Ethiopia
| | - Martha Zewdie
- Armauer Hansen Research Institute and ALERT hospital, Addis Ababa, Ethiopia
| | | | - Markos Abebe
- Armauer Hansen Research Institute and ALERT hospital, Addis Ababa, Ethiopia
| | - Jemal Hussein
- Armauer Hansen Research Institute and ALERT hospital, Addis Ababa, Ethiopia
| | - Saraswoti Khadge
- Mycobacterial Research Laboratory, Anandaban Hospital, Kathmandu, Nepal
| | - Kapil D. Neupane
- Mycobacterial Research Laboratory, Anandaban Hospital, Kathmandu, Nepal
| | - Deanna A. Hagge
- Mycobacterial Research Laboratory, Anandaban Hospital, Kathmandu, Nepal
| | - Ekaterina S. Jordanova
- The Dept. of Obstetrics and Gynaecology, Free University Amsterdam, Center for Gynaecologic Oncology, Amsterdam, The Netherlands
| | - Abraham Aseffa
- Armauer Hansen Research Institute and ALERT hospital, Addis Ababa, Ethiopia
| | - Tom H. M. Ottenhoff
- The Dept. of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Annemieke Geluk
- The Dept. of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
29
|
Boer MC, van Meijgaarden KE, Joosten SA, Ottenhoff THM. CD8+ regulatory T cells, and not CD4+ T cells, dominate suppressive phenotype and function after in vitro live Mycobacterium bovis-BCG activation of human cells. PLoS One 2014; 9:e94192. [PMID: 24714620 PMCID: PMC3979753 DOI: 10.1371/journal.pone.0094192] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 03/01/2014] [Indexed: 01/24/2023] Open
Abstract
Mycobacterium bovis bacillus Calmette-Guérin (M. bovis BCG), the only currently available vaccine against tuberculosis, has been reported to induce regulatory T cells in humans. The activity of regulatory T cells may not only dampen immunogenicity and protective efficacy of tuberculosis-vaccines, but also hamper diagnosis of infection of tuberculosis, when using immune (e.g. IFNγ-release) assays. Still, in settings of infectious diseases and vaccination, most studies have focused on CD4+ regulatory T cells, and not CD8+ regulatory T-cells. Here, we present a comparative analysis of the suppressive phenotype and function of CD4+ versus CD8+ T cells after in vitro live BCG activation of human cells. Moreover, as BCG is administered as a (partly) live vaccine, we also compared the ability of live versus heatkilled BCG in activating CD4+ and CD8+ regulatory T cell responses. BCG-activated CD8+ T cells consistently expressed higher levels of regulatory T cell markers, and after live BCG activation, density and (co-)expression of markers were significantly higher, compared to CD4+ T cells. Furthermore, selection on CD25-expression after live BCG activation enriched for CD8+ T cells, and selection on co-expression of markers further increased CD8+ enrichment. Ultimately, only T cells activated by live BCG were functionally suppressive and this suppressive activity resided predominantly in the CD8+ T cell compartment. These data highlight the important contribution of live BCG-activated CD8+ Treg cells to immune regulation and emphasize their possible negative impact on immunity and protection against tuberculosis, following BCG vaccination.
Collapse
Affiliation(s)
- Mardi C. Boer
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Simone A. Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
30
|
Lu H. TLR Agonists for Cancer Immunotherapy: Tipping the Balance between the Immune Stimulatory and Inhibitory Effects. Front Immunol 2014; 5:83. [PMID: 24624132 PMCID: PMC3939428 DOI: 10.3389/fimmu.2014.00083] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 02/17/2014] [Indexed: 01/07/2023] Open
Affiliation(s)
- Hailing Lu
- Tumor Vaccine Group, University of Washington , Seattle, WA , USA
| |
Collapse
|
31
|
Boor PPC, Metselaar HJ, Mancham S, van der Laan LJW, Kwekkeboom J. Rapamycin has suppressive and stimulatory effects on human plasmacytoid dendritic cell functions. Clin Exp Immunol 2014; 174:389-401. [PMID: 23968562 DOI: 10.1111/cei.12191] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2013] [Indexed: 01/23/2023] Open
Abstract
Plasmacytoid dendritic cells (PDC) are involved in innate immunity by interferon (IFN)-α production, and in adaptive immunity by stimulating T cells and inducing generation of regulatory T cells (Treg ). In this study we studied the effects of mammalian target of rapamycin (mTOR) inhibition by rapamycin, a commonly used immunosuppressive and anti-cancer drug, on innate and adaptive immune functions of human PDC. A clinically relevant concentration of rapamycin inhibited Toll-like receptor (TLR)-7-induced IFN-α secretion potently (-64%) but TLR-9-induced IFN-α secretion only slightly (-20%), while the same concentration suppressed proinflammatory cytokine production by TLR-7-activated and TLR-9-activated PDC with similar efficacy. Rapamycin inhibited the ability of both TLR-7-activated and TLR-9-activated PDC to stimulate production of IFN-γ and interleukin (IL)-10 by allogeneic T cells. Surprisingly, mTOR-inhibition enhanced the capacity of TLR-7-activated PDC to stimulate naive and memory T helper cell proliferation, which was caused by rapamycin-induced up-regulation of CD80 expression on PDC. Finally, rapamycin treatment of TLR-7-activated PDC enhanced their capacity to induce CD4(+) forkhead box protein 3 (FoxP3)(+) regulatory T cells, but did not affect the generation of suppressive CD8(+) CD38(+) lymphocyte activation gene (LAG)-3(+) Treg . In general, rapamycin inhibits innate and adaptive immune functions of TLR-stimulated human PDC, but enhances the ability of TLR-7-stimulated PDC to stimulate CD4(+) T cell proliferation and induce CD4(+) FoxP3(+) regulatory T cell generation.
Collapse
Affiliation(s)
- P P C Boor
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Centre, Rotterdam, the Netherlands
| | | | | | | | | |
Collapse
|
32
|
High doses of CpG oligodeoxynucleotides stimulate a tolerogenic TLR9-TRIF pathway. Nat Commun 2013; 4:1852. [PMID: 23673637 DOI: 10.1038/ncomms2874] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 04/12/2013] [Indexed: 02/07/2023] Open
Abstract
CpG-rich oligodeoxynucleotides activate the immune system, leading to innate and acquired immune responses. The immune-stimulatory effects of CpG-rich oligodeoxynucleotides are being exploited as a therapeutic approach. Here we show that at high doses, CpG-rich oligodeoxynucleotides promote an opposite, tolerogenic response in mouse plasmacytoid dendritic cells in vivo and in a human in vitro model. Unveiling a previously undescribed role for TRIF and TRAF6 proteins in Toll-like receptor 9 (TLR9) signalling, we demonstrate that physical association of TLR9, TRIF and TRAF6 leads to activation of noncanonical NF-κB signalling and the induction of IRF3- and TGF-β-dependent immune-suppressive tryptophan catabolism. In vivo, the TLR9-TRIF circuit--but not MyD88 signalling--was required for CpG protection against allergic inflammation. Our findings may be relevant to an increased understanding of the complexity of Toll-like receptor signalling and optimal exploitation of CpG-rich oligodeoxynucleotides as immune modulators.
Collapse
|
33
|
Zheng J, Liu Y, Liu Y, Liu M, Xiang Z, Lam KT, Lewis DB, Lau YL, Tu W. Human CD8+ regulatory T cells inhibit GVHD and preserve general immunity in humanized mice. Sci Transl Med 2013; 5:168ra9. [PMID: 23325802 DOI: 10.1126/scitranslmed.3004943] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Graft-versus-host disease (GVHD) is a lethal complication of allogeneic bone marrow transplantation (BMT). Immunosuppressive agents are currently used to control GVHD but may cause general immune suppression and limit the effectiveness of BMT. Adoptive transfer of regulatory T cells (T(regs)) can prevent GVHD in rodents, suggesting a therapeutic potential of T(regs) for GVHD in humans. However, the clinical application of T(reg)-based therapy is hampered by the low frequency of human T(regs) and the lack of a reliable model to test their therapeutic effects in vivo. Recently, we successfully generated human alloantigen-specific CD8(hi) T(regs) in a large scale from antigenically naïve precursors ex vivo using allogeneic CD40-activated B cells as stimulators. We report a human allogeneic GVHD model established in humanized mice to mimic GVHD after BMT in humans. We demonstrate that ex vivo-induced CD8(hi) T(regs) controlled GVHD in an allospecific manner by reducing alloreactive T cell proliferation as well as decreasing inflammatory cytokine and chemokine secretion within target organs through a CTLA-4-dependent mechanism in humanized mice. These CD8(hi) T(regs) induced long-term tolerance effectively without compromising general immunity and graft-versus-tumor activity. Our results support testing of human CD8(hi) T(regs) in GVHD in clinical trials.
Collapse
Affiliation(s)
- Jian Zheng
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 000000, China
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Maazi H, Lam J, Lombardi V, Akbari O. Role of plasmacytoid dendritic cell subsets in allergic asthma. Allergy 2013; 68:695-701. [PMID: 23662841 DOI: 10.1111/all.12166] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2013] [Indexed: 12/19/2022]
Abstract
Plasmacytoid dendritic cells (pDCs) are major type-I interferon-producing cells that play important roles in antiviral immunity and tolerance induction. These cells share a common DC progenitor with conventional DCs, and Fms-like tyrosine kinase-3 ligand is essential for their development. Several subsets of pDCs have been identified to date including CCR9(+) , CD9(+) , and CD2(+) pDCs. Recently, three subsets of pDCs were described, namely CD8α(-) β(-) , CD8α(+) β(-) , and CD8α(+) β(+) subsets. Interestingly, CD8α(+) β(-) and CD8α(+) β(+) but not CD8α(-) β(-) pDCs were shown to have tolerogenic effects in experimentally induced allergic asthma. These tolerogenic effects were shown to be mediated by the generation of FOXP3(+) regulatory T cells through retinoic acid and the induction of retinaldehyde dehydrogenase enzymes. These newly described subsets of pDCs show high potentials for novel therapeutic approaches for the treatment of allergic diseases. In this review, we will address the new progress in our understanding of pDC biology with respect to allergic disease, in particular allergic asthma.
Collapse
Affiliation(s)
- H. Maazi
- Department of Molecular Microbiology and Immunology; Keck School of Medicine; University of Southern California; Los Angeles; CA; USA
| | - J. Lam
- Department of Molecular Microbiology and Immunology; Keck School of Medicine; University of Southern California; Los Angeles; CA; USA
| | - V. Lombardi
- Department of Molecular Microbiology and Immunology; Keck School of Medicine; University of Southern California; Los Angeles; CA; USA
| | - O. Akbari
- Department of Molecular Microbiology and Immunology; Keck School of Medicine; University of Southern California; Los Angeles; CA; USA
| |
Collapse
|
35
|
Eloranta ML, Alm GV, Rönnblom L. Disease mechanisms in rheumatology--tools and pathways: plasmacytoid dendritic cells and their role in autoimmune rheumatic diseases. ACTA ACUST UNITED AC 2013; 65:853-63. [PMID: 23280551 DOI: 10.1002/art.37821] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 12/04/2012] [Indexed: 12/19/2022]
Affiliation(s)
- Maija-Leena Eloranta
- Department of Medical Sciences, Rheumatology, Uppsala University, Uppsala, Sweden
| | | | | |
Collapse
|
36
|
Chen Z, Han Y, Gu Y, Liu Y, Jiang Z, Zhang M, Cao X. CD11c(high)CD8+ regulatory T cell feedback inhibits CD4 T cell immune response via Fas ligand-Fas pathway. THE JOURNAL OF IMMUNOLOGY 2013; 190:6145-54. [PMID: 23677464 DOI: 10.4049/jimmunol.1300060] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Regulatory T cells can restrict the uncontrolled immune response and inflammation, avoiding pathologic immune injury to the host and thus playing important roles in the maintenance of immune homeostasis. Until recently, many subsets of CD4 and CD8 regulatory T cells have been reported. In this study, we identified CD11c(high)CD8(+) T cells as a new subset of CD8(+) regulatory T cells. During Listeria monocytogenes and Staphylococcus aureus infection, two subsets of CD8 T cells were classified according to the expression level of CD11c, including CD11c(low)CD8(+) and CD11c(high)CD8(+) T cells. CD11c(low)CD8(+) T cells, existing during the whole period of infection, act as conventional activated T cells to kill target cells in a perforin-dependent manner. Interestingly, CD11c(high)CD8(+) T cells appeared only at a late stage of infection, expressed relatively high CD122 and low CD69, did not secrete IFN-γ, IL-10, TGF-β, and exhibited much more potent cytotoxicity against target cells via Fas ligand-Fas pathway in an Ag-independent manner. Ligation of CD11c was important in the cytotoxicity of CD11c(high)CD8(+) T cells. Furthermore, CD11c(high)CD8(+) T cells could directly kill the activated CD4 T cells both in vitro and in vivo, whereas CD11c(low)CD8(+) T cells could not. Thus, we identified an infection-induced new subset of CD11c(high)CD8(+) regulatory T cells, which might contribute to protect host from pathological immune injure. Our results indicate that CD11c(+)CD8(+) T cells are constitute a heterogeneous population that can be divided further into regulatory CD11c(high)CD8(+) T cell subset and effector CD11c(low)CD8(+) T cell subset, thus adding insight to the role of CD8 T cells in immune response and regulation.
Collapse
Affiliation(s)
- Zhubo Chen
- Institute of Immunology, Tsinghua University School of Medicine, Beijing 100084, China
| | | | | | | | | | | | | |
Collapse
|
37
|
Lerret NM, Houlihan JL, Kheradmand T, Pothoven KL, Zhang ZJ, Luo X. Donor-specific CD8+ Foxp3+ T cells protect skin allografts and facilitate induction of conventional CD4+ Foxp3+ regulatory T cells. Am J Transplant 2012; 12:2335-47. [PMID: 22681667 PMCID: PMC3429694 DOI: 10.1111/j.1600-6143.2012.04120.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
CD4(+) regulatory T cells play a critical role in tolerance induction in transplantation. CD8(+) suppressor T cells have also been shown to control alloimmune responses in preclinical and clinical models. However, the exact nature of the CD8(+) suppressor T cells, their induction and mechanism of function in allogeneic transplantation remain elusive. In this study, we show that functionally suppressive, alloantigen-specific CD8(+) Foxp3(+) T cells can be induced and significantly expanded by stimulating naïve CD8(+) T cells with donor dendritic cells in the presence of IL-2, TGF-β1 and retinoic acid. These CD8(+) Foxp3(+) T cells express enhanced levels of CTLA-4, CCR4 and CD103, inhibit the up-regulation of costimulatory molecules on dendritic cells, and suppress CD4 and CD8 T cell proliferation and cytokine production in a donor-specific and contact-dependent manner. Importantly, upon adoptive transfer, the induced CD8(+) Foxp3(+) T cells protect full MHC-mismatched skin allografts. In vivo, the CD8(+) Foxp3(+) T cells preferentially traffic to the graft draining lymph node where they induce conventional CD4(+) Foxp3(+) T cells and concurrently suppress effector T cell expansion. We conclude that donor-specific CD8(+) Foxp3(+) suppressor T cells can be induced and exploited as an effective form of cell therapy for graft protection in transplantation.
Collapse
Affiliation(s)
- Nadine M. Lerret
- Division of Nephrology and Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago IL, USA,Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| | - Josetta L. Houlihan
- Division of Nephrology and Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago IL, USA,Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| | - Taba Kheradmand
- Division of Nephrology and Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| | - Kathryn L. Pothoven
- Division of Nephrology and Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago IL, USA,Department of Allergy and Immunology, Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| | - Zheng J. Zhang
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| | - Xunrong Luo
- Division of Nephrology and Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago IL, USA,Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago IL, USA,Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| |
Collapse
|
38
|
Hubo M, Jonuleit H. Plasmacytoid dendritic cells are inefficient in activation of human regulatory T cells. PLoS One 2012; 7:e44056. [PMID: 22952871 PMCID: PMC3430613 DOI: 10.1371/journal.pone.0044056] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 07/30/2012] [Indexed: 12/20/2022] Open
Abstract
Background Dendritic cells (DC) play a key role in initiation and regulation of immune responses. Plasmacytoid DC (pDC), a small subset of DC, characterized as type-I interferon producing cells, are critically involved in anti-viral immune responses, but also mediate tolerance by induction of regulatory T cells (Treg). In this study, we compared the capacity of human pDC and conventional DC (cDC) to modulate T cell activity in presence of Foxp3+ Treg. Principal Findings In coculture of T effector cells (Teff) and Treg, activated cDC overcome Treg anergy, abrogate their suppressive function and induce Teff proliferation. In contrast, pDC do not break Treg anergy but induce Teff proliferation even in coculture with Treg. Lack of Treg-mediated suppression is independent of proinflammatory cytokines like IFN-α, IL-1, IL-6 and TNF-α. Phenotyping of pDC-stimulated Treg reveals a reduced expression of Treg activation markers GARP and CTLA-4. Additional stimulation by anti-CD3 antibodies enhances surface expression of GARP and CTLA-4 on Treg and consequently reconstitutes their suppressive function, while increased costimulation with anti-CD28 antibodies is ineffective. Conclusions/Significance Our data show that activated pDC induce Teff proliferation, but are insufficient for functional Treg activation and, therefore, allow expansion of Teff also in presence of Treg.
Collapse
Affiliation(s)
- Mario Hubo
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany.
| | | |
Collapse
|
39
|
Luo L, Sun Z, Cheng H, Luo G. Memory T-cell-specific therapeutics attenuate allograft rejection via mediation of alloreactivity in memory cells. Immunol Lett 2012; 148:53-8. [PMID: 22902239 DOI: 10.1016/j.imlet.2012.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 07/30/2012] [Accepted: 08/01/2012] [Indexed: 02/01/2023]
Abstract
Many means in inbred rodent models promoted long-term graft survival or donor-specific tolerance, but less so in nonhuman primates, outbred rodents or human patients. A diverse repertoire of memory T cells, derived from heterologous immunity or prior to exposure to alloantigen, has been believed to be an important part of this barrier. Memory T cells have a unique capacity to generate effector functions quickly upon re-exposure to antigen, and this capacity is achieved by reduced activation thresholds, and expressed high level trafficking and adhesion molecules, which is likely responsible for their exhibiting differential susceptibility to immune therapeutics compared with naïve T cells. This review outlines recent progress on characteristics of memory T cells and focuses on these potential therapies targeting memory T cells which are likely to ameliorate allograft rejection by inducing transplant tolerance.
Collapse
Affiliation(s)
- Lei Luo
- Department of Research and Education, Guizhou Province People's Hospital, Guiyang, China
| | | | | | | |
Collapse
|
40
|
Lanzinger M, Jürgens B, Hainz U, Dillinger B, Raberger J, Fuchs D, Heitger A. Ambivalent effects of dendritic cells displaying prostaglandin E2-induced indoleamine 2,3-dioxygenase. Eur J Immunol 2012; 42:1117-28. [PMID: 22539287 DOI: 10.1002/eji.201141765] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Prostaglandin E2 (PGE(2)), an abundantly produced lipid messenger in mammalian organisms, has been attributed to possess potent albeit ambivalent immunological functions. Recently, PGE(2) has been reported to stimulate the commonly believed immunosuppressive indoleamine 2,3-dioxygenase (IDO) pathway in human dendritic cells (DCs), but without promoting DC immunosuppressive activity. Here, we report that PGE(2) used as a DC maturation agent apparently has more diverse functions. PGE(2)-matured DCs acquired powerful IDO activity, which was sustained even after removing PGE(2). These IDO-competent DCs were able to stimulate allogeneic T-cell proliferation, but achieved inhibitory activity as their content in DC/T-cell co-cultures increased. The DC inhibitory activity was reversed upon blockade of IDO activity, confirming that the suppressive effect was in fact mediated by IDO and occurred in a dose-dependent fashion. IDO-mediated T-cell suppression was restored upon re-stimulation of T cells in the absence of IDO activity, confirming its reversibility. T cells stimulated by PGE(2)-matured IDO-competent DCs were sensitized to produce multiple cytokines, comprising Th1, Th2, and Th17 phenotypes. Collectively, these data suggest that T cells stimulated by PGE(2)-matured DCs are not terminally differentiated and their ultimate type of response may be formed by microenvironmental conditions.
Collapse
|
41
|
Krummey SM, Ford ML. Heterogeneity within T Cell Memory: Implications for Transplant Tolerance. Front Immunol 2012; 3:36. [PMID: 22566919 PMCID: PMC3342058 DOI: 10.3389/fimmu.2012.00036] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 02/15/2012] [Indexed: 12/16/2022] Open
Abstract
Adaptive immunity in both mouse and man results in the generation of immunological memory. Memory T cells are both friend and foe to transplant recipients, as they are intimately involved and in many cases absolutely required for the maintenance of protective immunity in the face immunosuppression, yet from the evidence presented herein they clearly constitute a formidable barrier for the successful implementation of tolerance induction strategies in transplantation. This review describes the experimental evidence demonstrating the increased resistance of memory T cells to many distinct tolerance induction strategies, and outlines recent advances in our knowledge of the ways in which alloreactive memory T cells arise in previously untransplanted individuals. Understanding the impact of alloreactive memory T cell specificity, frequency, and quality might allow for better donor selection in order to minimize the donor-reactive memory T cell barrier in an individual transplant recipient, thus allowing stratification of relative risk of alloreactive memory T cell mediated rejection, and conversely increase the likelihood of successful establishment of tolerance. However, further research into the molecular and cellular pathways involved in alloreactive memory T cell-mediated rejection is required in order to design new strategies to overcome the memory T cell barrier, without critically impairing protective immunity.
Collapse
Affiliation(s)
- Scott M Krummey
- Department of Surgery, Emory Transplant Center, Emory University Atlanta, GA, USA
| | | |
Collapse
|
42
|
Ezzelarab M, Thomson AW. Tolerogenic dendritic cells and their role in transplantation. Semin Immunol 2011; 23:252-63. [PMID: 21741270 DOI: 10.1016/j.smim.2011.06.007] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 06/10/2011] [Indexed: 01/09/2023]
Abstract
The pursuit of clinical transplant tolerance has led to enhanced understanding of mechanisms underlying immune regulation, including the characterization of immune regulatory cells, in particular antigen-presenting cells (APC) and regulatory T cells (Treg), that may play key roles in promoting operational tolerance. Dendritic cells (DC) are highly efficient APC that have been studied extensively in rodents and humans, and more recently in non-human primates. Owing to their ability to regulate both innate and adaptive immune responses, DC are considered to play crucial roles in directing the alloimmune response towards transplant tolerance or rejection. Mechanisms via which they can promote central and peripheral tolerance include clonal deletion, the induction of Treg, and inhibition of memory T cell responses. These properties have led to the use of tolerogenic DC as a therapeutic strategy to promote organ transplant tolerance. In rodents, infusion of donor- or recipient-derived tolerogenic DC can extensively prolong donor-specific allograft survival, in association with regulation of the host T cell response. In clinical transplantation, progress has been made in monitoring DC in relation to graft outcome, including studies in operational liver transplant tolerance. Although clinical trials involving immunotherapeutic DC for patients with cancer are ongoing, implementation of human DC therapy in clinical transplantation will require assessment of various critical issues. These include cell isolation and purification techniques, source, route and timing of administration, and combination immunosuppressive therapy. With ongoing non-human primate studies focused on DC therapy, these logistics can be investigated seeking the optimal approaches. The scientific rationale for implementation of tolerogenic DC therapy to promote clinical transplant tolerance is strong. Evaluation of technical and therapeutic logistic issues is an important next step prior to the application of tolerogenic DC in clinical organ transplantation.
Collapse
Affiliation(s)
- Mohamed Ezzelarab
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, 200 Lothrop Street, BST W1540, Pittsburgh, PA 15261, USA
| | | |
Collapse
|