151
|
Delia M, Carluccio P, Mestice A, Frappampina R, Albano F, Specchia G, Musto P. After Treatment Decrease of Bone Marrow Tregs and Outcome in Younger Patients with Newly Diagnosed Acute Myeloid Leukemia. J Immunol Res 2020; 2020:2134647. [PMID: 33204734 PMCID: PMC7657695 DOI: 10.1155/2020/2134647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/16/2020] [Accepted: 09/29/2020] [Indexed: 12/17/2022] Open
Abstract
An emerging body of evidence demonstrates that defects in antileukemic effector cells in patients with acute myeloid leukemia (AML) can contribute to the development and/or persistence of the disease. In particular, immune suppressive regulatory T cells (Tregs) may contribute to this defective antileukemic immune response, being recruited by bone marrow leukemic cells to evade immune surveillance. We evaluated Tregs (CD4+/CD45RA-/CD25high/CD127low), performing multiparametric flow cytometry on freshly collected bone marrow aspirate (BMA), in addition to the usual molecular and cytogenetic work-up in newly diagnosed AML patients to look for any correlation between Tregs and the overall response rate (ORR). We studied 39 AML younger patients (<65 years), all treated with standard induction chemotherapy. ORR (complete remission (CR)+CR with incomplete hematologic recovery (CRi)) was documented in 21 out of 39 patients (54%); two partial responder patients were also recorded. Apart from the expected impact of the molecular-cytogenetic group (p = 0.03) and the NPM mutation (p = 0.05), diagnostic BMA Tregs did not show any correlation with ORR. However, although BMA Tregs did not differ in the study population after treatment, their counts significantly decreased in responder patients (p = 0.039), while no difference was documented in nonresponder ones. This suggested that the removal of Treg cells is able to evoke and enhance anti-AML immune response. However, the role of BMA Tregs in mediating immune system-AML interactions in the diagnostic and posttreatment phase should be confirmed in a greater number of patients.
Collapse
MESH Headings
- Adult
- Aged
- Biomarkers
- Biomarkers, Tumor
- Biopsy, Needle
- Bone Marrow/pathology
- Disease Management
- Female
- Humans
- Immunophenotyping
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/etiology
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/therapy
- Lymphocyte Count
- Male
- Middle Aged
- Mutation
- Prognosis
- ROC Curve
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Young Adult
Collapse
Affiliation(s)
- Mario Delia
- Hematology and Bone Marrow Transplantation Unit-Azienda Ospedaliero-Universitaria Consorziale Policlinico, Department of Emergency and Organ Transplantation-University of Bari “Aldo Moro”, Bari, Italy
| | - Paola Carluccio
- Hematology and Bone Marrow Transplantation Unit-Azienda Ospedaliero-Universitaria Consorziale Policlinico, Department of Emergency and Organ Transplantation-University of Bari “Aldo Moro”, Bari, Italy
| | - Anna Mestice
- Hematology and Bone Marrow Transplantation Unit-Azienda Ospedaliero-Universitaria Consorziale Policlinico, Department of Emergency and Organ Transplantation-University of Bari “Aldo Moro”, Bari, Italy
| | - Roberta Frappampina
- Hematology and Bone Marrow Transplantation Unit-Azienda Ospedaliero-Universitaria Consorziale Policlinico, Department of Emergency and Organ Transplantation-University of Bari “Aldo Moro”, Bari, Italy
| | - Francesco Albano
- Hematology and Bone Marrow Transplantation Unit-Azienda Ospedaliero-Universitaria Consorziale Policlinico, Department of Emergency and Organ Transplantation-University of Bari “Aldo Moro”, Bari, Italy
| | | | - Pellegrino Musto
- Hematology and Bone Marrow Transplantation Unit-Azienda Ospedaliero-Universitaria Consorziale Policlinico, Department of Emergency and Organ Transplantation-University of Bari “Aldo Moro”, Bari, Italy
| |
Collapse
|
152
|
Wang K, Fu W. Transcriptional regulation of Treg homeostasis and functional specification. Cell Mol Life Sci 2020; 77:4269-4287. [PMID: 32350553 PMCID: PMC7606275 DOI: 10.1007/s00018-020-03534-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/19/2020] [Accepted: 04/20/2020] [Indexed: 12/15/2022]
Abstract
CD4+Foxp3+ regulatory T (Treg) cells are key players in keeping excessive inflammation in check. Mounting evidence has shown that Treg cells exert much more diverse functions in both immunological and non-immunological processes. The development, maintenance and functional specification of Treg cells are regulated by multilayered factors, including antigens and TCR signaling, cytokines, epigenetic modifiers and transcription factors (TFs). In the review, we will focus on TFs by summarizing their unique and redundant roles in Treg cells under physiological and pathophysiological conditions. We will also discuss the recent advances of Treg trajectories between lymphoid organs and non-lymphoid tissues. This review will provide an updated view of the newly identified TFs and new functions of known TFs in Treg biology.
Collapse
Affiliation(s)
- Ke Wang
- Pediatric Diabetes Research Center, Department of Pediatrics, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Wenxian Fu
- Pediatric Diabetes Research Center, Department of Pediatrics, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
- Moores Cancer Center, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| |
Collapse
|
153
|
Antigen presentation, autoantibody production, and therapeutic targets in autoimmune liver disease. Cell Mol Immunol 2020; 18:92-111. [PMID: 33110250 PMCID: PMC7852534 DOI: 10.1038/s41423-020-00568-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023] Open
Abstract
The liver is an important immunological organ that controls systemic tolerance. The liver harbors professional and unconventional antigen-presenting cells that are crucial for tolerance induction and maintenance. Orchestrating the immune response in homeostasis depends on a healthy and well-toned immunological liver microenvironment, which is maintained by the crosstalk of liver-resident antigen-presenting cells and intrahepatic and liver-infiltrating leukocytes. In response to pathogens or autoantigens, tolerance is disrupted by unknown mechanisms. Intrahepatic parenchymal and nonparenchymal cells exhibit unique antigen-presenting properties. The presentation of microbial and endogenous lipid-, metabolite- and peptide-derived antigens from the gut via conventional and nonconventional mechanisms can educate intrahepatic immune cells and elicit effector responses or tolerance. Perturbation of this balance results in autoimmune liver diseases, such as autoimmune hepatitis, primary biliary cholangitis, and primary sclerosing cholangitis. Although the exact etiologies of these autoimmune liver diseases are unknown, it is thought that the disruption of tolerance towards self-antigens and microbial metabolites and lipids, as well as alterations in bile acid composition, may result in changes in effector cell activation and polarization and may reduce or impair protective anti-inflammatory regulatory T and B cell responses. Additionally, the canonical and noncanonical transmission of antigens and antigen:MHC complexes via trogocytosis or extracellular vesicles between different (non) immune cells in the liver may play a role in the induction of hepatic inflammation and tolerance. Here, we summarize emerging aspects of antigen presentation, autoantibody production, and the application of novel therapeutic approaches in the characterization and treatment of autoimmune liver diseases.
Collapse
|
154
|
Duggleby RC, Tsang HP, Strange K, McWhinnie A, Lamikanra AA, Roberts DJ, Hernandez D, Madrigal JA, Danby RD. Enumerating regulatory T cells in cryopreserved umbilical cord blood samples using FOXP3 methylation specific quantitative PCR. PLoS One 2020; 15:e0240190. [PMID: 33095809 PMCID: PMC7584164 DOI: 10.1371/journal.pone.0240190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 09/22/2020] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Allogeneic haematopoietic cell transplantation (HCT) is a curative therapy for severe haematological disorders. However, it carries significant risk of morbidity and mortality. To improve patient outcomes, better graft selection strategies are needed, incorporating HLA matching with clinically important graft characteristics. Studies have shown that the cellular content of HCT grafts, specifically higher ratios of T regulatory (Tregs)/T cells, are important factors influencing outcomes when using adult peripheral blood mobilised grafts. So far, no equivalent study exists in umbilical cord blood (CB) transplantation due to the limitations of cryopreserved CB samples. STUDY DESIGN AND METHODS To establish the most robust and efficient way to measure the Treg content of previously cryopreserved CB units, we compared the enumeration of Treg and CD3+ cells using flow cytometry and an epigenetic, DNA-based methodology. The two methods were assessed for their agreement, consistency and susceptibility to error when enumerating Treg and CD3+ cell numbers in both fresh and cryopreserved CB samples. RESULTS Epigenetic enumeration gave consistent and comparable results in both fresh and frozen CB samples. By contrast, assessment of Tregs and CD3+ cells by flow cytometry was only possible in fresh samples due to significant cell death following cryopreservation and thawing. CONCLUSION Epigenetic assessment offers significant advantages over flow cytometry for analysing cryopreserved CB; similar cell numbers were observed both in fresh and frozen samples. Furthermore, multiple epigenetic assessments can be performed from DNA extracted from small cryopreserved CB segments; often the only CB sample available for clinical studies.
Collapse
Affiliation(s)
- Richard C. Duggleby
- Anthony Nolan Research Institute, London, United Kingdom
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Hoi Pat Tsang
- National Health Service Blood and Transplant, Oxford, United Kingdom
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Kathryn Strange
- Anthony Nolan Research Institute, London, United Kingdom
- UCL Cancer Institute, University College London, London, United Kingdom
| | | | - Abigail A. Lamikanra
- National Health Service Blood and Transplant, Oxford, United Kingdom
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - David J. Roberts
- National Health Service Blood and Transplant, Oxford, United Kingdom
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Diana Hernandez
- Anthony Nolan Research Institute, London, United Kingdom
- UCL Cancer Institute, University College London, London, United Kingdom
| | - J. Alejandro Madrigal
- Anthony Nolan Research Institute, London, United Kingdom
- UCL Cancer Institute, Royal Free NHS Trust, London, United Kingdom
| | - Robert D. Danby
- Anthony Nolan Research Institute, London, United Kingdom
- UCL Cancer Institute, University College London, London, United Kingdom
- Department of Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
155
|
Payen D, Cravat M, Maadadi H, Didelot C, Prosic L, Dupuis C, Losser MR, De Carvalho Bittencourt M. A Longitudinal Study of Immune Cells in Severe COVID-19 Patients. Front Immunol 2020; 11:580250. [PMID: 33178207 PMCID: PMC7597438 DOI: 10.3389/fimmu.2020.580250] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 10/02/2020] [Indexed: 01/09/2023] Open
Abstract
Little is known about the time-dependent immune responses in severe COVID-19. Data of 15 consecutive patients were sequentially recorded from intensive care unit admission. Lymphocyte subsets and total monocyte and subsets counts were monitored as well as the expression of HLA-DR. For 5 patients, SARS-CoV-2-specific T-cell polyfunctionality was assessed against Spike and Nucleoprotein SARS-CoV-2 peptides. Non-specific inflammation markers were increased in all patients. Median monocyte HLA-DR expression was below the 8,000 AB/C threshold defining acquired immunodepression. A "V" trend curve for lymphopenia, monocyte numbers, and HLA-DR expression was observed with a nadir between days 11 and 14 after symptoms' onset. Intermediate CD14++CD16+ monocytes increased early with a reduction in classic CD14++CD16- monocytes. Polyfunctional SARS-Cov-2-specific CD4 T-cells were present and functional, whereas virus-specific CD8 T-cells were less frequent and not efficient. We report a temporal variation of both innate and adaptive immunity in severe COVID-19 patients, helpful in guiding therapeutic decisions (e.g. anti-inflammatory vs. immunostimulatory ones). We describe a defect in virus-specific CD8 T-cells, a potential biomarker of clinical severity. These combined data also provide helpful knowledge for vaccine design. CLINICAL TRIAL REGISTRATION https://clinicaltrials.gov/, identifier NCT04386395.
Collapse
Affiliation(s)
- Didier Payen
- Université Paris 7 Denis Diderot, UMR 1160 INSERM, Paris, France
| | - Maxime Cravat
- Université de Lorraine, CHRU-Nancy, Laboratoire d’Immunologie, Nancy, France
| | - Hadil Maadadi
- Université de Lorraine, CHRU-Nancy, Département d’Anesthésie Réanimation Brabois Adulte, Nancy, France
| | - Carole Didelot
- CHRU-Nancy, Plateforme de Cytométrie en Flux Diagnostique, Nancy, France
| | - Lydia Prosic
- CHRU-Nancy, Plateforme de Cytométrie en Flux Diagnostique, Nancy, France
| | - Claire Dupuis
- Service de Médecine Intensive et Réanimation, CHU de Clermont-Ferrand, Clermont-Ferrand, France
| | - Marie-Reine Losser
- Université de Lorraine, CHRU-Nancy, Département d’Anesthésie Réanimation Brabois Adulte, Nancy, France
- Université de Lorraine, INSERM UMR 1116, Nancy, France
| | - Marcelo De Carvalho Bittencourt
- Université de Lorraine, CHRU-Nancy, Laboratoire d’Immunologie, Nancy, France
- CHRU-Nancy, Plateforme de Cytométrie en Flux Diagnostique, Nancy, France
- Université de Lorraine, CNRS UMR 7365, IMoPA, Nancy, France
| |
Collapse
|
156
|
Terry LV, Oo YH. The Next Frontier of Regulatory T Cells: Promising Immunotherapy for Autoimmune Diseases and Organ Transplantations. Front Immunol 2020; 11:565518. [PMID: 33072105 PMCID: PMC7538686 DOI: 10.3389/fimmu.2020.565518] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/17/2020] [Indexed: 12/18/2022] Open
Abstract
Regulatory T cells (Tregs) are crucial in maintaining tolerance. Hence, Treg immunotherapy is an attractive therapeutic option in autoimmune diseases and organ transplantations. Currently, autoimmune diseases do not have a curative treatment and transplant recipients require life-long immunosuppression to prevent graft rejection. There has been significant progress in understanding polyclonal and antigen-specific Treg biology over the last decade. Clinical trials with good manufacturing practice (GMP) Treg cells have demonstrated safety and early efficacy of Treg therapy. GMP Treg cells can also be tracked following infusion. In order to improve efficacy of Tregs immunotherapy, it is necessary that Tregs migrate, survive and function at the specific target tissue. Application of antigen specific Tregs and maintaining cells' suppressive function and survival with low dose interleukin-2 (IL-2) will enhance the efficacy and longevity of infused GMP-grade Tregs. Notably, stability of Tregs in the local tissue can be manipulated by understanding the microenvironment. With the recent advances in GMP-grade Tregs isolation and antigen-specific chimeric antigen receptor (CAR)-Tregs development will allow functionally superior cells to migrate to the target organ. Thus, Tregs immunotherapy may be a promising option for patients with autoimmune diseases and organ transplantations in near future.
Collapse
Affiliation(s)
- Lauren V Terry
- Centre for Liver and Gastrointestinal Research, National Institute for Health Research Birmingham Biomedical Research Council, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Ye Htun Oo
- Centre for Liver and Gastrointestinal Research, National Institute for Health Research Birmingham Biomedical Research Council, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,European Reference Network (ERN) Centre-Rare Liver, Queen Elizabeth Hospital, Birmingham, United Kingdom.,Liver Transplant Unit, University Hospital of Birmingham National Health Service Foundation Trust, Birmingham, United Kingdom
| |
Collapse
|
157
|
Brown CY, Sadlon T, Hope CM, Wong YY, Wong S, Liu N, Withers H, Brown K, Bandara V, Gundsambuu B, Pederson S, Breen J, Robertson SA, Forrest A, Beyer M, Barry SC. Molecular Insights Into Regulatory T-Cell Adaptation to Self, Environment, and Host Tissues: Plasticity or Loss of Function in Autoimmune Disease. Front Immunol 2020; 11:1269. [PMID: 33072063 PMCID: PMC7533603 DOI: 10.3389/fimmu.2020.01269] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 05/19/2020] [Indexed: 12/19/2022] Open
Abstract
There has been much interest in the ability of regulatory T cells (Treg) to switch function in vivo, either as a result of genetic risk of disease or in response to environmental and metabolic cues. The relationship between levels of FOXP3 and functional fitness plays a significant part in this plasticity. There is an emerging role for Treg in tissue repair that may be less dependent on FOXP3, and the molecular mechanisms underpinning this are not fully understood. As a result of detailed, high-resolution functional genomics, the gene regulatory networks and key functional mediators of Treg phenotype downstream of FOXP3 have been mapped, enabling a mechanistic insight into Treg function. This transcription factor-driven programming of T-cell function to generate Treg requires the switching on and off of key genes that form part of the Treg gene regulatory network and raises the possibility that this is reversible. It is plausible that subtle shifts in expression levels of specific genes, including transcription factors and non-coding RNAs, change the regulation of the Treg gene network. The subtle skewing of gene expression initiates changes in function, with the potential to promote chronic disease and/or to license appropriate inflammatory responses. In the case of autoimmunity, there is an underlying genetic risk, and the interplay of genetic and environmental cues is complex and impacts gene regulation networks frequently involving promoters and enhancers, the regulatory elements that control gene expression levels and responsiveness. These promoter–enhancer interactions can operate over long distances and are highly cell type specific. In autoimmunity, the genetic risk can result in changes in these enhancer/promoter interactions, and this mainly impacts genes which are expressed in T cells and hence impacts Treg/conventional T-cell (Tconv) function. Genetic risk may cause the subtle alterations to the responsiveness of gene regulatory networks which are controlled by or control FOXP3 and its target genes, and the application of assays of the 3D organization of chromatin, enabling the connection of non-coding regulatory regions to the genes they control, is revealing the direct impact of environmental/metabolic/genetic risk on T-cell function and is providing mechanistic insight into susceptibility to inflammatory and autoimmune conditions.
Collapse
Affiliation(s)
- Cheryl Y Brown
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Timothy Sadlon
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia.,Women's and Children's Health Network, North Adelaide, SA, Australia
| | | | - Ying Y Wong
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Soon Wong
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Ning Liu
- Bioinformatics Hub, University of Adelaide, Adelaide, SA, Australia
| | - Holly Withers
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Katherine Brown
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Veronika Bandara
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Batjargal Gundsambuu
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Stephen Pederson
- Bioinformatics Hub, University of Adelaide, Adelaide, SA, Australia
| | - James Breen
- Bioinformatics Hub, University of Adelaide, Adelaide, SA, Australia
| | - Sarah Anne Robertson
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Alistair Forrest
- QEII Medical Centre and Centre for Medical Research, Harry Perkins Institute of Medical Research, Murdoch, WA, Australia
| | - Marc Beyer
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Simon Charles Barry
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia.,Women's and Children's Health Network, North Adelaide, SA, Australia
| |
Collapse
|
158
|
Sattler FR, Mert M, Sankaranarayanan I, Mack WJ, Galle-Treger L, Gonzalez E, Baronikian L, Lee K, Jahani PS, Hodis HN, Dieli-Conwright C, Akbari O. Feasibility of quantifying change in immune white cells in abdominal adipose tissue in response to an immune modulator in clinical obesity. PLoS One 2020; 15:e0237496. [PMID: 32881912 PMCID: PMC7470412 DOI: 10.1371/journal.pone.0237496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 07/27/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Obesity is often associated with inflammation in adipose tissue (AT) with release of mediators of atherogenesis. We postulated that it would be feasible to collect sufficient abdominal AT to quantify changes in a broad array of adaptive and innate mononuclear white cells in obese non-diabetic adults in response to a dipeptidyl protease inhibitor (DPP4i), known to inhibit activation of immune white cells. METHODS Adults 18-55 years-of-age were screened for abdominal obesity and insulin resistance or impaired glucose tolerance but without known inflammatory conditions. Twenty-one eligible participants consented for study and were randomized 3:1 to receive sitagliptin (DPP4i) at 100mg or matching placebo daily for 28 days. Abdominal AT collected by percutaneous biopsy and peripheral blood mononuclear cell fractions were evaluated before and after treatment; plasma was stored for batch testing. RESULTS Highly sensitive C-reactive protein, a global marker of inflammation, was not elevated in the study population. Innate lymphoid cells (ILC) type 3 (ILC-3) in abdominal AT decreased with active treatment compared with placebo (p = 0.04). Other immune white cells in AT and peripheral blood mononuclear cell (PBMC) fractions did not change with treatment compared to placebo (p>0.05); although ILC-2 declined in PBMCs (p = 0.007) in the sitagliptin treatment group. Two circulating biomarkers of atherogenesis, interferon-inducible protein-10 (IP-10) and sCD40L declined in plasma (p = 0.02 and p = 0.07, respectively) in the active treatment group, providing indirect validation of a net reduction in inflammation. CONCLUSIONS In this pilot study, two cell types of the innate lymphoid system, ILC-3 in AT and ILC-2 PBMCs declined during treatment and as did circulating biomarkers of atherogenesis. Changes in other immune cells were not demonstrable. The study showed that sufficient abdominal AT could be obtained to quantify white cells of both innate and adaptive immunity and to demonstrate changes during therapy with an immune inhibitor. TRIAL REGISTRATION ClinicalTrials.gov identifier (NCT number): NCT02576.
Collapse
Affiliation(s)
- Fred R. Sattler
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
| | - Melissa Mert
- Department of Preventive Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
| | - Ishwarya Sankaranarayanan
- Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
| | - Wendy J. Mack
- Department of Preventive Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
| | - Lauriane Galle-Treger
- Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
| | - Evelyn Gonzalez
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
| | - Lilit Baronikian
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
| | - Kyuwan Lee
- Ostrow School of Dentistry, Division of Physical Therapy and Biokinesiology, University of Southern California, Los Angeles, California, United States of America
- Department of Population Sciences, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Pedram Shafiei Jahani
- Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
| | - Howard N. Hodis
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
| | - Christina Dieli-Conwright
- Ostrow School of Dentistry, Division of Physical Therapy and Biokinesiology, University of Southern California, Los Angeles, California, United States of America
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
| |
Collapse
|
159
|
Sasson SC, Zaunders JJ, Nahar K, Munier CML, Fairfax BP, Olsson-Brown A, Jolly C, Read SA, Ahlenstiel G, Palendira U, Scolyer RA, Carlino MS, Payne MJ, Cheung VTF, Gupta T, Klenerman P, Long GV, Brain O, Menzies AM, Kelleher AD. Mucosal-associated invariant T (MAIT) cells are activated in the gastrointestinal tissue of patients with combination ipilimumab and nivolumab therapy-related colitis in a pathology distinct from ulcerative colitis. Clin Exp Immunol 2020; 202:335-352. [PMID: 32734627 PMCID: PMC7670140 DOI: 10.1111/cei.13502] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/09/2020] [Accepted: 07/07/2020] [Indexed: 12/15/2022] Open
Abstract
The aim of this study was to investigate the pathogenesis of combination ipilimumab and nivolumab-associated colitis (IN-COL) by measuring gut-derived and peripheral blood mononuclear cell (GMNC; PBMC) profiles. We studied GMNC and PBMC from patients with IN-COL, IN-treated with no adverse-events (IN-NAE), ulcerative colitis (UC) and healthy volunteers using flow cytometry. In the gastrointestinal-derived cells we found high levels of activated CD8+ T cells and mucosal-associated invariant T (MAIT) cells in IN-COL, changes that were not evident in IN-NAE or UC. UC, but not IN-C, was associated with a high proportion of regulatory T cells (Treg ). We sought to determine if local tissue responses could be measured in peripheral blood. Peripherally, checkpoint inhibition instigated a rise in activated memory CD4+ and CD8+ T cells, regardless of colitis. Low circulating MAIT cells at baseline was associated with IN-COL patients compared with IN-NAE in one of two cohorts. UC, but not IN-COL, was associated with high levels of circulating plasmablasts. In summary, the alterations in T cell subsets measured in IN-COL-affected tissue, characterized by high levels of activated CD8+ T cells and MAIT cells and a low proportion of Treg , reflected a pathology distinct from UC. These tissue changes differed from the periphery, where T cell activation was a widespread on-treatment effect, and circulating MAIT cell count was low but not reliably predictive of colitis.
Collapse
Affiliation(s)
- S C Sasson
- Translational Gastroenterology Unit and Oxford Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - J J Zaunders
- Centre for Applied Medical Research, St Vincent's Hospital, Sydney, Australia
| | - K Nahar
- Melanoma Institute Australia and The University of Sydney, Sydney, Australia
| | - C M L Munier
- The Kirby Institute, University of New South Wales, Sydney, Australia
| | - B P Fairfax
- Department of Oncology, Churchill Hospital, Oxford, UK.,Department of Oncology, University of Oxford, Oxford, UK.,MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - A Olsson-Brown
- The Clatterbridge Cancer Centre NHS Foundation Trust and Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - C Jolly
- The Clatterbridge Cancer Centre NHS Foundation Trust and Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - S A Read
- Westmead Institute of Medical Research, Sydney, Australia.,Western Sydney University, Sydney, Australia
| | - G Ahlenstiel
- Westmead Institute of Medical Research, Sydney, Australia.,Department of Gastroenterology, Blacktown Hospital, Sydney, Australia
| | - U Palendira
- Discipline of Infectious Diseases and Immunology, The University of Sydney, Sydney, Australia
| | - R A Scolyer
- Melanoma Institute Australia and The University of Sydney, Sydney, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, Australia
| | - M S Carlino
- Melanoma Institute Australia and The University of Sydney, Sydney, Australia.,Department of Medical Oncology, Westmead and Blacktown Hospitals, Sydney, Australia
| | - M J Payne
- Department of Oncology, Churchill Hospital, Oxford, UK
| | - V T F Cheung
- Translational Gastroenterology Unit and Oxford Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - T Gupta
- Translational Gastroenterology Unit and Oxford Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, UK.,MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - P Klenerman
- Translational Gastroenterology Unit and Oxford Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, UK.,Peter Medawar Building of Pathogen Research, University of Oxford, Oxford, UK
| | - G V Long
- Melanoma Institute Australia and The University of Sydney, Sydney, Australia.,Department of Medical Oncology, Royal North Shore Hospital and Mater Hospitals, Sydney, Australia
| | - O Brain
- Translational Gastroenterology Unit and Oxford Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, UK.,Department of Gastroenterology, John Radcliffe Hospital, Oxford, UK
| | - A M Menzies
- Melanoma Institute Australia and The University of Sydney, Sydney, Australia.,Department of Medical Oncology, Royal North Shore Hospital and Mater Hospitals, Sydney, Australia
| | - A D Kelleher
- Centre for Applied Medical Research, St Vincent's Hospital, Sydney, Australia.,The Kirby Institute, University of New South Wales, Sydney, Australia
| |
Collapse
|
160
|
Lopes SM, Roncon S, Pinho AC, Bordalo F, Antunes L, Campilho F, Campos A, Costa-Pereira A. Should we use extracorporeal photopheresis more often? Evidence from graft-versus-host disease patients monitored with Treg as a biomarker. Future Sci OA 2020; 6:FSO623. [PMID: 32983569 PMCID: PMC7491011 DOI: 10.2144/fsoa-2020-0107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 07/23/2020] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Chronic graft-versus-host disease (cGvHD) is a major complication after allogeneic hematopoietic cell transplantation. Extracorporeal photopheresis (ECP) is an immunotherapy treatment for cGvHD, although suitable response biomarkers are lacking. MATERIALS & METHODS We analyzed data from six cGvHD patients undergoing ECP at a reference center from 826 to 2866 days. Circulating Tregs were enumerated, patient's clinical evolution, immunosuppression dose and adverse events (AEs) registered. RESULTS We observed an increase in Tregs, a decrease in immunosuppression dosage and symptoms improvement. Mild AEs occurred at a very low rate. CONCLUSION In these patients, the improvement of cGvHD, with low AEs, confirms a place for ECP as treatment. Improvements were accompanied by an increase in circulating Tregs, suggesting their role as a biomarker.
Collapse
Affiliation(s)
- Sérgio Machado Lopes
- Department of Cellular Therapy, Instituto Português de Oncologia do Porto, 4200-072, Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Susana Roncon
- Department of Cellular Therapy, Instituto Português de Oncologia do Porto, 4200-072, Porto, Portugal
| | - Ana Catarina Pinho
- Department of Cellular Therapy, Instituto Português de Oncologia do Porto, 4200-072, Porto, Portugal
| | - Filipa Bordalo
- Department of Cellular Therapy, Instituto Português de Oncologia do Porto, 4200-072, Porto, Portugal
| | - Luís Antunes
- Department of Bone Marrow Transplant, Instituto Português de Oncologia do Porto, 4200-072, Porto, Portugal
| | - Fernando Campilho
- Department of Epidemiology, Instituto Português de Oncologia do Porto, 4200-072, Porto, Portugal
| | - António Campos
- Department of Epidemiology, Instituto Português de Oncologia do Porto, 4200-072, Porto, Portugal
| | - Altamiro Costa-Pereira
- Center for Health Technology & Services Research (CINTESIS), Department of Community Medicine, Information & Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
| |
Collapse
|
161
|
Tang Y, Zhu L, Xu Q, Zhang X, Li B, Lee LJ. The co-stimulation of anti-CD28 and IL-2 enhances the sensitivity of ELISPOT assays for detection of neoantigen-specific T cells in PBMC. J Immunol Methods 2020; 484-485:112831. [PMID: 32758496 DOI: 10.1016/j.jim.2020.112831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 05/05/2020] [Accepted: 07/29/2020] [Indexed: 12/30/2022]
Abstract
Neoantigen-based cancer immunotherapies hold the promise of being a truly personalized, effective treatment for diverse cancer types. ELISPOT assays, as a powerful experimental technique, can verify the existence of antigen specific T cells to support basic clinical research and monitor clinical trials. However, despite the high sensitivity of ELISPOT assays, detecting immune responses of neoantigen specific T cells in a patient or healthy donor's PBMCs is still extremely difficult, since the frequency of these T cells can be very low. We developed a novel experimental method, by co-stimulation of T cells with anti-CD28 and IL-2 at the beginning of ELISPOT, to further increase the sensitivity of ELISPOT and mitigate the challenge introduced by low frequency T cells. Under the optimal concentration of 1 μg/ml for anti-CD28 and 1 U/ml for IL-2, an 11.7-fold increase of T cell response against CMV peptide was observed by using our method, and it outperforms other cytokine stimulation alternatives (5-10 folds). We also showed that this method can be effectively applied to detect neoantigen-specific T cells in healthy donors' and a melanoma patient's PBMCs. To the best of our knowledge, this is the first report that the co-stimulation of anti-CD28 and IL-2 is able to significantly improve the sensitivity of ELISPOT assays, indicating that anti-CD28 and IL-2 signaling can act in synergy to lower the T cell activation threshold and trigger more neoantigen-specific T cells.
Collapse
Affiliation(s)
- Yunxia Tang
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China; BGI-Shenzhen, Shenzhen 518083, China
| | | | - Qumiao Xu
- BGI-Shenzhen, Shenzhen 518083, China
| | - Xiuqing Zhang
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China; BGI-Shenzhen, Shenzhen 518083, China; BGI-GenoImmune, Wuhan 490079, China.
| | - Bo Li
- BGI-Shenzhen, Shenzhen 518083, China; BGI-GenoImmune, Wuhan 490079, China.
| | - Leo J Lee
- BGI-Shenzhen, Shenzhen 518083, China; BGI-GenoImmune, Wuhan 490079, China; Department of Electrical and Computer Engineering, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3G4, Canada.
| |
Collapse
|
162
|
Waickman AT, Keller HR, Kim TH, Luckey MA, Tai X, Hong C, Molina-París C, Walsh STR, Park JH. The Cytokine Receptor IL-7Rα Impairs IL-2 Receptor Signaling and Constrains the In Vitro Differentiation of Foxp3 + Treg Cells. iScience 2020; 23:101421. [PMID: 32791329 PMCID: PMC7424196 DOI: 10.1016/j.isci.2020.101421] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/16/2020] [Accepted: 07/24/2020] [Indexed: 01/09/2023] Open
Abstract
IL-7 receptor signaling is essential for the generation and maintenance of conventional T cells. Immunosuppressive Foxp3+ Treg cells, however, express uniquely low amounts of the IL-7-proprietary IL-7Rα so that they are impaired in IL-7 signaling. Because Treg cells depend on IL-2, the loss of IL-7Rα has been considered irrelevant for Treg cells. In contrast, here, we report that IL-7Rα downregulation is necessary to maximize IL-2R signaling. Although IL-7Rα overexpression promoted IL-7 signaling, unexpectedly, IL-2 signaling was suppressed in the same cells. Mechanistically, we found that γc, which is a receptor subunit shared by IL-7R and IL-2R, directly binds and pre-associates with IL-7Rα, thus limiting its availability for IL-2R binding. Consequently, overexpression of signaling-deficient, tailless IL-7Rα proteins inhibited IL-2R signaling, demonstrating that IL-7Rα sequesters γc and suppresses IL-2R signaling by extracellular interactions. Collectively, these results reveal a previously unappreciated regulatory mechanism of IL-2 receptor signaling that is governed by IL-7Rα abundance.
Collapse
Affiliation(s)
- Adam T Waickman
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NIH, Bethesda, MD 20892, USA
| | - Hilary R Keller
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NIH, Bethesda, MD 20892, USA; Department of Surgery, Guthrie Robert Packer Hospital, Sayre, PA, USA
| | - Tae-Hyoun Kim
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NIH, Bethesda, MD 20892, USA
| | - Megan A Luckey
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NIH, Bethesda, MD 20892, USA
| | - Xuguang Tai
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NIH, Bethesda, MD 20892, USA
| | - Changwan Hong
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, South Korea
| | - Carmen Molina-París
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds, UK
| | - Scott T R Walsh
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Frederick, MD 21702, USA
| | - Jung-Hyun Park
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
163
|
Seddiki N, Picard F, Dupaty L, Lévy Y, Godot V. The Potential of Immune Modulation in Therapeutic HIV-1 Vaccination. Vaccines (Basel) 2020; 8:vaccines8030419. [PMID: 32726934 PMCID: PMC7565497 DOI: 10.3390/vaccines8030419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 02/07/2023] Open
Abstract
We discuss here some of the key immunological elements that are at the crossroads and need to be combined to develop a potent therapeutic HIV-1 vaccine. Therapeutic vaccines have been commonly used to enhance and/or recall pre-existing HIV-1-specific cell-mediated immune responses aiming to suppress virus replication. The current success of immune checkpoint blockers in cancer therapy renders them very attractive to use in HIV-1 infected individuals with the objective to preserve the function of HIV-1-specific T cells from exhaustion and presumably target the persistent cellular reservoir. The major latest advances in our understanding of the mechanisms responsible for virus reactivation during therapy-suppressed individuals provide the scientific basis for future combinatorial therapeutic vaccine development.
Collapse
Affiliation(s)
- Nabila Seddiki
- Inserm, U955, Equipe 16, 94000 Créteil, France; (F.P.); (L.D.); (Y.L.); (V.G.)
- Faculté de médecine, Université Paris Est, 94000 Créteil, France
- Vaccine Research Institute (VRI), 94000 Créteil, France
- INSERM U955 Equipe 16, Université Paris-Est Créteil, Vaccine Research Institute (VRI), 51, Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
- Correspondence: ; Tel.: +33-01-4981-3902; Fax: +33-01-4981-3709
| | - Florence Picard
- Inserm, U955, Equipe 16, 94000 Créteil, France; (F.P.); (L.D.); (Y.L.); (V.G.)
- Vaccine Research Institute (VRI), 94000 Créteil, France
| | - Léa Dupaty
- Inserm, U955, Equipe 16, 94000 Créteil, France; (F.P.); (L.D.); (Y.L.); (V.G.)
- Vaccine Research Institute (VRI), 94000 Créteil, France
| | - Yves Lévy
- Inserm, U955, Equipe 16, 94000 Créteil, France; (F.P.); (L.D.); (Y.L.); (V.G.)
- Faculté de médecine, Université Paris Est, 94000 Créteil, France
- Vaccine Research Institute (VRI), 94000 Créteil, France
- AP-HP Hôpital H. Mondor—A. Chenevier, Service d’Immunologie clinique et maladies infectieuses, 94010 Créteil, France
| | - Véronique Godot
- Inserm, U955, Equipe 16, 94000 Créteil, France; (F.P.); (L.D.); (Y.L.); (V.G.)
- Faculté de médecine, Université Paris Est, 94000 Créteil, France
- Vaccine Research Institute (VRI), 94000 Créteil, France
| |
Collapse
|
164
|
Zohouri M, Mehdipour F, Razmkhah M, Faghih Z, Ghaderi A. CD4 +CD25 -FoxP3 + T cells: a distinct subset or a heterogeneous population? Int Rev Immunol 2020; 40:307-316. [PMID: 32705909 DOI: 10.1080/08830185.2020.1797005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In addition to generating effective immunity against infectious agents, the immune system helps to fight against different noninfectious human diseases while maintaining the balance between self and non-self discrimination. The breakdown of tolerance in autoimmune diseases or sustainable tolerance in an abnormal microenvironment such as chronic inflammation may initiate the process of malignancy. Immune system regulation is controlled by a complex, dynamic network of cells and mediators. Understanding the cellular and molecular basis of immune regulation provides better insight into the mechanisms governing the immune pathology of diseases. Among several cellular subsets and mediators with regulatory roles, a subpopulation of CD4+ T cells was recently reported to be positive for FoxP3 and negative for CD25, with a suggested range of functional activities in both cancer and autoimmune diseases. This CD4 subset was first reported in 2006 and thought to have a role in the pathogenesis of cancer. However, the spectrum of roles played by this T cell subset is broad, and no consensus has been reached regarding its immunological functions. In this review, we focused on the possible origin of CD4+CD25‒FoxP3+ T cells and their function in cancer and autoimmune diseases.
Collapse
Affiliation(s)
- Mahshid Zohouri
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fereshteh Mehdipour
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahboobeh Razmkhah
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Faghih
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Ghaderi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
165
|
Arroyo Hornero R, Georgiadis C, Hua P, Trzupek D, He LZ, Qasim W, Todd JA, Ferreira RC, Wood KJ, Issa F, Hester J. CD70 expression determines the therapeutic efficacy of expanded human regulatory T cells. Commun Biol 2020; 3:375. [PMID: 32665635 PMCID: PMC7360768 DOI: 10.1038/s42003-020-1097-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 06/17/2020] [Indexed: 12/27/2022] Open
Abstract
Regulatory T cells (Tregs) are critical mediators of immune homeostasis. The co-stimulatory molecule CD27 is a marker of highly suppressive Tregs, although the role of the CD27-CD70 receptor-ligand interaction in Tregs is not clear. Here we show that after prolonged in vitro stimulation, a significant proportion of human Tregs gain stable CD70 expression while losing CD27. The expression of CD70 in expanded Tregs is associated with a profound loss of regulatory function and an unusual ability to provide CD70-directed co-stimulation to TCR-activated conventional T cells. Genetic deletion of CD70 or its blockade prevents Tregs from delivering this co-stimulatory signal, thus maintaining their regulatory activity. High resolution targeted single-cell RNA sequencing of human peripheral blood confirms the presence of CD27-CD70+ Treg cells. These findings have important implications for Treg-based clinical studies where cells are expanded over extended periods in order to achieve sufficient treatment doses.
Collapse
Affiliation(s)
- Rebeca Arroyo Hornero
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Christos Georgiadis
- Molecular and Cellular Immunology Unit, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
| | - Peng Hua
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Dominik Trzupek
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, OX3 7BN, UK
| | - Li-Zhen He
- Celldex Therapeutics, Inc., Hampton, NJ, 08827, USA
| | - Waseem Qasim
- Molecular and Cellular Immunology Unit, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
| | - John A Todd
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, OX3 7BN, UK
| | - Ricardo C Ferreira
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, OX3 7BN, UK
| | - Kathryn J Wood
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Fadi Issa
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Joanna Hester
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK.
| |
Collapse
|
166
|
Khadge S, Sharp JG, Thiele GM, McGuire TR, Talmadge JE. Fatty Acid Mediators in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1259:125-153. [PMID: 32578175 DOI: 10.1007/978-3-030-43093-1_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Patients with cancer frequently overexpress inflammatory cytokines with an associated neutrophilia both of which may be downregulated by diets with high omega-3 polyunsaturated fatty acids (ω-3 PUFA). The anti-inflammatory activity of dietary ω-3 PUFA has been suggested to have anticancer properties and to improve survival of cancer patients. Currently, the majority of dietary research efforts do not differentiate between obesity and dietary fatty acid consumption as mediators of inflammatory cell expansion and tumor microenvironmental infiltration, initiation, and progression. In this chapter, we discuss the relationships between dietary lipids, inflammation, neoplasia and strategies to regulate these relationships. We posit that dietary composition, notably the ratio of ω-3 vs. ω-6 PUFA, regulates tumor initiation and progression and the frequency and sites of metastasis that, together, impact overall survival (OS). We focus on three broad topics: first, the role of dietary lipids in chronic inflammation and tumor initiation, progression, and regression; second, lipid mediators linking inflammation and cancer; and third, dietary lipid regulation of murine and human tumor initiation, progression, and metastasis.
Collapse
Affiliation(s)
- Saraswoti Khadge
- Department of Pathology and Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA.,Vanderbilt University, Nashville, TN, USA
| | - John Graham Sharp
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Geoffrey M Thiele
- Department of Pathology and Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.,Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Timothy R McGuire
- Department of Pharmacy Practice, University of Nebraska Medical Center, Omaha, NE, USA
| | - James E Talmadge
- Department of Pathology and Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA. .,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
167
|
Luo L, Zeng X, Huang Z, Luo S, Qin L, Li S. Reduced frequency and functional defects of CD4 +CD25 highCD127 low/- regulatory T cells in patients with unexplained recurrent spontaneous abortion. Reprod Biol Endocrinol 2020; 18:62. [PMID: 32522204 PMCID: PMC7285476 DOI: 10.1186/s12958-020-00619-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 06/02/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Unexplained recurrent spontaneous abortion (URSA) is defined as two or more consecutive pregnancy losses, generally of unknown cause; it is related to a failure of fetal-maternal immunological tolerance. Regulatory T cells (Tregs) exert immunosuppressive effects, which are essential to maintain fetal-maternal immunological tolerance and regulate immune balance. In this study, we used the specific cell-surface phenotype of CD4+CD25highCD127low/- Tregs to investigate the number and suppressive function of Tregs isolated from the peripheral blood of patients with URSA with the aim of expanding our understanding of their role in URSA. METHODS We isolated a relatively pure population of peripheral CD4+CD25highCD127low/- Tregs and CD4+CD25- responder T cells (Tresps) from the patients with URSA and normal fertile nonpregnant control women via fluorescence-activated cell sorting. We compared the frequency, suppressive capacity, and forkhead box transcription factor P3 (FOXP3) expression of Tregs in the peripheral blood between patients with URSA and normal controls. RESULTS The frequency of CD4+CD25highCD127low/- Tregs in the peripheral blood was lower in URSA patients than in the controls (P < 0.05). The mean fluorescence intensity of FOXP3 and FOXP3 mRNA expression in Tregs was also significantly lower in the URSA patients (P < 0.01). Tregs suppressed the activity of autologous Tresps stimulated with anti-CD3/CD28 beads in a concentration-dependent manner, with the strongest suppression occurring in cocultures with a 1:1 Treg:Tresp ratio in both groups; however, patient-derived Tregs exhibited a poorer capacity to suppress the proliferation of autologous Tresps than the Tregs from normal controls (P < 0.01). Moreover, Tregs isolated from URSA patients inhibited the proliferation of Tresps from normal controls less potently than the Tregs from normal controls (P < 0.01), and Tresps from URSA patients were less effectively suppressed by autologous Tregs than by those from normal controls (P < 0.01). Tresp activity were intact in both groups. CONCLUSIONS We observed a lower frequency of peripheral CD4+CD25highCD127low/- Tregs with lower FOXP3 expression in the peripheral blood of URSA patients. In addition, highly purified Tregs from patients with URSA exhibited impaired suppressive effects. The defect in immune regulation in URSA patients appears to be primarily related to impaired Tregs, and not to increased resistance of Tresps to suppression. Our findings reveal a potential novel therapeutic target for URSA.
Collapse
Affiliation(s)
- Li Luo
- grid.461863.e0000 0004 1757 9397Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- grid.13291.380000 0001 0807 1581Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Xun Zeng
- grid.461863.e0000 0004 1757 9397Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- grid.13291.380000 0001 0807 1581Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Zhongying Huang
- grid.461863.e0000 0004 1757 9397Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- grid.13291.380000 0001 0807 1581Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Shan Luo
- grid.461863.e0000 0004 1757 9397Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- grid.13291.380000 0001 0807 1581Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Lang Qin
- grid.461863.e0000 0004 1757 9397Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- grid.13291.380000 0001 0807 1581Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Shangwei Li
- grid.461863.e0000 0004 1757 9397Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- grid.13291.380000 0001 0807 1581Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| |
Collapse
|
168
|
Namdeo M, Kandel R, Thakur PK, Mohan A, Dey AB, Mitra DK. Old age-associated enrichment of peripheral T regulatory cells and altered redox status in pulmonary tuberculosis patients. Eur J Immunol 2020; 50:1195-1208. [PMID: 32365223 DOI: 10.1002/eji.201948261] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 02/23/2020] [Accepted: 04/28/2020] [Indexed: 12/29/2022]
Abstract
Aging influences the susceptibility and prognosis to various infectious diseases including tuberculosis (TB). Despite the impairment of T-cell function and immunity in older individuals, the mechanism for the higher incidence of TB in the elderly remains largely unknown. Here, we evaluated the age-associated immune alterations, particularly in effector and Treg responses in pulmonary TB patients. We also evaluated the impact of redox status and its modulation with N-acetyl-cysteine (NAC) in elderly TB. Higher frequency of Treg cells and reduced IFN-γ positive T cells were observed among older TB patients. The elevated number of Treg cells correlated tightly with bacillary load (i.e. disease severity); which declined significantly in response to successful anti-tubercular treatment. We could rescue Myobacterium tuberculosis-specific effector T cell (Th1) responses through various in vitro approaches, for example, Treg cell depletion and co-culture experiments, blocking experiments using antibodies against IL-10, TGF-β, and programmed death-1 (PD-1) as well as NAC supplementation. We report old age-associated enrichment of Treg cells and suppression of M. tuberculosis-specific effector T (Th1) cell immune responses. Monitoring these immune imbalances in older patients may assist in immune potentiation through selectively targeting Treg cells and/or optimizing redox status by NAC supplementation.
Collapse
Affiliation(s)
- Manju Namdeo
- Department of Transplant Immunology and Immunogenetics, All India Institute of Medical Sciences, New Delhi, India
| | - Ramesh Kandel
- Department of Geriatric Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Prabhakar Kumar Thakur
- Department of Geriatric Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Anant Mohan
- Department of Pulmonary Critical Care and Sleep Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Aparajit Ballav Dey
- Department of Geriatric Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Dipendra Kumar Mitra
- Department of Transplant Immunology and Immunogenetics, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
169
|
Mitchell J, Kelly J, Kvedaraite E, von Bahr Greenwood T, Henter JI, Pellicci DG, Berzins SP, Kannourakis G. Foxp3+ Tregs from Langerhans cell histiocytosis lesions co-express CD56 and have a definitively regulatory capacity. Clin Immunol 2020; 215:108418. [DOI: 10.1016/j.clim.2020.108418] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 03/30/2020] [Accepted: 04/09/2020] [Indexed: 12/13/2022]
|
170
|
Walton K, Fernandez MR, Sagatys EM, Reff J, Kim J, Lee MC, Kiluk JV, Hui JYC, McKenna D, Hupp M, Forster C, Linden MA, Lawrence NJ, Lawrence HR, Pidala J, Pavletic SZ, Blazar BR, Sebti SM, Cleveland JL, Anasetti C, Betts BC. Metabolic reprogramming augments potency of human pSTAT3-inhibited iTregs to suppress alloreactivity. JCI Insight 2020; 5:136437. [PMID: 32255769 DOI: 10.1172/jci.insight.136437] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/01/2020] [Indexed: 12/18/2022] Open
Abstract
Immunosuppressive donor Tregs can prevent graft-versus-host disease (GVHD) or solid-organ allograft rejection. We previously demonstrated that inhibiting STAT3 phosphorylation (pSTAT3) augments FOXP3 expression, stabilizing induced Tregs (iTregs). Here we report that human pSTAT3-inhibited iTregs prevent human skin graft rejection and xenogeneic GVHD yet spare donor antileukemia immunity. pSTAT3-inhibited iTregs express increased levels of skin-homing cutaneous lymphocyte-associated antigen, immunosuppressive GARP and PD-1, and IL-9 that supports tolerizing mast cells. Further, pSTAT3-inhibited iTregs significantly reduced alloreactive conventional T cells, Th1, and Th17 cells implicated in GVHD and tissue rejection and impaired infiltration by pathogenic Th2 cells. Mechanistically, pSTAT3 inhibition of iTregs provoked a shift in metabolism from oxidative phosphorylation (OxPhos) to glycolysis and reduced electron transport chain activity. Strikingly, cotreatment with coenzyme Q10 restored OxPhos in pSTAT3-inhibited iTregs and augmented their suppressive potency. These findings support the rationale for clinically testing the safety and efficacy of metabolically tuned, human pSTAT3-inhibited iTregs to control alloreactive T cells.
Collapse
Affiliation(s)
- Kelly Walton
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | | | | - Jongphil Kim
- Department of Biostatistics and Bioinformatics, and
| | | | - John V Kiluk
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | | | - David McKenna
- Department of Laboratory Medicine and Pathology, and
| | - Meghan Hupp
- Department of Laboratory Medicine and Pathology, and
| | - Colleen Forster
- Bionet Histology Research Laboratory, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | | | | - Joseph Pidala
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida, USA
| | - Steven Z Pavletic
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Said M Sebti
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia USA
| | | | - Claudio Anasetti
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida, USA
| | - Brian C Betts
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
171
|
Truffault F, Nazzal D, Verdier J, Gradolatto A, Fadel E, Roussin R, Eymard B, Le Panse R, Berrih-Aknin S. Comparative Analysis of Thymic and Blood Treg in Myasthenia Gravis: Thymic Epithelial Cells Contribute to Thymic Immunoregulatory Defects. Front Immunol 2020; 11:782. [PMID: 32435245 PMCID: PMC7218102 DOI: 10.3389/fimmu.2020.00782] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/06/2020] [Indexed: 11/13/2022] Open
Abstract
The thymus is involved in autoimmune Myasthenia gravis (MG) associated with anti-acetylcholine (AChR) antibodies. In MG, thymic regulatory T cells (Treg) are not efficiently suppressive, and conventional T cells (Tconv) are resistant to suppression. To better understand the specific role of the thymus in MG, we compared the phenotype and function of peripheral and thymic Treg and Tconv from controls and MG patients. Suppression assays with thymic or peripheral CD4 + T cells showed that the functional impairment in MG was more pronounced in the thymus than in the periphery. Phenotypic analysis of Treg showed a significant reduction of resting and effector Treg in the thymus but not in the periphery of MG patients. CD31, a marker lost with excessive immunoreactivity, was significantly reduced in thymic but not blood resting Treg. These results suggest that an altered thymic environment may explain Treg differences between MG patients and controls. Since thymic epithelial cells (TECs) play a major role in the generation of Treg, we co-cultured healthy thymic CD4 + T cells with control or MG TECs and tested their suppressive function. Co-culture with MG TECs consistently hampers regulatory activity, as compared with control TECs, suggesting that MG TECs contribute to the immune regulation defects of MG CD4 + T cells. MG TECs produced significantly higher thymic stromal lymphopoietin (TSLP) than control TECs, and a neutralizing anti-TSLP antibody partially restored the suppressive capacity of Treg derived from co-cultures with MG TECs, suggesting that TSLP contributed to the defect of thymic Treg in MG patients. Finally, a co-culture of MG CD4 + T cells with control TECs restored numbers and function of MG Treg, demonstrating that a favorable environment could correct the immune regulation defects of T cells in MG. Altogether, our data suggest that the severe defect of thymic Treg is at least partially due to MG TECs that overproduce TSLP. The Treg defects could be corrected by replacing dysfunctional TECs by healthy TECs. These findings highlight the role of the tissue environment on the immune regulation.
Collapse
Affiliation(s)
- Frédérique Truffault
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Dani Nazzal
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Julien Verdier
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Angeline Gradolatto
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Elie Fadel
- Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | | | - Bruno Eymard
- AP-HP, Referral Center for Neuromuscular Disorders, Pitié-Salpêtrière Hospital, Institute of Myology, Paris, France
| | - Rozen Le Panse
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Sonia Berrih-Aknin
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| |
Collapse
|
172
|
Opstelten R, de Kivit S, Slot MC, van den Biggelaar M, Iwaszkiewicz-Grześ D, Gliwiński M, Scott AM, Blom B, Trzonkowski P, Borst J, Cuadrado E, Amsen D. GPA33: A Marker to Identify Stable Human Regulatory T Cells. THE JOURNAL OF IMMUNOLOGY 2020; 204:3139-3148. [DOI: 10.4049/jimmunol.1901250] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 04/16/2020] [Indexed: 12/16/2022]
|
173
|
Mapping the extent of heterogeneity of human CCR5+ CD4+ T cells in peripheral blood and lymph nodes. AIDS 2020; 34:833-848. [PMID: 32044843 DOI: 10.1097/qad.0000000000002503] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND CD4 T cells that express the chemokine receptor, CCR5, are the most important target of HIV-1 infection, but their functions, phenotypes and anatomical locations are poorly understood. We aimed to use multiparameter flow cytometry to better define the full breadth of these cells. METHODS High-parameter fluorescence flow and mass cytometry were optimized to analyse subsets of CCR5 memory CD4 T cells, including CD25CD127 Tregs, CXCR3CCR6- Th1-like, CCR6CD161CXCR3- Th17-like, integrins α4ß7 gut-homing, CCR4 skin-homing, CD62L lymph node-homing, CD38HLA-DR activated cells, and CD27-CD28- cytotoxic T lymphocytes, in a total of 22 samples of peripheral blood, ultrasound-guided fine needle biopsies of lymph nodes and excised tonsils. CCR5 antigen-specific CD4 T cells were studied using the OX40 flow-based assay. RESULTS 10-20% of CCR5 memory CD4 T cells were Tregs, 10-30% were gut-homing, 10-30% were skin-homing, 20-40% were lymph node-homing, 20-50% were Th1-like and 20-40% were Th17-like cells. Up to 30% were cytotoxic T lymphocytes in CMV-seropositive donors, including cells that were either CCR5Granzyme K or CCR5Granzyme B. When all possible phenotypes were exhaustively analysed, more than 150 different functional and trafficking subsets of CCR5 CD4 T cells were seen. Moreover, a small population of resident CD69Granzyme KCCR5 CD4 T cells was found in lymphoid tissues. CMV- and Mycobacterium tuberculosis-specific CD4 T cells were predominantly CCR5. CONCLUSION These results reveal for the first time the prodigious heterogeneity of function and trafficking of CCR5 CD4 T cells in blood and in lymphoid tissue, with significant implications for rational approaches to prophylaxis for HIV-1 infection and for purging of the HIV-1 reservoir in those participants already infected.
Collapse
|
174
|
Platzer H, Nees TA, Reiner T, Tripel E, Gantz S, Hagmann S, Moradi B, Rosshirt N. Impact of Mononuclear Cell Infiltration on Chondrodestructive MMP/ADAMTS Production in Osteoarthritic Knee Joints-An Ex Vivo Study. J Clin Med 2020; 9:jcm9051279. [PMID: 32354196 PMCID: PMC7288002 DOI: 10.3390/jcm9051279] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 12/15/2022] Open
Abstract
Progressive loss of joint function in osteoarthritis (OA) is driven by degenerative and inflammatory processes and their complex interaction. Decoding the link between degeneration and inflammation is one of the most exciting approaches in understanding OA pathophysiology and holds the promise to open new therapeutic avenues. The overarching goal of this project was to analyze the impact of mononuclear cells (MNC) on enzymatic chondrodestructive processes (MMP/ADAMTS) in OA. Synovial membrane (SM), articular cartilage (AC) and peripheral blood (PB) were obtained from a total of 21 patients with advanced knee OA who underwent arthroplastic surgery. In supernatants of native synovial cell cultures, T cell-depleted synovial cell cultures and macrophage-depleted synovial cell cultures, the concentrations of various metalloproteinases were examined by Enzyme Linked Immunosorbent Assay (ELISA). Furthermore, ELISA was used to analyze concentrations of metalloproteinases in supernatants of chondrocyte monocultures and chondrocyte co-cultures with CD4+CD127dim/- enriched peripheral blood mononuclear cells (PBMC), Treg depleted CD4+CD25-CD127dim/- enriched PBMC and CD4+CD25+CD127dim/- Treg. Compared to native synovial cell culture, T cell depletion led to significantly lower levels of MMP-1, MMP-3 and MMP-9 and macrophage depletion led to a significant decline of MMP-1, MMP-3, MMP-9 and ADAMTS-5 concentration. Compared to T cell depletion, macrophage depletion resulted in a significantly stronger reduction of MMP-1, MMP-3, MMP-9 and ADAMTS-5. In chondrocyte co-culture with CD4+CD127dim/- enriched PBMC the concentration of MMP-1 and ADAMTS-5 was significantly increased compared to chondrocyte monoculture. No significant differences were found between chondrocyte monoculture and chondrocyte co-culture with Treg as well as between co-culture with CD4+CD127dim/- enriched PBMC containing Treg and co-culture with Treg-depleted CD4+CD25-CD127dim/- enriched PBMC. In conclusion, our data suggests that both synovial macrophages and T cells have a catabolic potential by inducing the release of chondrodestructive metalloproteinases in OA synovium. This study also supports the hypothesis that MNC affect the release of metalloproteinases by chondrocytes and are hereby involved in the cartilage-induced chondrodestructive process. In this study no suppressive effect of Treg was shown.
Collapse
|
175
|
Zhang X, Olsen N, Zheng SG. The progress and prospect of regulatory T cells in autoimmune diseases. J Autoimmun 2020; 111:102461. [PMID: 32305296 DOI: 10.1016/j.jaut.2020.102461] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 12/16/2022]
Abstract
Regulatory T cells (Treg) are an important immune cell population, playing a crucial role in regulating immune tolerance and preventing autoimmune diseases. These cells consist of various cell sub-populations and generally have an immunoregulatory or suppressive role against immune responses. They also have a different cell heterogeneity and each populations has own biological characteristics. Treg deficiency, reduction, instability, reduced vitality and dysfunction all account for multiple autoimmune diseases. In this review, we have systemically reviewed Treg classification, phenotypic features, regulation of Foxp3 expression, plasticity and stability of Treg as well as their relationship with several important autoimmune diseases. We particularly focus on why and how inflammatory and diet environments affect the functional capacity and underlying mechanisms of Treg cell populations. We also summarize new advances in technologies which help to analyze and dissect these cells in molecular levels in-depth. We also clarify the possible clinical relevance on application of these cells in patients with autoimmune diseases. The advantages and weaknesses have been carefully discussed as well. We also propose the possible approaches to overcome these weaknesses of Treg cells in complicate environments. Thus, we have displayed the updated knowledge of Treg cells, which provides an overall insight into the role and mechanisms of Treg cells in autoimmune diseases.
Collapse
Affiliation(s)
- Ximei Zhang
- Institute of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China; Division of Rheumatology and Immunology, Department of Internal Medicine, Ohio State University College of Medicine, Columbus, PA, 43201, USA
| | - Nancy Olsen
- Division of Rheumatology, Department of Medicine at Penn State College of Medicine and Milton S. Hershey Medical Center, Hershey, 17033, USA
| | - Song Guo Zheng
- Division of Rheumatology and Immunology, Department of Internal Medicine, Ohio State University College of Medicine, Columbus, PA, 43201, USA.
| |
Collapse
|
176
|
Rackaityte E, Halkias J. Mechanisms of Fetal T Cell Tolerance and Immune Regulation. Front Immunol 2020; 11:588. [PMID: 32328065 PMCID: PMC7160249 DOI: 10.3389/fimmu.2020.00588] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/13/2020] [Indexed: 12/19/2022] Open
Abstract
The developing human fetus generates both tolerogenic and protective immune responses in response to the unique requirements of gestation. Thus, a successful human pregnancy depends on a fine balance between two opposing immunological forces: the semi-allogeneic fetus learns to tolerate both self- and maternal- antigens and, in parallel, develops protective immunity in preparation for birth. This critical window of immune development bridges prenatal immune tolerance with the need for postnatal environmental protection, resulting in a vulnerable neonatal period with heightened risk of infection. The fetal immune system is highly specialized to mediate this transition and thus serves a different function from that of the adult. Adaptive immune memory is already evident in the fetal intestine. Fetal T cells with pro-inflammatory potential are born in a tolerogenic environment and are tightly controlled by both cell-intrinsic and -extrinsic mechanisms, suggesting that compartmentalization and specialization, rather than immaturity, define the fetal immune system. Dysregulation of fetal tolerance generates an inflammatory response with deleterious effects to the pregnancy. This review aims to discuss the recent advances in our understanding of the cellular and molecular composition of fetal adaptive immunity and the mechanisms that govern T cell development and function. We also discuss the tolerance promoting environment that impacts fetal immunity and the consequences of its breakdown. A greater understanding of fetal mechanisms of immune activation and regulation has the potential to uncover novel paradigms of immune balance which may be leveraged to develop therapies for transplantation, autoimmune disease, and birth-associated inflammatory pathologies.
Collapse
Affiliation(s)
- Elze Rackaityte
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, United States
| | - Joanna Halkias
- Division of Neonatology, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, CA, United States
| |
Collapse
|
177
|
Sturm R, Xanthopoulos L, Heftrig D, Oppermann E, Vrdoljak T, Dunay IR, Marzi I, Relja B. Regulatory T Cells Modulate CD4 Proliferation after Severe Trauma via IL-10. J Clin Med 2020; 9:jcm9041052. [PMID: 32276346 PMCID: PMC7230720 DOI: 10.3390/jcm9041052] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 12/19/2022] Open
Abstract
Objective: Severely injured patients frequently develop an immunological imbalance following the traumatic insult, which might result in infectious complications evoked by a persisting immunosuppression. Regulatory T cells (Tregs) maintain the immune homeostasis by suppressing proinflammatory responses, however, their functionality after trauma is unclear. Here, we characterized the role of Tregs in regulating the proliferation of CD4+ lymphocytes in traumatized patients (TP). Methods: Peripheral blood was obtained daily from 29 severely injured TP (Injury Severity Score, ISS ≥16) for ten days following admission to the emergency department (ED). Ten healthy volunteers (HV) served as controls. The frequency and activity of Tregs were assessed by flow cytometry. Proliferation of CD4+ cells was analyzed either in presence or absence of Tregs, or after blocking of either IL-10 or IL-10R1. Results: The frequencies of CD4+CD25high and CD4+CD25+CD127− Tregs were significantly decreased immediately upon admission of TP to the ED and during the following 10 post-injury days. Compared with HV CD4+ T cell proliferation in TP increased significantly upon their admission and on the following days. As expected, CD4+CD25+CD127− Tregs reduced the proliferation of CD4+ cells in HV, nevertheless, CD4+ proliferation in TP was increased by Tregs. Neutralization of IL-10 as well as blocking the IL-10R1 increased further CD4+ T cell proliferation in Tregs-depleted cultures, thereby confirming an IL-10-mediated mechanism of IL-10-regulated CD4+ T cell proliferation. Neutralization of IL-10 in TP decreased CD4+ T cell proliferation in Tregs-depleted cultures, whereas blocking of the IL-10R1 receptor had no significant effects. Conclusions: The frequency of Tregs in the CD4+ T lymphocyte population is reduced after trauma; however, their inductiveness is increased. The mechanisms of deregulated influence of Tregs on CD4+ T cell proliferation are mediated via IL-10 but not via the IL-10R1.
Collapse
Affiliation(s)
- Ramona Sturm
- Department of Trauma Surgery, Goethe University, 60590 Frankfurt, Germany; (R.S.); (L.X.); (I.M.)
| | - Lara Xanthopoulos
- Department of Trauma Surgery, Goethe University, 60590 Frankfurt, Germany; (R.S.); (L.X.); (I.M.)
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, 39120 Magdeburg, Germany;
| | - David Heftrig
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, 39120 Magdeburg, Germany;
- Clinic for Radiology, Neuroradiology and Nuclear Medicine, Klinikum Frankfurt Höchst, 60590 Frankfurt, Germany
| | - Elsie Oppermann
- Department of General and Visceral Surgery, University Hospital Frankfurt, Goethe-University, 60590 Frankfurt, Germany;
| | - Teodora Vrdoljak
- Department of Diagnostic and Interventional Radiology, University Hospital Dubrava, University of Zagreb School of Medicine, 10000 Zagreb, Croatia;
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany;
| | - Ingo Marzi
- Department of Trauma Surgery, Goethe University, 60590 Frankfurt, Germany; (R.S.); (L.X.); (I.M.)
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, 39120 Magdeburg, Germany;
- Correspondence: ; Tel.: +49-391-67-28242
| |
Collapse
|
178
|
Snyder ME, Finlayson MO, Connors TJ, Dogra P, Senda T, Bush E, Carpenter D, Marboe C, Benvenuto L, Shah L, Robbins H, Hook JL, Sykes M, D'Ovidio F, Bacchetta M, Sonett JR, Lederer DJ, Arcasoy S, Sims PA, Farber DL. Generation and persistence of human tissue-resident memory T cells in lung transplantation. Sci Immunol 2020; 4:4/33/eaav5581. [PMID: 30850393 DOI: 10.1126/sciimmunol.aav5581] [Citation(s) in RCA: 214] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/28/2019] [Indexed: 12/20/2022]
Abstract
Tissue-resident memory T cells (TRM) maintain immunity in diverse sites as determined in mouse models, whereas their establishment and role in human tissues have been difficult to assess. Here, we investigated human lung TRM generation, maintenance, and function in airway samples obtained longitudinally from human leukocyte antigen (HLA)-disparate lung transplant recipients, where donor and recipient T cells could be localized and tracked over time. Donor T cells persist specifically in the lungs (and not blood) of transplant recipients and express high levels of TRM signature markers including CD69, CD103, and CD49a, whereas lung-infiltrating recipient T cells gradually acquire TRM phenotypes over months in vivo. Single-cell transcriptome profiling of airway T cells reveals that donor T cells comprise two TRM-like subsets with varying levels of expression of TRM-associated genes, whereas recipient T cells comprised non-TRM and similar TRM-like subpopulations, suggesting de novo TRM generation. Transplant recipients exhibiting higher frequencies of persisting donor TRM experienced fewer adverse clinical events such as primary graft dysfunction and acute cellular rejection compared with recipients with low donor TRM persistence, suggesting that monitoring TRM dynamics could be clinically informative. Together, our results provide spatial and temporal insights into how human TRM develop, function, persist, and affect tissue integrity within the complexities of lung transplantation.
Collapse
Affiliation(s)
- Mark E Snyder
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA.,Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Michael O Finlayson
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Thomas J Connors
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA.,Department of Pediatrics, Columbia University Medical Center, New York, NY 10032, USA
| | - Pranay Dogra
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA.,Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Takashi Senda
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA.,Department of Surgery, Columbia University Medical Center, New York, NY 10032, USA
| | - Erin Bush
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Dustin Carpenter
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA.,Department of Surgery, Columbia University Medical Center, New York, NY 10032, USA
| | - Charles Marboe
- Department of Pathology, Columbia University Medical Center, New York, NY 10032, USA
| | - Luke Benvenuto
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Lori Shah
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Hilary Robbins
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Jaime L Hook
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Megan Sykes
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA.,Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA.,Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Frank D'Ovidio
- Department of Surgery, Columbia University Medical Center, New York, NY 10032, USA
| | - Matthew Bacchetta
- Department of Surgery, Columbia University Medical Center, New York, NY 10032, USA
| | - Joshua R Sonett
- Department of Surgery, Columbia University Medical Center, New York, NY 10032, USA
| | - David J Lederer
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA.,Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
| | - Selim Arcasoy
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA.,Department of Pediatrics, Columbia University Medical Center, New York, NY 10032, USA
| | - Peter A Sims
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Donna L Farber
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA. .,Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA.,Department of Surgery, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
179
|
Marsh-Wakefield F, Byrne SN, Hawke S, Grau G. Mass cytometry provides unprecedented insight into the role of B cells during the pathogenesis of multiple sclerosis. ADVANCES IN CLINICAL NEUROSCIENCE & REHABILITATION 2020. [DOI: 10.47795/fzhz8873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
180
|
Naisbitt DJ, Olsson‐Brown A, Gibson A, Meng X, Ogese MO, Tailor A, Thomson P. Immune dysregulation increases the incidence of delayed-type drug hypersensitivity reactions. Allergy 2020; 75:781-797. [PMID: 31758810 DOI: 10.1111/all.14127] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 11/05/2019] [Accepted: 11/18/2019] [Indexed: 12/14/2022]
Abstract
Delayed-type, T cell-mediated, drug hypersensitivity reactions are a serious unwanted manifestation of drug exposure that develops in a small percentage of the human population. Drugs and drug metabolites are known to interact directly and indirectly (through irreversible protein binding and processing to the derived adducts) with HLA proteins that present the drug-peptide complex to T cells. Multiple forms of drug hypersensitivity are strongly linked to expression of a single HLA allele, and there is increasing evidence that drugs and peptides interact selectively with the protein encoded by the HLA allele. Despite this, many individuals expressing HLA risk alleles do not develop hypersensitivity when exposed to culprit drugs suggesting a nonlinear, multifactorial relationship in which HLA risk alleles are one factor. This has prompted a search for additional susceptibility factors. Herein, we argue that immune regulatory pathways are one key determinant of susceptibility. As expression and activity of these pathways are influenced by disease, environmental and patient factors, it is currently impossible to predict whether drug exposure will result in a health benefit, hypersensitivity or both. Thus, a concerted effort is required to investigate how immune dysregulation influences susceptibility towards drug hypersensitivity.
Collapse
Affiliation(s)
- Dean J. Naisbitt
- MRC Centre for Drug Safety Science Department of Clinical and Molecular Pharmacology The University of Liverpool Liverpool UK
| | - Anna Olsson‐Brown
- MRC Centre for Drug Safety Science Department of Clinical and Molecular Pharmacology The University of Liverpool Liverpool UK
| | - Andrew Gibson
- MRC Centre for Drug Safety Science Department of Clinical and Molecular Pharmacology The University of Liverpool Liverpool UK
| | - Xiaoli Meng
- MRC Centre for Drug Safety Science Department of Clinical and Molecular Pharmacology The University of Liverpool Liverpool UK
| | - Monday O. Ogese
- MRC Centre for Drug Safety Science Department of Clinical and Molecular Pharmacology The University of Liverpool Liverpool UK
| | - Arun Tailor
- MRC Centre for Drug Safety Science Department of Clinical and Molecular Pharmacology The University of Liverpool Liverpool UK
| | - Paul Thomson
- MRC Centre for Drug Safety Science Department of Clinical and Molecular Pharmacology The University of Liverpool Liverpool UK
| |
Collapse
|
181
|
Hessam S, Gambichler T, Höxtermann S, Skrygan M, Sand M, Garcovich S, Meyer T, Stockfleth E, Bechara F. Frequency of circulating subpopulations of T‐regulatory cells in patients with hidradenitis suppurativa. J Eur Acad Dermatol Venereol 2020; 34:834-838. [DOI: 10.1111/jdv.16071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/24/2019] [Indexed: 12/14/2022]
Affiliation(s)
- S. Hessam
- Department of Dermatology, Venereology and Allergology Ruhr‐University Bochum Bochum Germany
| | - T. Gambichler
- Department of Dermatology, Venereology and Allergology Ruhr‐University Bochum Bochum Germany
| | - S. Höxtermann
- Department of Dermatology, Venereology and Allergology Ruhr‐University Bochum Bochum Germany
| | - M. Skrygan
- Department of Dermatology, Venereology and Allergology Ruhr‐University Bochum Bochum Germany
| | - M. Sand
- Department of Dermatology, Venereology and Allergology Ruhr‐University Bochum Bochum Germany
- Department of Plastic and Reconstructive Surgery St. Josef‐Hospital Essen‐Kupferdreh Germany
| | - S. Garcovich
- Institute of Dermatology F. Policlinico Gemelli IRCCS Università Cattolica del Sacro Cuore Rome Italy
| | - T. Meyer
- Department of Dermatology, Venereology and Allergology Ruhr‐University Bochum Bochum Germany
| | - E. Stockfleth
- Department of Dermatology, Venereology and Allergology Ruhr‐University Bochum Bochum Germany
| | - F.G. Bechara
- Department of Dermatology, Venereology and Allergology Ruhr‐University Bochum Bochum Germany
| |
Collapse
|
182
|
Guedes MCE, Arroz MJ, Martins C, Angelo-Dias M, Proença RD, Borrego LM. Regulatory T cells and IL-17A levels in noninfectious uveitis. Graefes Arch Clin Exp Ophthalmol 2020; 258:1269-1278. [PMID: 32200408 DOI: 10.1007/s00417-020-04649-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/06/2020] [Accepted: 03/13/2020] [Indexed: 12/28/2022] Open
Abstract
PURPOSE Regulatory T cells (Tregs) have been intensively studied in a myriad of autoimmune diseases. As for noninfectious uveitis (NIU), results have been contradictory, and studies have failed to demonstrate a consistent reduction in Treg cell frequency in patients with active disease. The present study aims to characterize T lymphocyte subsets, including naïve and memory Tregs as well as their respective CD39 expression, in the peripheral blood of NIU patients. Inflammatory as well as suppressive cytokine profiles were also evaluated. METHODS T cell subpopulations were evaluated by multiparametric flow cytometry using anti-CD3, anti-CD4, anti-CD45, anti-CD45RA, anti-CD197, anti-CD25, anti-CD127, and anti-CD39. Treg cells were defined as CD3 + CD4+CD25hiCD127low. A multiplex bead-based immunoassay was used to determine TNF-α, IFN-ɣ, IL-17A, IL-10, and TGF-β levels. RESULTS Twenty-nine patients with active NIU were included as well as 15 sex- and age-matched controls. There were no significant differences in T lymphocyte subsets, including Tregs, between patients and controls. However, patients with a lower grade of anterior chamber or vitreous inflammatory cellular reaction showed higher memory Treg counts than controls, with no respective increase in CD39+ expression, and a tendency for higher IL-17A levels (p = 0.06). This IL-17A elevation was present in the total NIU group (p = 0.08) as well as a positive correlation between IL-17A levels and the absolute counts of memory Tregs (p = 0.013; R = 0.465). Patients with higher IL-17A levels also showed higher serum concentrations of memory (p = 0.001) and naïve (p = 0.003) Tregs as well as elevated TNF-α (p < 0.0001) and IFN-ɣ (p = 0.016) levels. Negative correlations were observed between IL-10 and TGF-β levels and the percentages of memory (p = 0.030; R = - 0.411) and total CD39+ Tregs (p = 0.051; R = - 0.373) in the peripheral blood of NIU patients. CONCLUSION Our results showed that total Treg levels were not reduced in patients with NIU. Further characterization of Treg subsets, including memory Tregs and respective CD39 expression, may provide additional insight on the role of Treg cells in NIU. Consistent high levels of circulating IL-17A in NIU patients are in accordance with previous studies and reinforce this cytokine's vital role in uveitis pathogenesis and its possible use as a therapeutic target.
Collapse
Affiliation(s)
| | - Maria Jorge Arroz
- Clinical Pathology Department, Western Lisbon Hospital Center-São Francisco Xavier Hospital, Lisbon, Portugal
| | - Catarina Martins
- CEDOC, NOVA Medical School and Comprehensive Health Research Center (CHRC), Lisbon, Portugal
| | - Miguel Angelo-Dias
- CEDOC, NOVA Medical School and Comprehensive Health Research Center (CHRC), Lisbon, Portugal
| | - Rui Daniel Proença
- Coimbra Surgical Center and Coimbra Hospital and Universitary Center, Coimbra, Portugal
| | - Luis Miguel Borrego
- Luz Lisbon Hospital and CEDOC, NOVA Medical School and Comprehensive Health Research Center (CHRC), Lisbon, Portugal
| |
Collapse
|
183
|
Singh AK, Salwe S, Padwal V, Velhal S, Sutar J, Bhowmick S, Mukherjee S, Nagar V, Patil P, Patel V. Delineation of Homeostatic Immune Signatures Defining Viremic Non-progression in HIV-1 Infection. Front Immunol 2020; 11:182. [PMID: 32194543 PMCID: PMC7066316 DOI: 10.3389/fimmu.2020.00182] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 01/23/2020] [Indexed: 01/07/2023] Open
Abstract
Viremic non-progressors (VNPs), a distinct group of HIV-1-infected individuals, exhibit no signs of disease progression and maintain persistently elevated CD4+ T cell counts for several years despite high viral replication. Comprehensive characterization of homeostatic cellular immune signatures in VNPs can provide unique insights into mechanisms responsible for coping with viral pathogenesis as well as identifying strategies for immune restoration under clinically relevant settings such as antiretroviral therapy (ART) failure. We report a novel homeostatic signature in VNPs, the preservation of the central memory CD4+ T cell (CD4+ TCM) compartment. In addition, CD4+ TCM preservation was supported by ongoing interleukin-7 (IL-7)-mediated thymic repopulation of naive CD4+ T cells leading to intact CD4+ T cell homeostasis in VNPs. Regulatory T cell (Treg) expansion was found to be a function of preserved CD4+ T cell count and CD4+ T cell activation independent of disease status. However, in light of continual depletion of CD4+ T cell count in progressors but not in VNPs, Tregs appear to be involved in lack of disease progression despite high viremia. In addition to these homeostatic mechanisms resisting CD4+ T cell depletion in VNPs, a relative diminution of terminally differentiated effector subset was observed exclusively in these individuals that might ameliorate consequences of high viral replication. VNPs also shared signatures of impaired CD8+ T cell cytotoxic function with progressors evidenced by increased exhaustion (PD-1 upregulation) and CD127 (IL-7Rα) downregulation contributing to persistent viremia. Thus, the homeostatic immune signatures reported in our study suggest a complex multifactorial mechanism accounting for non-progression in VNPs.
Collapse
Affiliation(s)
- Amit Kumar Singh
- Department of Biochemistry and Virology, Indian Council of Medical Research (ICMR)-National Institute for Research in Reproductive Health, Mumbai, India
| | - Sukeshani Salwe
- Department of Biochemistry and Virology, Indian Council of Medical Research (ICMR)-National Institute for Research in Reproductive Health, Mumbai, India
| | - Varsha Padwal
- Department of Biochemistry and Virology, Indian Council of Medical Research (ICMR)-National Institute for Research in Reproductive Health, Mumbai, India
| | - Shilpa Velhal
- Department of Biochemistry and Virology, Indian Council of Medical Research (ICMR)-National Institute for Research in Reproductive Health, Mumbai, India
| | - Jyoti Sutar
- Department of Biochemistry and Virology, Indian Council of Medical Research (ICMR)-National Institute for Research in Reproductive Health, Mumbai, India
| | - Shilpa Bhowmick
- Department of Biochemistry and Virology, Indian Council of Medical Research (ICMR)-National Institute for Research in Reproductive Health, Mumbai, India
| | - Srabani Mukherjee
- Department of Molecular Endocrinology, Indian Council of Medical Research (ICMR)-National Institute for Research in Reproductive Health, Mumbai, India
| | - Vidya Nagar
- Department of Medicine, Grant Medical College & Sir J. J. Group of Hospitals, Mumbai, India
| | - Priya Patil
- Department of Medicine, Grant Medical College & Sir J. J. Group of Hospitals, Mumbai, India
| | - Vainav Patel
- Department of Biochemistry and Virology, Indian Council of Medical Research (ICMR)-National Institute for Research in Reproductive Health, Mumbai, India
| |
Collapse
|
184
|
Santamaría-Cadavid M, Rodríguez-Castro E, Rodríguez-Yáñez M, Arias-Rivas S, López-Dequidt I, Pérez-Mato M, Rodríguez-Pérez M, López-Loureiro I, Hervella P, Campos F, Castillo J, Iglesias-Rey R, Sobrino T. Regulatory T cells participate in the recovery of ischemic stroke patients. BMC Neurol 2020; 20:68. [PMID: 32111174 PMCID: PMC7048127 DOI: 10.1186/s12883-020-01648-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 02/19/2020] [Indexed: 01/09/2023] Open
Abstract
Background Recent preclinical studies have shown that regulatory T cells (Treg) play a key role in the immune response after ischemic stroke (IS). However, the role of Treg in human acute IS has been poorly investigated. Our aim was to study the relationship between circulating Treg and outcome in human IS patients. Methods A total of 204 IS patients and 22 control subjects were recruited. The main study variable was good functional outcome at 3 months (modified Rankin scale ≤2) considering infarct volume, Early Neurological Deterioration (END) and risk of infections as secondary variables. The percentage of circulating Treg was measured at admission, 48, 72 h and at day 7 after stroke onset. Results Circulating Treg levels were higher in IS patients compared to control subjects. Treg at 48 h were independently associated with good functional outcome (OR, 3.5; CI: 1.9–7.8) after adjusting by confounding factors. Patients with lower Treg at 48 h showed higher frequency of END and risk of infections. In addition, a negative correlation was found between circulating Treg at 48 h (r = − 0.414) and 72 h (r = − 0.418) and infarct volume. Conclusions These findings suggest that Treg may participate in the recovery of IS patients. Therefore, Treg may be considered a potential therapeutic target in acute ischemic stroke.
Collapse
Affiliation(s)
- María Santamaría-Cadavid
- Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico, c/ Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - Emilio Rodríguez-Castro
- Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico, c/ Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - Manuel Rodríguez-Yáñez
- Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico, c/ Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - Susana Arias-Rivas
- Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico, c/ Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - Iria López-Dequidt
- Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico, c/ Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - María Pérez-Mato
- Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico, c/ Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - Manuel Rodríguez-Pérez
- Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico, c/ Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - Ignacio López-Loureiro
- Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico, c/ Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - Pablo Hervella
- Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico, c/ Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - Francisco Campos
- Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico, c/ Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - José Castillo
- Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico, c/ Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain
| | - Ramón Iglesias-Rey
- Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico, c/ Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain.
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico, c/ Travesa da Choupana, s/n, 15706, Santiago de Compostela, Spain.
| |
Collapse
|
185
|
Massalska M, Radzikowska A, Kuca-Warnawin E, Plebanczyk M, Prochorec-Sobieszek M, Skalska U, Kurowska W, Maldyk P, Kontny E, Gober HJ, Maslinski W. CD4 +FOXP3 + T Cells in Rheumatoid Arthritis Bone Marrow Are Partially Impaired. Cells 2020; 9:E549. [PMID: 32111105 PMCID: PMC7140449 DOI: 10.3390/cells9030549] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/13/2020] [Accepted: 02/20/2020] [Indexed: 12/12/2022] Open
Abstract
There is evolving evidence that dysregulation of immune homeostasis in the bone marrow (BM) adjacent to the inflamed joints is involved in the pathogenesis of. In this study, we are addressing the phenotype and function of regulatory T cells (Tregs) residing in the BM of patients with rheumatoid arthritis (RA) and osteoarthritis (OA). BM and peripheral blood samples were obtained from RA and OA patients undergoing hip replacement surgery. The number and phenotype of Tregs were analyzed by flow cytometry and immunohistochemistry. The function of Tregs was investigated ex vivo, addressing their suppressive activity on effector T cells. [3H]-Thymidine incorporation assay and specific enzyme-linked immunosorbent assay were used for quantification of cell proliferation and pro-inflammatory (TNF, IFN-γ) cytokine release, respectively. Significantly lower numbers of CD4+FOXP3+ T cells were found in the BM of patients with RA compared to control patients with OA. High expression of CD127 (IL-7 receptor) and relatively low expression of CXCR4 (receptor for stromal cell-derived factor CXCL12) are characteristics of the CD4+FOXP3+ cells residing in the BM of RA patients. The BM-resident Tregs of RA patients demonstrated a limited suppressive activity on the investigated immune response. Our results indicate that the reduced number and impaired functional properties of CD4+FOXP3+ T cells present in the BM of RA patients may favor the inflammatory process, which is observed in RA BM.
Collapse
Affiliation(s)
- Magdalena Massalska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology, and Rehabilitation, 02-637 Warsaw, Poland; (A.R.); (E.K.-W.); (M.P.); (U.S.); (W.K.); (E.K.); (W.M.)
| | - Anna Radzikowska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology, and Rehabilitation, 02-637 Warsaw, Poland; (A.R.); (E.K.-W.); (M.P.); (U.S.); (W.K.); (E.K.); (W.M.)
| | - Ewa Kuca-Warnawin
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology, and Rehabilitation, 02-637 Warsaw, Poland; (A.R.); (E.K.-W.); (M.P.); (U.S.); (W.K.); (E.K.); (W.M.)
| | - Magdalena Plebanczyk
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology, and Rehabilitation, 02-637 Warsaw, Poland; (A.R.); (E.K.-W.); (M.P.); (U.S.); (W.K.); (E.K.); (W.M.)
| | - Monika Prochorec-Sobieszek
- Department of Pathology, National Institute of Geriatrics, Rheumatology, and Rehabilitation, 02-637 Warsaw, Poland;
- Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, 02-776 Warsaw, Poland
| | - Urszula Skalska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology, and Rehabilitation, 02-637 Warsaw, Poland; (A.R.); (E.K.-W.); (M.P.); (U.S.); (W.K.); (E.K.); (W.M.)
| | - Weronika Kurowska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology, and Rehabilitation, 02-637 Warsaw, Poland; (A.R.); (E.K.-W.); (M.P.); (U.S.); (W.K.); (E.K.); (W.M.)
| | - Pawel Maldyk
- Department of Rheumoorthopaedic Surgery, National Institute of Geriatrics, Rheumatology, and Rehabilitation, 02-637 Warsaw, Poland;
- Clinical Department of Orthopedic and Traumatology of Locomotor System, Enfant-Jesus Clinical Hospital, 02-005 Warsaw, Poland
| | - Ewa Kontny
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology, and Rehabilitation, 02-637 Warsaw, Poland; (A.R.); (E.K.-W.); (M.P.); (U.S.); (W.K.); (E.K.); (W.M.)
| | - Hans-Jürgen Gober
- Department of Pharmacy, Kepler University Hospital, 4020 Linz, Austria;
- Pharmaceutical Outcomes Programme, British Columbia Children’s Hospital, Vancouver, BC V5Z 4H4, Canada
| | - Wlodzimierz Maslinski
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology, and Rehabilitation, 02-637 Warsaw, Poland; (A.R.); (E.K.-W.); (M.P.); (U.S.); (W.K.); (E.K.); (W.M.)
| |
Collapse
|
186
|
Atif M, Conti F, Gorochov G, Oo YH, Miyara M. Regulatory T cells in solid organ transplantation. Clin Transl Immunology 2020; 9:e01099. [PMID: 32104579 PMCID: PMC7036337 DOI: 10.1002/cti2.1099] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 12/02/2019] [Accepted: 12/04/2019] [Indexed: 12/19/2022] Open
Abstract
The induction of graft tolerance remains the holy grail of transplantation. This is important as chronic allograft dysfunction and the side effects of immunosuppression regimens place a major burden on the lives of transplant patients and their healthcare systems. This has mandated the need to understand the immunobiology of graft rejection and identify novel therapeutics. Regulatory T (Treg) cells play an important role in modulating pro-inflammatory microenvironments and maintaining tissue homeostasis. However, there are fundamental unanswered questions regarding Treg cell immunobiology. These cells are a heterogeneous entity with functionally diverse roles. Moreover, the adoption of novel deeper immunophenotyping and genomic sequencing technologies has identified this phenotype and function to be more complex than expected. Hence, a comprehensive understanding of Treg cell heterogeneity is needed to safely and effectively exploit their therapeutic potential. From a clinical perspective, the recent decade has seen different clinical teams commence and complete first-in-man clinical trials utilising Treg cells as an adoptive cellular therapy. In this review, we discuss these trials from a translational perspective with an important focus on safety. Finally, we identify crucial knowledge gaps for future study.
Collapse
Affiliation(s)
- Muhammad Atif
- Sorbonne UniversitéInserm U1135Centre d'Immunologie et des Maladies Infectieuses (CIMI‐Paris)Hôpital Pitié‐SalpêtrièreAP‐HPParisFrance
- Unité de Transplantation HépatiqueHôpital Pitié‐SalpêtrièreAP‐HPParisFrance
- Centre for Liver and Gastro ResearchNIHR Birmingham Biomedical Research CentreUniversity of BirminghamBirminghamUK
- Academic Department of SurgeryUniversity of BirminghamBirminghamUK
| | - Filomena Conti
- Unité de Transplantation HépatiqueHôpital Pitié‐SalpêtrièreAP‐HPParisFrance
| | - Guy Gorochov
- Sorbonne UniversitéInserm U1135Centre d'Immunologie et des Maladies Infectieuses (CIMI‐Paris)Hôpital Pitié‐SalpêtrièreAP‐HPParisFrance
| | - Ye Htun Oo
- Centre for Liver and Gastro ResearchNIHR Birmingham Biomedical Research CentreUniversity of BirminghamBirminghamUK
- Liver Transplant and HPB UnitQueen Elizabeth HospitalUniversity Hospital Birmingham NHS Foundation TrustBirminghamUK
| | - Makoto Miyara
- Sorbonne UniversitéInserm U1135Centre d'Immunologie et des Maladies Infectieuses (CIMI‐Paris)Hôpital Pitié‐SalpêtrièreAP‐HPParisFrance
| |
Collapse
|
187
|
Kim JH, Kim BS, Lee SK. Regulatory T Cells in Tumor Microenvironment and Approach for Anticancer Immunotherapy. Immune Netw 2020; 20:e4. [PMID: 32158592 PMCID: PMC7049587 DOI: 10.4110/in.2020.20.e4] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 01/30/2020] [Accepted: 02/02/2020] [Indexed: 12/21/2022] Open
Abstract
Tregs have a role in immunological tolerance and immune homeostasis by suppressing immune reactions, and its therapeutic potential is critical in autoimmune diseases and cancers. There have been multiple studies conducted on Tregs because of their roles in immune suppression and therapeutic potential. In tumor immunity, Tregs can promote the development and progression of tumors by preventing effective anti-tumor immune responses in tumor-bearing hosts. High infiltration of Tregs into tumor tissue results in poor survival in various types of cancer patients. Identifying factors specifically expressed in Tregs that affect the maintenance of stability and function of Tregs is important for understanding cancer pathogenesis and identifying therapeutic targets. Thus, manipulation of Tregs is a promising anticancer strategy, but finding markers for Treg-specific depletion and controlling these cells require fine-tuning and further research. Here, we discuss the role of Tregs in cancer and the development of Treg-targeted therapies to promote cancer immunotherapy.
Collapse
Affiliation(s)
- Jung-Ho Kim
- Research Institute for Precision Immune-Medicine, Good T Cells, Inc., Seoul 03722, Korea
| | - Beom Seok Kim
- Research Institute for Precision Immune-Medicine, Good T Cells, Inc., Seoul 03722, Korea
| | - Sang-Kyou Lee
- Research Institute for Precision Immune-Medicine, Good T Cells, Inc., Seoul 03722, Korea
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
188
|
Whangbo JS, Antin JH, Koreth J. The role of regulatory T cells in graft-versus-host disease management. Expert Rev Hematol 2020; 13:141-154. [PMID: 31874061 DOI: 10.1080/17474086.2020.1709436] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: Despite improvements in human leukocyte antigen (HLA) matching algorithms and supportive care, graft-versus-host disease (GVHD) remains the leading cause of non-relapse morbidity and mortality following allogeneic hematopoietic stem cell transplantation (HSCT). Acute GVHD, typically occurring in the first 100 days post-HSCT, is mediated by mature effector T cells from the donor (graft) that become activated after encountering alloantigens in the recipient (host). Chronic GVHD, characterized by aberrant immune responses to both autoantigens and alloantigens, occurs later and arises from a failure to develop tolerance after HSCT. CD4+ CD25+ CD127- FOXP3+ regulatory T cells (Tregs) function to suppress auto- and alloreactive immune responses and are key mediators of immune tolerance.Areas covered: In this review, authors discuss the biologic and therapeutic roles of Tregs in acute and chronic GVHD, including in vivo and ex vivo strategies for Treg expansion and adoptive Treg cellular therapy.Expert opinion: Although they comprise only a small subset of circulating CD4 + T cells, Tregs play an important role in establishing and maintaining immune tolerance following allogeneic HSCT. The development of GVHD has been associated with reduced Treg frequency or numbers. Consequently, the immunosuppressive properties of Tregs are being harnessed in clinical trials for GVHD prevention and treatment.
Collapse
Affiliation(s)
- Jennifer S Whangbo
- Division of Hematology-Oncology, Boston Children's Hospital, Boston, MA and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Joseph H Antin
- Harvard Medical School, Boston, MA, USA.,Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA, USA
| | - John Koreth
- Harvard Medical School, Boston, MA, USA.,Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
189
|
Battaglia A, Buzzonetti A, Fossati M, Scambia G, Fattorossi A, Madiyalakan MR, Mahnke YD, Nicodemus C. Translational immune correlates of indirect antibody immunization in a randomized phase II study using scheduled combination therapy with carboplatin/paclitaxel plus oregovomab in ovarian cancer patients. Cancer Immunol Immunother 2020; 69:383-397. [PMID: 31897661 DOI: 10.1007/s00262-019-02456-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 12/13/2019] [Indexed: 12/12/2022]
Abstract
The standard-of-care (SOC) first-line therapy for ovarian cancer (OC) patients is plagued with high relapse rates. Several studies indicated the immune system's prominent role changing the disease course in OC patients. Chemo-immunotherapy regimens, currently being explored, include oregovomab, which is a monoclonal antibody specific for the OC associated antigen carbohydrate/cancer antigen 125 (CA125) that yielded promising results when administered together with SOC in a previous study. The QPT-ORE-002 multi-site phase II randomized study demonstrated that in patients with advanced OC, oregovomab combined with first-line SOC improved overall and progression-free survival, compared to SOC alone. The study included an Italian cohort in which we demonstrated that adding oregovomab to SOC resulted in increased patient numbers with amplified CA125-specific CD8+T lymphocytes/ml peripheral blood counts, which might explain the improved therapeutic effect of SOC + oregovomab over SOC alone. Predictive for oregovomab efficacy was a less suppressive immune environment at baseline as indicated by low numbers of circulating myeloid-derived suppressor cells, subset type 4, and a low neutrophil-and-monocyte to lymphocyte ratio.
Collapse
Affiliation(s)
- Alessandra Battaglia
- Istituto di Clinica Ostetrica e Ginecologica, Università Cattolica del Sacro Cuore, L.go F.Vito 1, 00168, Rome, Italy.
| | - Alexia Buzzonetti
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Marco Fossati
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giovanni Scambia
- Istituto di Clinica Ostetrica e Ginecologica, Università Cattolica del Sacro Cuore, L.go F.Vito 1, 00168, Rome, Italy.,Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Andrea Fattorossi
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | | | | |
Collapse
|
190
|
Williams JH, Udata C, Ganguly BJ, Bucktrout SL, Joh T, Shannon M, Wong GY, Levisetti M, Garzone PD, Meng X. Model-Based Characterization of the Pharmacokinetics, Target Engagement Biomarkers, and Immunomodulatory Activity of PF-06342674, a Humanized mAb Against IL-7 Receptor-α, in Adults with Type 1 Diabetes. AAPS JOURNAL 2020; 22:23. [PMID: 31900603 PMCID: PMC6942017 DOI: 10.1208/s12248-019-0401-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/21/2019] [Indexed: 12/22/2022]
Abstract
IL-7 receptor-α (IL-7Rα) blockade has been shown to reverse autoimmune diabetes in the non-obese diabetic mouse by promoting inhibition of effector T cells and consequently altering the balance of regulatory T (Treg) and effector memory (TEM) cells. PF-06342674 is a humanized monoclonal antibody that binds to and inhibits the function of IL-7Rα. In the current phase 1b study, subjects with type 1 diabetes (T1D) received subcutaneous doses of either placebo or PF-06342674 (1, 3, 8 mg/kg/q2w or 6 mg/kg/q1w) for 10 weeks and were followed up to 18 weeks. Nonlinear mixed effects models were developed to characterize the pharmacokinetics (PK), target engagement biomarkers, and immunomodulatory activity. PF-06342674 was estimated to have 20-fold more potent inhibitory effect on TEM cells relative to Treg cells resulting in a non-monotonic dose-response relationship for the Treg:TEM ratio, reaching maximum at ~ 3 mg/kg/q2w dose. Target-mediated elimination led to nonlinear PK with accelerated clearance at lower doses due to high affinity binding and rapid clearance of the drug-target complex. Doses ≥ 3 mg/kg q2w result in sustained PF-06342674 concentrations higher than the concentration of cellular IL-7 receptor and, in turn, maintain near maximal receptor occupancy over the dosing interval. The results provide important insight into the mechanism of IL-7Rα blockade and immunomodulatory activity of PF-06342674 and establish a rational framework for dose selection for subsequent clinical trials of PF-06342674. Furthermore, this analysis serves as an example of mechanistic modeling to support dose selection of a drug candidate in the early phases of development.
Collapse
Affiliation(s)
- Jason H Williams
- Worldwide Research & Development, Pfizer Inc, 10777 Science Center Dr, CB1/1130, San Diego, California, 92121, USA.
| | - Chandrasekhar Udata
- Worldwide Research & Development, Pfizer Inc, 10777 Science Center Dr, CB1/1130, San Diego, California, 92121, USA
| | - Bishu J Ganguly
- Pfizer Inc, South San Francisco, California, USA.,Lyell Immunopharma, South San Francisco, California, USA
| | - Samantha L Bucktrout
- Pfizer Inc, South San Francisco, California, USA.,Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Tenshang Joh
- Worldwide Research & Development, Pfizer Inc, 10777 Science Center Dr, CB1/1130, San Diego, California, 92121, USA
| | - Megan Shannon
- Worldwide Research & Development, Pfizer Inc, 10777 Science Center Dr, CB1/1130, San Diego, California, 92121, USA
| | - Gilbert Y Wong
- Worldwide Research & Development, Pfizer Inc, 10777 Science Center Dr, CB1/1130, San Diego, California, 92121, USA
| | - Matteo Levisetti
- Worldwide Research & Development, Pfizer Inc, 10777 Science Center Dr, CB1/1130, San Diego, California, 92121, USA.,DNAtrix Therapeutics, San Diego, California, USA
| | - Pamela D Garzone
- Pfizer Inc, South San Francisco, California, USA.,Calibr, a division of Scripps Research, La Jolla, California, USA
| | - Xu Meng
- Worldwide Research & Development, Pfizer Inc, 10777 Science Center Dr, CB1/1130, San Diego, California, 92121, USA
| |
Collapse
|
191
|
Abstract
The transcription factor FOXP3 controls the immunosuppressive program in CD4+ T cells that is crucial for systemic immune regulation. Mutations of the single X-chromosomal FOXP3 gene in male individuals cause the inherited autoimmune disease immune dysregulation, polyendocrinopathy, enteropathy, and X-linked (IPEX) syndrome. Insufficient gene expression and impaired function of mutant FOXP3 protein prevent the generation of anti-inflammatory regulatory T (Treg) cells and fail to inhibit autoreactive T cell responses. Diversification of FOXP3 functional properties is achieved through alternative splicing that leads to isoforms lacking exon 2 (FOXP3Δ2), exon 7 (FOXP3Δ7), or both (FOXP3Δ2Δ7) specifically in human CD4+ T cells. Several IPEX mutations targeting these exons or promoting their alternative splicing revealed that those truncated isoforms cannot compensate for the loss of the full-length isoform (FOXP3fl). In this review, IPEX mutations that change the FOXP3 isoform profile and the resulting consequences for the CD4+ T-cell phenotype are discussed.
Collapse
Affiliation(s)
- Reiner K Mailer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
192
|
Mai HL, Nguyen TVH, Branchereau J, Poirier N, Renaudin K, Mary C, Belarif L, Minault D, Hervouet J, Le Bas-Berdardet S, Soulillou JP, Vanhove B, Blancho G, Brouard S. Interleukin-7 receptor blockade by an anti-CD127 monoclonal antibody in nonhuman primate kidney transplantation. Am J Transplant 2020; 20:101-111. [PMID: 31344323 DOI: 10.1111/ajt.15543] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/28/2019] [Accepted: 07/19/2019] [Indexed: 01/25/2023]
Abstract
IL-7 is an important cytokine for T cell lymphopoiesis. Blockade of the IL-7 signaling pathway has been shown to induce long-term graft survival or graft tolerance in murine transplant models through inhibiting T cell homeostasis and favoring immunoregulation. In this study, we assessed for the first time the effects of a blocking anti-human cluster of differentiation 127 (CD127) mAb administered in combination with low-dose tacrolimus or thymoglobulin in a life-sustaining kidney allograft model in baboons. Contrary to our expectation, the addition of an anti-CD127 mAb to the treatment protocols did not prolong graft survival compared to low-dose tacrolimus alone or thymoglobulin alone. Anti-CD127 mAb administration led to full CD127 receptor occupancy during the follow-up period. However, all treated animals lost their kidney graft between 1 week and 2 weeks after transplantation. Unlike in rodents, in nonhuman primates, anti-CD127 mAb treatment does not decrease the absolute numbers of lymphocyte and lymphocyte subsets and does not effectively inhibit postdepletional T cell proliferation and homeostasis, suggesting that IL-7 is not a limiting factor for T cell homeostasis in primates.
Collapse
Affiliation(s)
- Hoa Le Mai
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Thi Van Ha Nguyen
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Julien Branchereau
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,Service d'Urologie, CHU Nantes, France
| | | | - Karine Renaudin
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,Service d'Anatomie et Cytologie Pathologiques, CHU Nantes, France
| | | | | | - David Minault
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Jeremy Hervouet
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Stéphanie Le Bas-Berdardet
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Jean-Paul Soulillou
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | | | - Gilles Blancho
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Sophie Brouard
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| |
Collapse
|
193
|
Ruiter B, Smith NP, Monian B, Tu AA, Fleming E, Virkud YV, Patil SU, Whittaker CA, Love JC, Shreffler WG. Expansion of the CD4 + effector T-cell repertoire characterizes peanut-allergic patients with heightened clinical sensitivity. J Allergy Clin Immunol 2020; 145:270-282. [PMID: 31654649 PMCID: PMC6949413 DOI: 10.1016/j.jaci.2019.09.033] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 08/06/2019] [Accepted: 09/06/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Individuals with peanut allergy range in clinical sensitivity: some can consume grams of peanut before experiencing any symptoms, whereas others suffer systemic reactions to 10 mg or less. Current diagnostic testing only partially predicts this clinical heterogeneity. OBJECTIVE We sought to identify characteristics of the peanut-specific CD4+ T-cell response in peanut-allergic patients that correlate with high clinical sensitivity. METHODS We studied the T-cell receptor β-chain (TCRβ) usage and phenotypes of peanut-activated, CD154+ CD4+ memory T cells using fluorescence-activated cell sorting, TCRβ sequencing, and RNA-Seq, in reactive and hyporeactive patients who were stratified by clinical sensitivity. RESULTS TCRβ analysis of the CD154+ and CD154- fractions revealed more than 6000 complementarity determining region 3 sequences and motifs that were significantly enriched in the activated cells and 17% of the sequences were shared between peanut-allergic individuals, suggesting strong convergent selection of peanut-specific clones. These clones were more numerous among the reactive patients, and this expansion was identified within effector, but not regulatory T-cell populations. The transcriptional profile of CD154+ T cells in the reactive group skewed toward a polarized TH2 effector phenotype, and expression of TH2 cytokines strongly correlated with peanut-specific IgE levels. There were, however, also non-TH2-related differences in phenotype. Furthermore, the ratio of peanut-specific clones in the effector versus regulatory T-cell compartment, which distinguished the clinical groups, was independent of specific IgE concentration. CONCLUSIONS Expansion of the peanut-specific effector T-cell repertoire is correlated with clinical sensitivity, and this observation may be useful to inform our assessment of disease phenotype and to monitor disease longitudinally.
Collapse
Affiliation(s)
- Bert Ruiter
- Center for Immunology & Inflammatory Diseases, Massachusetts General Hospital, Boston, Mass; Harvard Medical School, Boston, Mass.
| | - Neal P Smith
- Center for Immunology & Inflammatory Diseases, Massachusetts General Hospital, Boston, Mass
| | - Brinda Monian
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Mass
| | - Ang A Tu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Mass
| | - Elizabeth Fleming
- Center for Immunology & Inflammatory Diseases, Massachusetts General Hospital, Boston, Mass
| | - Yamini V Virkud
- Center for Immunology & Inflammatory Diseases, Massachusetts General Hospital, Boston, Mass; Harvard Medical School, Boston, Mass; Food Allergy Center, Massachusetts General Hospital, Boston, Mass
| | - Sarita U Patil
- Center for Immunology & Inflammatory Diseases, Massachusetts General Hospital, Boston, Mass; Harvard Medical School, Boston, Mass; Food Allergy Center, Massachusetts General Hospital, Boston, Mass
| | - Charles A Whittaker
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Mass; The Barbara K. Ostrom (1978) Bioinformatics and Computing Facility in the Swanson Biotechnology Center, Massachusetts Institute of Technology, Cambridge, Mass
| | - J Christopher Love
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Mass
| | - Wayne G Shreffler
- Center for Immunology & Inflammatory Diseases, Massachusetts General Hospital, Boston, Mass; Harvard Medical School, Boston, Mass; Food Allergy Center, Massachusetts General Hospital, Boston, Mass
| |
Collapse
|
194
|
Fu J, Duan J, Mo J, Xiao H, Huang Y, Chen W, Xiang S, Yang F, Chen Y, Xu S. Mild Cognitive Impairment Patients Have Higher Regulatory T-Cell Proportions Compared With Alzheimer's Disease-Related Dementia Patients. Front Aging Neurosci 2020; 12:624304. [PMID: 33551792 PMCID: PMC7862571 DOI: 10.3389/fnagi.2020.624304] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/21/2020] [Indexed: 02/05/2023] Open
Abstract
Objectives: The role of neuroinflammation in the pathogenesis of Alzheimer's disease (AD) has attracted much attention recently. Regulatory T-cells (Tregs) play an important role in modulating inflammation. We aimed to explore the Treg-related immunosuppression status at different stages of AD. Methods: Thirty healthy control (HC) subjects, 26 patients with mild cognitive impairment (MCI), 30 patients with mild probable AD-related dementia, and 28 patients with moderate-to-severe probable AD-related dementia underwent detailed clinical history taking, structural MRI scanning, and neuropsychological assessment. Peripheral blood samples were taken to measure the percentage of CD4+CD25+CD127low/- Tregs by flow cytometry and the levels of interleukin (IL-10), interleukin (IL-35), and transforming growth factor β (TGF-β) by ELISA. Results: The percentage of Tregs in the blood of MCI patients was the highest (9.24%); there was a significant difference between patients with MCI and patients with probable AD-related dementia. The level of TGF-β in patients with MCI (47.02 ng/ml) was significantly increased compared with patients with AD-related dementia. There were positive correlations between Treg percentage, IL-35, and Mini-mental state evaluation scores in patients with MCI and probable AD-related dementia. Conclusions: Patients with MCI have stronger Treg-related immunosuppression status compared with patients with probable AD-related dementia.
Collapse
Affiliation(s)
- Jiping Fu
- Eastern Department of Neurology, Guangdong Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Department of Neurology, First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Jinhai Duan
- Eastern Department of Neurology, Guangdong Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jianwei Mo
- Eastern Department of Neurology, Guangdong Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hao Xiao
- Eastern Department of Neurology, Guangdong Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yuedong Huang
- Eastern Department of Neurology, Guangdong Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Weiping Chen
- Eastern Department of Neurology, Guangdong Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shaotong Xiang
- Eastern Department of Neurology, Guangdong Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Fan Yang
- Eastern Department of Neurology, Guangdong Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yongjun Chen
- Eastern Department of Neurology, Guangdong Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shuwen Xu
- Eastern Department of Neurology, Guangdong Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- *Correspondence: Shuwen Xu
| |
Collapse
|
195
|
Tawfeik AM, Mora A, Osman A, Moneer MM, El-Sheikh N, Elrefaei M. Frequency of CD4+ regulatory T cells, CD8+ T cells, and human papilloma virus infection in Egyptian Women with breast cancer. Int J Immunopathol Pharmacol 2020; 34:2058738420966822. [PMID: 33103515 PMCID: PMC7786412 DOI: 10.1177/2058738420966822] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 09/22/2020] [Indexed: 12/11/2022] Open
Abstract
Several subsets of regulatory CD4+ T cells (CD4+ Tregs) have been described in peripheral blood and tumor microenvironment of breast cancer (BC) patients and may play a role in the progression of BC. High-risk human papilloma virus (HR-HPV) has a causal role in cervical, head, and neck tumors but the role of HR-HPV in evoking neoplasia in BC is still unclear. In this study we assessed the prevalence of CD4+CD25+ FOXP3+ regulatory T cells (CD4+Tregs) and CD3+ CD8+ T cells by flow cytometry in peripheral blood from a total of 55 Egyptian women, including 20 treatment-naïve BC, 15 with breast benign lesions (BBL), and 20 healthy volunteers (HV). HR-HPV genotypes type 16, 18, and 31 were investigated in breast tissue from all BC and BBL patients using Real-Time PCR. HR-HPV was detected in 4/20 (20%) and 0/15 (0%) BC and BBL patients respectively. The frequency of CD4+ Tregs was significantly higher in BC compared to BBL and HV, (P < 0.001). In addition, we observed a significantly higher frequency of CD3+ CD8+ T cells in peripheral blood of patients with late stage III BC compared to early stage I and II BC (P = 0.011). However, there was no significant association between the ratio of CD8+ T cell to CD4+ Tregs frequencies and the expression of Estrogen Receptor (ER), Progesterone Receptor (PR), and Human Epidermal Growth Factor Receptor 2 (HER2). These results lead us to postulate that the association between the frequency of CD4+ Tregs and CD8+ T cells in the peripheral blood may be a prognostic or predictive parameter in Egyptian women with BC. In addition, HR-HPV infection may be implicated in the development of some types of BC in Egyptian women.
Collapse
Affiliation(s)
- Amany M Tawfeik
- Molecular Immunology Unit for Infectious Diseases, Department of Microbiology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Ahmed Mora
- Department of Chemistry, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Ahmed Osman
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
- Biotechnology Program, Basic and Applied Sciences Institute, Egypt-Japan University of Science and Technology, Cairo, Egypt
| | - Manar M Moneer
- Department of Epidemiology and Statistics, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Nabila El-Sheikh
- Molecular Immunology Unit for Infectious Diseases, Department of Microbiology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Mohamed Elrefaei
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
196
|
Zhou W, Deng J, Chen Q, Li R, Xu X, Guan Y, Li W, Xiong X, Li H, Li J, Cai X. Expression of CD4+CD25+CD127 Low regulatory T cells and cytokines in peripheral blood of patients with primary liver carcinoma. Int J Med Sci 2020; 17:712-719. [PMID: 32218692 PMCID: PMC7085268 DOI: 10.7150/ijms.44088] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 02/16/2020] [Indexed: 12/26/2022] Open
Abstract
Objective: To assess the clinical utility of the ratio of CD4+CD25+CD127low regulatory T cells (Tregs) in subjects at high risk of HCC, investigate the relationship between the percentage of Tregs and the expression of transforming growth factor (TGF)-β1 and interleukin (IL)-10 in patients with hepatocellular carcinoma before and after treatment. Methods: Peripheral venous blood was collected from patients with liver cancer before and after treatment. The proportion of CD4+CD25+CD127low Tregs was detected by flow cytometry. The levels of TGF-β1 and IL-10 in serum were detected by enzyme-linked immunosorbent assay, and were compared with healthy subjects as a control group. Results: The proportion of CD4+CD25+CD127low to CD4+T lymphocytes in patients with hepatocellular carcinoma was significantly higher than that in healthy controls (P<0.01). The proportion of CD4+CD25+CD127lowTregs, whose AUC of ROC curve was 0.917, could effectively separate the HCC patients from the healthy subjects with a diagnostic sensitivity of 90%, specificity of 80%. The proportion of CD4+CD25+CD127low to CD4+T lymphocytes and the levels of TGF-β1 and IL-10 in patients with hepatocellular carcinoma after the operation and chemotherapy were significantly lower than those before treatment (P<0.05).The proportion of CD4+CD25+CD127lowTregs was positively correlated with the concentrations of TGF-β1 and IL-10 before and after treatment of primary liver cancer (P<0.05). Conclusion: CD4+CD25+CD127lowTregs may be a significant predictor of HCC biopsy outcome and play an inhibitory role on effector T cells by regulating cytokines.
Collapse
Affiliation(s)
- Wenchao Zhou
- Clinical laboratory, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Jianxin Deng
- Department of Endocrinology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen 518035, People's Republic of China
| | - Qianmei Chen
- Clinical laboratory, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Ruiying Li
- Clinical laboratory, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Xiaosong Xu
- Clinical laboratory, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Yubin Guan
- Clinical laboratory, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Wei Li
- Clinical laboratory, Guangzhou Military Area Inspection Center, the General Hospital of Guangzhou Military Region, Guangzhou 510010, China
| | - Xiaomin Xiong
- Clinical laboratory, the Hospital of Dongguan Renkang, Dongguan 523952, China
| | - Hongwei Li
- Institute of Biotherapy, Southern Medical University Guangzhou 510515, China
| | - Jianpei Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Clinical Laboratory Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
- ✉ Corresponding authors: Jianpei Li, or Xiangsheng Cai,
| | - Xiangsheng Cai
- Clinical laboratory, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- ✉ Corresponding authors: Jianpei Li, or Xiangsheng Cai,
| |
Collapse
|
197
|
Scheinecker C, Göschl L, Bonelli M. Treg cells in health and autoimmune diseases: New insights from single cell analysis. J Autoimmun 2019; 110:102376. [PMID: 31862128 DOI: 10.1016/j.jaut.2019.102376] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 11/22/2019] [Indexed: 02/07/2023]
Abstract
Autoimmune diseases, such as Systemic Lupus Erythematosus (SLE) or Rheumatoid Arthritis (RA) are characterized by the breakdown of immunological tolerance. Defects of regulatory T cells have been described among the various mechanisms, that are important for the development of autoimmune diseases, due to their critical role as regulators of peripheral immune tolerance and homeostasis. Initially T suppressor cells have been described as one population of peripheral T cells. Based on new technological advances a new understanding of the heterogeneity of different Treg cell populations in the lymphoid and non-lymphoid tissue has evolved over the last years. While initially Foxp3 has been defined as the main master regulator of Treg cells, we have learned that Treg cells from various tissue can be identified by a specific transcriptomic and epigenetic signature. Epigenetic mechanisms allow Treg cell stability, but we have also learned that certain Treg subsets are plastic and can under specific circumstances even enhance autoimmunity and inflammatory processes. Quantitative and functional defects of Treg cells have been observed in a variety of autoimmune diseases. Due to our understanding of the nature of this cell population, Treg cells have been a target of new Treg based therapies, such as low-dose IL-2. In addition, ongoing clinical trials aim to test safety and efficacy of transferred, in vitro expanded Treg cells in patients with autoimmune diseases and transplant patients.
Collapse
Affiliation(s)
- Clemens Scheinecker
- Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria.
| | - Lisa Göschl
- Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria.
| | - Michael Bonelli
- Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
198
|
Zelm MC, McKenzie CI, Varese N, Rolland JM, O'Hehir RE. Recent developments and highlights in immune monitoring of allergen immunotherapy. Allergy 2019; 74:2342-2354. [PMID: 31587309 DOI: 10.1111/all.14078] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 10/01/2019] [Accepted: 10/02/2019] [Indexed: 12/15/2022]
Abstract
Allergic diseases are the most common chronic immune-mediated disorders and can manifest with an enormous diversity in clinical severity and symptoms. Underlying mechanisms for the adverse immune response to allergens and its downregulation by treatment are still being revealed. As a result, there have been, and still are, major challenges in diagnosis, prediction of disease progression/evolution and treatment. Currently, the only corrective treatment available is allergen immunotherapy (AIT). AIT modifies the immune response through long-term repeated exposure to defined doses of allergen. However, as the treatment usually needs to be continued for several years to be effective, and can be accompanied by adverse reactions, many patients face difficulties completing their schedule. Long-term therapy also potentially incurs high costs. Therefore, there is a great need for objective markers to predict or to monitor individual patient's beneficial changes in immune response during therapy so that efficacy can be identified as early as possible. In this review, we specifically address recent technical developments that have generated new insights into allergic disease pathogenesis, and how these could potentially be translated into routine laboratory assays for disease monitoring during AIT that are relatively inexpensive, robust and scalable.
Collapse
Affiliation(s)
- Menno C. Zelm
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Department of Respiratory Medicine Allergy and Clinical Immunology (Research) Central Clinical School Monash University, and Alfred Hospital Melbourne VIC Australia
| | - Craig I. McKenzie
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
| | - Nirupama Varese
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Department of Respiratory Medicine Allergy and Clinical Immunology (Research) Central Clinical School Monash University, and Alfred Hospital Melbourne VIC Australia
| | - Jennifer M. Rolland
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Department of Respiratory Medicine Allergy and Clinical Immunology (Research) Central Clinical School Monash University, and Alfred Hospital Melbourne VIC Australia
| | - Robyn E. O'Hehir
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Department of Respiratory Medicine Allergy and Clinical Immunology (Research) Central Clinical School Monash University, and Alfred Hospital Melbourne VIC Australia
| |
Collapse
|
199
|
Yu Y, Kiran Kumar MN, Wu MX. Delivery of allergen powder for safe and effective epicutaneous immunotherapy. J Allergy Clin Immunol 2019; 145:597-609. [PMID: 31783055 DOI: 10.1016/j.jaci.2019.11.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 11/02/2019] [Accepted: 11/07/2019] [Indexed: 01/16/2023]
Abstract
BACKGROUND More effective and safer immunotherapies to manage peanut allergy are in great demand despite extensive investigation of sublingual/oral immunotherapy and epicutaneous immunotherapy (EPIT) currently in the clinics. OBJECTIVE We sought to develop a powder-laden, dissolvable microneedle array (PLD-MNA) for epidermal delivery of powdered allergens and to evaluate the efficacy of this novel EPIT in peanut-sensitized mice. METHODS PLD-MNA was packaged with a mixture of powdered peanut allergen (PNA), 1,25-dihydroxyvitamin D3 (VD3), and CpG. Its epidermal delivery and therapeutic efficacy were evaluated alongside PNA-specific forkhead box P3-positive regulatory T cells and IL-10+ and TGF-β1+ skin-resident macrophages. RESULTS PLD-MNA was successfully laden with PNA/VD3/CpG powder and capable of epidermal delivery of most of its content 1 hour after application onto intact mouse skin concomitant with no significant leakage into the circulation or skin irritation. PLD-MNA-mediated EPIT substantially reduced clinical allergy scores to 1 from 3.5 in sham control mice (P < .001) after 6 treatments accompanied by lower levels of PNA-specific IgE and intestinal mucosal mast cells and eosinophils over sham treatments. Moreover, in comparison with allergens administered intradermally, powdered allergens delivered by means of PLD-MNA preferentially attracted immunoregulatory macrophages and stimulated the cells to produce IL-10, TGF-β, or both at the immunization site, which might account for increased numbers of regulatory T-like cells in lymph tissues in association with systemic tolerance. PNA/VD3/CpG-laden PLD-MNA was safe and required only 6 treatments and one fifth of the PNA adjuvant dose, with improved outcomes when compared with 12 conventional intradermal immunotherapies. CONCLUSIONS PLD-MNA holds great promise as a novel, safe, effective, and self-applicable modality to manage IgE-mediated allergies.
Collapse
Affiliation(s)
- Yang Yu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School
| | | | - Mei X Wu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School.
| |
Collapse
|
200
|
Cabral-Marques O, Schimke LF, de Oliveira EB, El Khawanky N, Ramos RN, Al-Ramadi BK, Segundo GRS, Ochs HD, Condino-Neto A. Flow Cytometry Contributions for the Diagnosis and Immunopathological Characterization of Primary Immunodeficiency Diseases With Immune Dysregulation. Front Immunol 2019; 10:2742. [PMID: 31849949 PMCID: PMC6889851 DOI: 10.3389/fimmu.2019.02742] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/08/2019] [Indexed: 12/24/2022] Open
Abstract
Almost 70 years after establishing the concept of primary immunodeficiency disorders (PIDs), more than 320 monogenic inborn errors of immunity have been identified thanks to the remarkable contribution of high-throughput genetic screening in the last decade. Approximately 40 of these PIDs present with autoimmune or auto-inflammatory symptoms as the primary clinical manifestation instead of infections. These PIDs are now recognized as diseases of immune dysregulation. Loss-of function mutations in genes such as FOXP3, CD25, LRBA, IL-10, IL10RA, and IL10RB, as well as heterozygous gain-of-function mutations in JAK1 and STAT3 have been reported as causative of these disorders. Identifying these syndromes has considerably contributed to expanding our knowledge on the mechanisms of immune regulation and tolerance. Although whole exome and whole genome sequencing have been extremely useful in identifying novel causative genes underlying new phenotypes, these approaches are time-consuming and expensive. Patients with monogenic syndromes associated with autoimmunity require faster diagnostic tools to delineate therapeutic strategies and avoid organ damage. Since these PIDs present with severe life-threatening phenotypes, the need for a precise diagnosis in order to initiate appropriate patient management is necessary. More traditional approaches such as flow cytometry are therefore a valid option. Here, we review the application of flow cytometry and discuss the relevance of this powerful technique in diagnosing patients with PIDs presenting with immune dysregulation. In addition, flow cytometry represents a fast, robust, and sensitive approach that efficiently uncovers new immunopathological mechanisms underlying monogenic PIDs.
Collapse
Affiliation(s)
- Otavio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Lena F Schimke
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, University of Freiburg, Freiburg im Breisgau, Germany
| | | | - Nadia El Khawanky
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Freiburg im Breisgau, Germany.,Precision Medicine Theme, The South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Rodrigo Nalio Ramos
- INSERM U932, SiRIC Translational Immunotherapy Team, Institut Curie, Paris Sciences et Lettres Research University, Paris, France
| | - Basel K Al-Ramadi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | | | - Hans D Ochs
- Department of Pediatrics, University of Washington School of Medicine, and Seattle Children's Research Institute, Seattle, WA, United States
| | - Antonio Condino-Neto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|