1
|
Nguyen AA, Platt CD. Flow Cytometry-based Immune Phenotyping of T and B Lymphocytes in the Evaluation of Immunodeficiency and Immune Dysregulation. Clin Lab Med 2024; 44:479-493. [PMID: 39089753 DOI: 10.1016/j.cll.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
There are approximately 500 congenital disorders that impair immune cell development and/or function. Patients with these disorders may present with a wide range of symptoms, including increased susceptibility to infection, autoimmunity, autoinflammation, lymphoproliferation, and/or atopy. Flow cytometry-based immune phenotyping of T and B lymphocytes plays an essential role in the evaluation of patients with these presentations. In this review, we describe the clinical utility of flow cytometry as part of a comprehensive evaluation of immune function and how this testing may be used as a diagnostic tool to identify underlying aberrant immune pathways, monitor disease activity, and assess infection risk.
Collapse
Affiliation(s)
- Alan A Nguyen
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Fegan Building 6th Floor, Boston, MA 02115, USA
| | - Craig D Platt
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, 1 Blackfan Circle, Karp Building 10th Floor, Boston, MA 02115, USA.
| |
Collapse
|
2
|
Guadalupe VOM, Abigail J SS, Patricia OR, Andrea HSD, Selma SM, Marco A YN, Carmen ZHM, Sara E EP, Berrón-Ruiz L. Analysis of B cell proliferation in response to in vitro stimulation in patients with CVID. Allergol Immunopathol (Madr) 2024; 52:94-102. [PMID: 39278858 DOI: 10.15586/aei.v52i5.1106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/29/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND Common variable immunodeficiency (CVID) is a heterogeneous disorder characterized by defective antibody production and impaired differentiation of B cells. B cell proliferation is an essential step for antibody synthesis. Depending on the nature of the stimulus, their response may be either T-cell-dependent or T-cell-independent. METHODS We studied 23 CVID patients and 14 healthy donors (HD). The patients were categorized based on their percentage of memory B cells. In addition to standard immunophenotyping of circulating human B and T cell subsets, an in vitro CFSE dilution assay was used to assess the proliferative capacity of B cells and to compare the activation of the T cell-dependent and T cell-independent response among the patients. RESULTS Patients with a reduction in memory B cells exhibited an increase in follicular T cells (Tfh) and showed low proliferation in response to PKW, CpG, and SAC stimuli (Condition II) (p= 0.0073). In contrast, patients with a normal percentage of memory B cells showed a high expression of IL-21R and low proliferation in response to CPG (Condition III); IL-21, CD40L, and anti-IgM (Condition IV) stimuli (p= 0.0163 and p = 0.0475, respectively). CONCLUSION Defective proliferation in patients depends on the type of stimulus used and the phenotypic characteristics of the patients. Further studies are necessary to understand the disease mechanisms, which may guide us toward identifying genetic defects associated with CVID.
Collapse
Affiliation(s)
| | - Saldaña-Solano Abigail J
- Departamento de Atención de la Salud, Universidad Autónoma Metropolitana, Unidad Xochimilco, Ciudad de México, México
| | | | | | - Scheffer-Mendoza Selma
- Servicio de Inmunología y Alergia, Instituto Nacional de Pediatría SSA, Ciudad de México, México
| | | | | | - Espinosa-Padilla Sara E
- Laboratorio en Inmunodeficiencias, Instituto Nacional de Pediatría SSA, Ciudad de México, México
| | - Laura Berrón-Ruiz
- Laboratorio en Inmunodeficiencias, Instituto Nacional de Pediatría SSA, Ciudad de México, México;
| |
Collapse
|
3
|
Sepahi A, Ho HE, Vyas P, Umiker B, Kis-Toth K, Wiederschain D, Radigan L, Cunningham-Rundles C. ICOS agonist vopratelimab modulates follicular helper T cells and improves B cell function in common variable immunodeficiency. Clin Immunol 2024; 264:110217. [PMID: 38621471 DOI: 10.1016/j.clim.2024.110217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 03/15/2024] [Accepted: 04/09/2024] [Indexed: 04/17/2024]
Abstract
Common variable immunodeficiency (CVID) is an immune defect characterized by hypogammaglobulinemia and impaired development of B cells into plasma cells. As follicular helper T cells (TFH) play a central role in humoral immunity, we examined TFH cells in CVID, and investigated whether an inducible T cell co-stimulator (ICOS) agonist, vopratelimab, could modulate TFH, B cell interactions and enhance immunoglobulin production. CVID subjects had decreased TFH17 and increased TFH1 subsets; this was associated with increased transitional B cells and decreased IgG+ B and IgD-IgM-CD27+ memory B cells. ICOS expression on CVID CD4+ T cells was also decreased. However, ICOS activation of CD4+ T cells by vopratelimab significantly increased total CVID TFH, TFH2, cell numbers, as well as IL-4, IL-10 and IL-21 secretion in vitro. Vopratelimab treatment also increased plasma cells, IgG+ B cells, reduced naïve & transitional B cells and significantly increased IgG1 secretion by CVID B cells. Interestingly, vopratelimab treatment also restored IgA secretion in PBMCs from several CVID patients who had a complete lack of endogenous serum IgA. Our data demonstrate the potential of TFH modulation in restoring TFH and enhancing B cell maturation in CVID. The effects of an ICOS agonist in antibody defects warrants further investigation. This biologic may also be of therapeutic interest in other clinical settings of antibody deficiency.
Collapse
Affiliation(s)
- Ali Sepahi
- PharmaEssentia Innovation Research Center, Bedford, MA, United States; Concentra Biosciences, LLC, Cambridge, MA, United States
| | - Hsi-En Ho
- Department of Medicine, Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Prapti Vyas
- ReNAgade Therapeutics, Cambridge, MA, United States; Concentra Biosciences, LLC, Cambridge, MA, United States
| | - Benjamin Umiker
- AstraZeneca, Cambridge, MA, United States; Concentra Biosciences, LLC, Cambridge, MA, United States
| | - Katalin Kis-Toth
- NextPoint Therapeutics, Inc., Cambridge, MA, United States; Concentra Biosciences, LLC, Cambridge, MA, United States
| | - Dmitri Wiederschain
- Crossbow Therapeutics, Cambridge, MA, United States; Concentra Biosciences, LLC, Cambridge, MA, United States
| | - Lin Radigan
- Department of Medicine, Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Charlotte Cunningham-Rundles
- Department of Medicine, Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
4
|
Satoh-Kanda Y, Nakayamada S, Kubo S, Yamagata K, Nawata A, Tanaka H, Kosaka S, Kanda R, Yu S, Fujita Y, Sonomoto K, Tanaka Y. Modifying T cell phenotypes using TYK2 inhibitor and its implications for the treatment of systemic lupus erythematosus. RMD Open 2024; 10:e003991. [PMID: 38871479 PMCID: PMC11177773 DOI: 10.1136/rmdopen-2023-003991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 05/31/2024] [Indexed: 06/15/2024] Open
Abstract
OBJECTIVES The tuning effects of JAK/TYK2 inhibitors on the imbalance between T follicular helper (Tfh) and T regulatory (Treg) cells, related to systemic lupus erythematosus (SLE) pathogenesis, were investigated using human peripheral blood samples. METHODS Peripheral blood mononuclear cells from untreated patients with SLE and healthy controls were analysed. Tfh1 cells were identified in nephritis tissue, and the effect of Tfh1 cells on B-cell differentiation was examined by coculturing naïve B cells with Tfh1 cells. RESULTS Tfh1 cell numbers were increased in the peripheral blood of patients, and activated Treg cell counts were decreased relative to Tfh1 cell counts. This imbalance in the Tfh to Treg ratio was remarkably pronounced in cases of lupus nephritis, especially in types III and IV active nephritis. Immunohistochemistry revealed Tfh1 cell infiltration in lupus nephritis tissues. Co-culture of Tfh1 cells (isolated from healthy individuals) with naïve B cells elicited greater induction of T-bet+ B cells than controls. In JAK/TYK2-dependent STAT phosphorylation assays using memory CD4+ T cells, IL-12-induced STAT1/4 phosphorylation and Tfh1 cell differentiation were inhibited by both JAK and TYK2 inhibitors. However, phosphorylation of STAT5 by IL-2 and induction of Treg cell differentiation by IL-2+TGFβ were inhibited by JAK inhibitors but not by TYK2 inhibitors, suggesting that TYK2 does not mediate the IL-2 signalling pathway. CONCLUSIONS Tfh1 cells can induce T-bet+ B cell production and may contribute to SLE pathogenesis-associated processes. TYK2 inhibitor may fine-tune the immune imbalance by suppressing Tfh1 differentiation and maintaining Treg cell differentiation, thereby preserving IL-2 signalling, unlike other JAK inhibitors.
Collapse
Affiliation(s)
- Yurie Satoh-Kanda
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | - Shingo Nakayamada
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | - Satoshi Kubo
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
- Department of Molecular Targeted Therapies (DMTT), University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | - Kaoru Yamagata
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | - Aya Nawata
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
- Department of Pathology and Oncology, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Hiroaki Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | - Shunpei Kosaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
- Department of Pathology and Oncology, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Ryuichiro Kanda
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | - Shan Yu
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
- Department of Pediatrics, Shenyang Women's and Children's Hospital, Shenyang, Liaoning, China
| | - Yuya Fujita
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | - Koshiro Sonomoto
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| |
Collapse
|
5
|
Surendar J, Hackenberg RK, Schmitt-Sánchez F, Ossendorff R, Welle K, Stoffel-Wagner B, Sage PT, Burger C, Wirtz DC, Strauss AC, Schildberg FA. Osteomyelitis is associated with increased anti-inflammatory response and immune exhaustion. Front Immunol 2024; 15:1396592. [PMID: 38736874 PMCID: PMC11082283 DOI: 10.3389/fimmu.2024.1396592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/12/2024] [Indexed: 05/14/2024] Open
Abstract
Introduction Osteomyelitis (OMS) is a bone infection causing bone pain and severe complications. A balanced immune response is critical to eradicate infection without harming the host, yet pathogens manipulate immunity to establish a chronic infection. Understanding OMS-driven inflammation is essential for disease management, but comprehensive data on immune profiles and immune cell activation during OMS are lacking. Methods Using high-dimensional flow cytometry, we investigated the detailed innate and adaptive systemic immune cell populations in OMS and age- and sex-matched controls. Results Our study revealed that OMS is associated with increased levels of immune regulatory cells, namely T regulatory cells, B regulatory cells, and T follicular regulatory cells. In addition, the expression of immune activation markers HLA-DR and CD86 was decreased in OMS, while the expression of immune exhaustion markers TIM-3, PD-1, PD-L1, and VISTA was increased. Members of the T follicular helper (Tfh) cell family as well as classical and typical memory B cells were significantly increased in OMS individuals. We also found a strong correlation between memory B cells and Tfh cells. Discussion We conclude that OMS skews the host immune system towards the immunomodulatory arm and that the Tfh memory B cell axis is evident in OMS. Therefore, immune-directed therapies may be a promising alternative for eradication and recurrence of infection in OMS, particularly in individuals and areas where antibiotic resistance is a major concern.
Collapse
Affiliation(s)
- Jayagopi Surendar
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Roslind K. Hackenberg
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Fabio Schmitt-Sánchez
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Robert Ossendorff
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Kristian Welle
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Birgit Stoffel-Wagner
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Peter T. Sage
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Christof Burger
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Dieter C. Wirtz
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Andreas C. Strauss
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Frank A. Schildberg
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
6
|
Tchen J, Simon Q, Chapart L, Thaminy MK, Vibhushan S, Saveanu L, Lamri Y, Saidoune F, Pacreau E, Pellefigues C, Bex-Coudrat J, Karasuyama H, Miyake K, Hidalgo J, Fallon PG, Papo T, Blank U, Benhamou M, Hanouna G, Sacre K, Daugas E, Charles N. PD-L1- and IL-4-expressing basophils promote pathogenic accumulation of T follicular helper cells in lupus. Nat Commun 2024; 15:3389. [PMID: 38649353 PMCID: PMC11035650 DOI: 10.1038/s41467-024-47691-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 04/04/2024] [Indexed: 04/25/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by anti-nuclear autoantibodies whose production is promoted by autoreactive T follicular helper (TFH) cells. During SLE pathogenesis, basophils accumulate in secondary lymphoid organs (SLO), amplify autoantibody production and disease progression through mechanisms that remain to be defined. Here, we provide evidence for a direct functional relationship between TFH cells and basophils during lupus pathogenesis, both in humans and mice. PD-L1 upregulation on basophils and IL-4 production are associated with TFH and TFH2 cell expansions and with disease activity. Pathogenic TFH cell accumulation, maintenance, and function in SLO were dependent on PD-L1 and IL-4 in basophils, which induced a transcriptional program allowing TFH2 cell differentiation and function. Our study establishes a direct mechanistic link between basophils and TFH cells in SLE that promotes autoantibody production and lupus nephritis.
Collapse
Affiliation(s)
- John Tchen
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Quentin Simon
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
- Inovarion, 75005, Paris, France
| | - Léa Chapart
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Morgane K Thaminy
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Shamila Vibhushan
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Loredana Saveanu
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Yasmine Lamri
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Fanny Saidoune
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Emeline Pacreau
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Christophe Pellefigues
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Julie Bex-Coudrat
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Hajime Karasuyama
- Inflammation, Infection and Immunity Laboratory, TMDU Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kensuke Miyake
- Inflammation, Infection and Immunity Laboratory, TMDU Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Juan Hidalgo
- Universidad Autonoma de Barcelona, Facultad de Biociencias, Unidad de Fisiologia Animal Bellaterra, Bellaterra Campus, 08193, Barcelona, Spain
| | | | - Thomas Papo
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
- Service de Médecine Interne, Hôpital Bichat, Assistance Publique - Hôpitaux de Paris, 75018, Paris, France
| | - Ulrich Blank
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Marc Benhamou
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
| | - Guillaume Hanouna
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
- Service de Néphrologie, Hôpital Bichat, Assistance Publique - Hôpitaux de Paris, 75018, Paris, France
| | - Karim Sacre
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
- Service de Médecine Interne, Hôpital Bichat, Assistance Publique - Hôpitaux de Paris, 75018, Paris, France
| | - Eric Daugas
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France
- Service de Néphrologie, Hôpital Bichat, Assistance Publique - Hôpitaux de Paris, 75018, Paris, France
| | - Nicolas Charles
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018, Paris, France.
- Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018, Paris, France.
| |
Collapse
|
7
|
Wu J, Zhang X, Lin S, Wei Q, Lin Z, Jin O, Gu J. Alterations in peripheral T- and B-cell subsets in patients with systemic sclerosis. Int J Rheum Dis 2024; 27:e15145. [PMID: 38661314 DOI: 10.1111/1756-185x.15145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 03/22/2024] [Accepted: 03/27/2024] [Indexed: 04/26/2024]
Abstract
OBJECTIVES To determine the alteration of peripheral T and B cell subsets in patients with systemic sclerosis (SSc) and to evaluate their correlation with the progression of SSc. METHODS We recruited 47 SSc patients and 45 healthy controls (HCs) in this study. Demographic and clinical data were then collected. Flow cytometry was used to detect the proportions of 44 different T and B cell subsets in circulating blood. RESULTS The proportion of total B cells (p = .043) decreased in SSc patients, together with similar frequencies of total T cells, CD4+ T cells, and CD8+ T cells in both groups. Several subsets of T and B cells differed significantly between these two groups. Follicular helper T cells-1 (Tfh1) (p < .001), helper T cells-1 (Th1) (p = .001), regulatory T cells (Treg) (p = .004), effector memory CD8+ T cells (p = .041), and cytotoxic T cells-17 (Tc17) (p = .01) were decreased in SSc patients. Follicular helper T cells-2 (Tfh2) (p = .001) and, helper T cells-2 (Th2) (p = .001) levels increased in the SSc group. Regulatory B cells (Breg) (p = .015) were lower in the SSc group, together with marginal zone (MZ) B cells (p < .001), memory B cells (p = .001), and non-switched B cells (p = .005). The modified Rodnan skin score (mRSS) correlated with helper T cells-17 (Th17) (r = -.410, p = .004), Tfh1 (r = -.321, p = .028), peripheral helper T cells (Tph) (r = -.364, p = .012) and plasma cells (r = -.312, p = .033). CONCLUSIONS The alterations in T and B cells implied immune dysfunction, which may play an essential role in systemic sclerosis.
Collapse
Affiliation(s)
- Jialing Wu
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xi Zhang
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Shen Lin
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Qiujing Wei
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhiming Lin
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Ou Jin
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jieruo Gu
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
8
|
Harris EM, Chamseddine S, Chu A, Senkpeil L, Nikiciuk M, Al-Musa A, Woods B, Ozdogan E, Saker S, van Konijnenburg DPH, Yee CS, Nelson R, Lee P, Halyabar O, Hale RC, Day-Lewis M, Henderson LA, Nguyen AA, Elkins M, Ohsumi TK, Gutierrez-Arcelus M, Peyper JM, Platt CD, Grace RF, LaBere B, Chou J. Integrating circulating T follicular memory cells and autoantibody repertoires for characterization of autoimmune disorders. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.25.24303331. [PMID: 38464255 PMCID: PMC10925364 DOI: 10.1101/2024.02.25.24303331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Introduction Autoimmune diseases are heterogeneous and often lack specific or sensitive diagnostic tests. Increased percentages of CD4+CXCR5+PD1+ circulating T follicular helper (cTfh) cells and skewed distributions of cTfh subtypes have been associated with autoimmunity. However, cTfh cell percentages can normalize with immunomodulatory treatment despite persistent disease activity, indicating the need for identifying additional cellular and/or serologic features correlating with autoimmunity. Methods The cohort included 50 controls and 56 patients with autoimmune cytopenias, gastrointestinal, pulmonary, and/or neurologic autoimmune disease. Flow cytometry was used to measure CD4+CXCR5+ T cell subsets expressing the chemokine receptors CXCR3 and/or CCR6: CXCR3+CCR6- Type 1, CXCR3-CCR6- Type 2, CXCR3+CCR6+ Type 1/17, and CXCR3- CCR6+ Type 17 T cells. IgG and IgA autoantibodies were quantified using a microarray featuring 1616 full-length, conformationally intact protein antigens. The 97.5th percentile in the control cohort defined normal limits for T cell subset percentages and total number (burden) of autoantibodies. Results This study focused on CD4+CXCR5+ T cells because CXCR5 upregulation occurs after cognate T-B cell interactions characteristic of autoimmune diseases. We refer to these cells as circulating T follicular memory (cTfm) cells to acknowledge the dynamic nature of antigen-experienced CXCR5+ T cells, which encompass progenitors of cTfh or Tfh cells as well as early effector memory T cells that have not yet lost CXCR5. Compared to controls, 57.1% of patients had increased CXCR5+CXCR3+CCR6+ cTfm1/17 and 25% had increased CXCR5+CXCR3-CCR6+ cTfm17 cell percentages. Patients had significantly more diverse IgG and IgA autoantibodies than controls and 44.6% had an increased burden of autoantibodies of either isotype. Unsupervised autoantibody clustering identified three clusters of patients with IgG autoantibody profiles distinct from those of controls, enriched for patients with active autoimmunity and monogenic diseases. An increased percentage of cTfm17 cells was most closely associated with an increased burden of high-titer IgG and IgA autoantibodies. A composite measure integrating increased cTfm1/17, cTfm17, and high-titer IgG and/or IgA autoantibodies had 91.1% sensitivity and 90.9% specificity for identifying patients with autoimmunity. Percentages of cTfm1/17 and cTfm17 percentages and numbers of high-titer autoantibodies in patients receiving immunomodulatory treatment did not differ from those in untreated patients, thus suggesting that measurements of cTfm can complement measurements of other cellular markers affected by treatment. Conclusions This study highlights two new approaches for assessing autoimmunity: measuring CD4+CXCR5+ cTfm subsets as well as total burden of autoantibodies. Our findings suggest that these approaches are particularly relevant to patients with rare autoimmune disorders for whom target antigens and prognosis are often unknown.
Collapse
Affiliation(s)
- Emily M. Harris
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Sarah Chamseddine
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Anne Chu
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Northeastern University, Boston, MA
| | - Leetah Senkpeil
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthew Nikiciuk
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Northeastern University, Boston, MA
| | - Amer Al-Musa
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian Woods
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Sarife Saker
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Christina S.K. Yee
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ryan Nelson
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Pui Lee
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Olha Halyabar
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Rebecca C. Hale
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Megan Day-Lewis
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Lauren A. Henderson
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Alan A. Nguyen
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Megan Elkins
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Craig D. Platt
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Rachael F. Grace
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Brenna LaBere
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Current affiliation: Division of Allergy and Immunology, Phoenix Children’s Hospital, Phoenix, AZ 85016
| | - Janet Chou
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Omotesho QA, Escamilla A, Pérez-Ruiz E, Frecha CA, Rueda-Domínguez A, Barragán I. Epigenetic targets to enhance antitumor immune response through the induction of tertiary lymphoid structures. Front Immunol 2024; 15:1348156. [PMID: 38333212 PMCID: PMC10851080 DOI: 10.3389/fimmu.2024.1348156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/02/2024] [Indexed: 02/10/2024] Open
Abstract
Tertiary lymphoid structures (TLS) are ectopic lymphoid aggregates found in sites of chronic inflammation such as tumors and autoimmune diseases. The discovery that TLS formation at tumor sites correlated with good patient prognosis has triggered extensive research into various techniques to induce their formation at the tumor microenvironment (TME). One strategy is the exogenous induction of specific cytokines and chemokine expression in murine models. However, applying such systemic chemokine expression can result in significant toxicity and damage to healthy tissues. Also, the TLS formed from exogenous chemokine induction is heterogeneous and different from the ones associated with favorable prognosis. Therefore, there is a need to optimize additional approaches like immune cell engineering with lentiviral transduction to improve the TLS formation in vivo. Similarly, the genetic and epigenetic regulation of the different phases of TLS neogenesis are still unknown. Understanding these molecular regulations could help identify novel targets to induce tissue-specific TLS in the TME. This review offers a unique insight into the molecular checkpoints of the different stages and mechanisms involved in TLS formation. This review also highlights potential epigenetic targets to induce TLS neogenesis. The review further explores epigenetic therapies (epi-therapy) and ongoing clinical trials using epi-therapy in cancers. In addition, it builds upon the current knowledge of tools to generate TLS and TLS phenotyping biomarkers with predictive and prognostic clinical potential.
Collapse
Affiliation(s)
- Quadri Ajibola Omotesho
- Medical Oncology Service (Group of Translational Research in Cancer Immunotherapy and Epigenetics), Regional and Clinical University Hospitals, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Alejandro Escamilla
- Medical Oncology Service (Group of Translational Research in Cancer Immunotherapy and Epigenetics), Regional and Clinical University Hospitals, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
- Department of Human Physiology, Human Histology, Pathological Anatomy and Physical Sport Education, University of Malaga, Malaga, Spain
| | - Elisabeth Pérez-Ruiz
- Medical Oncology Service (Group of Translational Research in Cancer Immunotherapy and Epigenetics), Regional and Clinical University Hospitals, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
| | - Cecilia A. Frecha
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Civil Hospital, Malaga, Spain
| | - Antonio Rueda-Domínguez
- Medical Oncology Service (Group of Translational Research in Cancer Immunotherapy and Epigenetics), Regional and Clinical University Hospitals, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
| | - Isabel Barragán
- Medical Oncology Service (Group of Translational Research in Cancer Immunotherapy and Epigenetics), Regional and Clinical University Hospitals, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
- Group of Pharmacoepigenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
LaBere B, Chu A, Platt CD, Chou J. The Integration of Patient-Reported Quality of Life and Systemic Biomarkers in Patients with Immune Dysregulation. RESEARCH SQUARE 2023:rs.3.rs-3270389. [PMID: 37674702 PMCID: PMC10479437 DOI: 10.21203/rs.3.rs-3270389/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Background Patient-reported quality of life measurements are an important method for improving the treatment of patients with a variety of diseases. These tools have been minimally investigated in patients with inborn errors of immunity (IEI). Patients with IEI may have immune dysregulation and autoimmune-mediated multi-system organ involvement, making treatment optimization vitally important. Routine laboratory and radiologic testing are typically used for treatment monitoring; however, these modalities have the potential to miss early organ damage. T follicular helper cells are T cells that contribute to antibody production and are known to be expanded in patients with active autoimmunity. We hypothesized that a combination of patient-reported quality of life measurements, in addition to T follicular helper cell percentages, would help us to better understand the level of disease activity in patients with IEI and autoimmunity. Methods Patients with immune dysregulation were consented to provide a blood sample and to complete a questionnaire. The Centers for Disease Control HRQOL-14 tool was utilized for the questionnaire portion, and T follicular helper cell levels were measured from whole blood using surface staining and flow cytometry analysis. Patient disease activity was abstracted from the patient medical record, and this was compared to the questionnaire and whole blood assay results. Results A total of 20 patients participated in the study; 8 patients had active disease and the remaining were found to be quiescent. There was no significant difference between the patient-reported general health ratings based on sex, age, disease activity, or category of immune dysregulation (p > 0.05). The cTfh percentages were expanded in patients with active disease as compared to those with quiescent (p < 0.05). However, there was no significant correlation between cTfh percentage and patient-reported unhealthy days from the questionnaire (R2 = 0.113, p > 0.05). Conclusions Patients with active immune dysregulation were found to have expanded cTfh percentages as compared to those with quiescent disease, however this was not reflected in patient-reported quality of life questionnaires. Better understanding of disease activity and the patient experience is vital to optimize appropriate treatments and outcomes for patients with IEI and immune dysregulation, and more investigation is needed.
Collapse
|
11
|
Yin S, Wang J, Chen L, Mao M, Rahma I, Geng Y, Huang R, Tong X, Liu Y, Wu C, Chen Y, Li J. Circulating Th2-biased T follicular helper cells impede antiviral humoral responses during chronic hepatitis B infection through upregulating CTLA4. Antiviral Res 2023:105665. [PMID: 37421985 DOI: 10.1016/j.antiviral.2023.105665] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 05/22/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
Failure in curing chronic hepatitis B (CHB) caused by hepatitis B virus (HBV) can lead to functional impairment of B cells. Cytotoxic T-lymphocyte associated antigen 4 (CTLA4) regulates B cell and T follicular helper (Tfh) cell differentiation. In addition, Tfh cells play a critical role in helping B cells generate antibodies upon pathogen exposure. Here, we analyzed the global and HBsAg-specific B cells and circulating Tfh (cTfh) cells using samples from treatment-naïve and Peg-IFN-α-treated CHB patients and healthy subjects. Compared to healthy subjects, CTLA4 expression was significantly increased in cTfh cells, from CHB patients. The frequency of CTLA4+cTfh2 cells was negatively correlated with that of HBsAg-specific resting memory B cells. Importantly, inhibition of CTLA4 restored HBsAb secretion and promoted plasma cell differentiation. In addition, CTLA4+cTfh2 cells from CHB patients were ineffective in providing B cell help. Both expression of CTLA4 in cTfh and cTfh2 cells and ratios of CLTA4+cTfh and CTLA4+cTfh2 cells were significantly decreased in Peg-IFN-α-treated CHB patients who showed complete responses. Thus, our results highlighted that cTh2-biased T follicular helper cells could impede antiviral humoral responses during chronic HBV infection by upregulating CTLA4, suggesting that further optimizing potent Tfh cell responses may promote functional cure of CHB.
Collapse
Affiliation(s)
- Shengxia Yin
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China; Institute of Viruses and Infectious Diseases, Nanjing University, Jiangsu, China
| | - Jian Wang
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China; Institute of Viruses and Infectious Diseases, Nanjing University, Jiangsu, China
| | - Lin Chen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Minxin Mao
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Issa Rahma
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yu Geng
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Rui Huang
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China; Institute of Viruses and Infectious Diseases, Nanjing University, Jiangsu, China
| | - Xin Tong
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China; Institute of Viruses and Infectious Diseases, Nanjing University, Jiangsu, China
| | - Yong Liu
- Institute of Viruses and Infectious Diseases, Nanjing University, Jiangsu, China; Department of Experimental Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chao Wu
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China; Institute of Viruses and Infectious Diseases, Nanjing University, Jiangsu, China.
| | - Yuxin Chen
- Institute of Viruses and Infectious Diseases, Nanjing University, Jiangsu, China; Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Jie Li
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China; Institute of Viruses and Infectious Diseases, Nanjing University, Jiangsu, China.
| |
Collapse
|
12
|
Gong M, Choi SC, Park YP, Zou X, Elshikha AS, Gerriets VA, Rathmell JC, Mohamazadeh M, Morel L. Transcriptional and metabolic programs promote the expansion of follicular helper T cells in lupus-prone mice. iScience 2023; 26:106774. [PMID: 37216123 PMCID: PMC10197114 DOI: 10.1016/j.isci.2023.106774] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/28/2023] [Accepted: 04/24/2023] [Indexed: 05/24/2023] Open
Abstract
The expansion of follicular helper T (Tfh) cells, which is tightly associated with the development of lupus, is reversed by the inhibition of either glycolysis or glutaminolysis in mice. Here we analyzed the gene expression and metabolome of Tfh cells and naive CD4+ T (Tn) cells in the B6.Sle1.Sle2.Sle3 (triple congenic, TC) mouse model of lupus and its congenic B6 control. Lupus genetic susceptibility in TC mice drives a gene expression signature starting in Tn cells and expanding in Tfh cells with enhanced signaling and effector programs. Metabolically, TC Tn and Tfh cells showed multiple defective mitochondrial functions. TC Tfh cells also showed specific anabolic programs including enhanced glutamate metabolism, malate-aspartate shuttle, and ammonia recycling, as well as altered dynamics of amino acid content and their transporters. Thus, our study has revealed specific metabolic programs that can be targeted to specifically limit the expansion of pathogenic Tfh cells in lupus.
Collapse
Affiliation(s)
- Minghao Gong
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Seung-Chul Choi
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Yuk Pheel Park
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Xueyang Zou
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Ahmed S. Elshikha
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Valerie A. Gerriets
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jeffrey C. Rathmell
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Mansour Mohamazadeh
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Laurence Morel
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
13
|
Starshinova A, Zinchenko Y, Malkova A, Kudlay D, Kudryavtsev I, Yablonskiy P. Sarcoidosis and Autoimmune Inflammatory Syndrome Induced by Adjuvants. Life (Basel) 2023; 13:1047. [PMID: 37109576 PMCID: PMC10145559 DOI: 10.3390/life13041047] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/16/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Currently, sarcoidosis remains one of the diseases with unknown etiology, which significantly complicates its diagnosis and treatment. Various causes of sarcoidosis have been studied for many years. Both organic and inorganic trigger factors, provoking the development of granulomatous inflammation are considered. However, the most promising and evidence-based hypothesis is the development of sarcoidosis as an autoimmune disease, provoked by various adjuvants in genetic predisposed individuals. This concept fits into the structure of the autoimmune/inflammatory syndrome, induced by adjuvants (ASIA) that was proposed in 2011 by Professor Shoenfeld Y. In this paper, the authors reveal the presence of major and minor ASIA criteria for sarcoidosis, propose a new concept of the course of sarcoidosis within the framework of ASIA, and point out the difficulties in creating a model of the disease and the selection of therapy. It is obvious that the data obtained not only bring us closer to understanding the nature of sarcoidosis, but also potentiate new studies confirming this hypothesis by obtaining a model of the disease.
Collapse
Affiliation(s)
- Anna Starshinova
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia;
| | - Yulia Zinchenko
- Saint-Petersburg Research Institute of Phthisiopulmonology, 194064 Saint-Petersburg, Russia (P.Y.)
| | - Anna Malkova
- Laboratory of the Mosaic of Autoimmunity, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia;
| | - Dmitriy Kudlay
- Medical Department, Sechenov First Moscow State Medical University, 119435 Moscow, Russia
- Institute of Immunology, 115478 Moscow, Russia
| | - Igor Kudryavtsev
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia;
- Department of Immunology, Institution of Experimental Medicine, 197022 Saint-Petersburg, Russia
| | - Piotr Yablonskiy
- Saint-Petersburg Research Institute of Phthisiopulmonology, 194064 Saint-Petersburg, Russia (P.Y.)
- Laboratory of the Mosaic of Autoimmunity, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia;
| |
Collapse
|
14
|
Kudryavtsev I, Zinchenko Y, Starshinova A, Serebriakova M, Malkova A, Akisheva T, Kudlay D, Glushkova A, Yablonskiy P, Shoenfeld Y. Circulating Regulatory T Cell Subsets in Patients with Sarcoidosis. Diagnostics (Basel) 2023; 13:1378. [PMID: 37189479 PMCID: PMC10137313 DOI: 10.3390/diagnostics13081378] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Over recent years, many researchers have supported the autoimmune theory of sarcoidosis. The presence of uncontrolled inflammatory response on local and system levels in patients with sarcoidosis did not define that the immunoregulatory mechanisms could be affected. The aim of this study was to evaluate the distribution and the disturbance circulating Treg cell subsets in the peripheral blood in patients with sarcoidosis. MATERIALS AND METHODS A prospective comparative study was performed in 2016-2018 (34 patients with sarcoidosis (men (67.6%), women (32.3%)) were examined). Healthy subjects-the control group (n = 40). The diagnosis of pulmonary sarcoidosis was performed according to the standard criteria. We used two ten-color combinations of antibodies for Treg immunophenotyping. The first one contained CD39-FITC, CD127-PE, CCR4-PE/Dazzle™ 594, CD25-PC5.5, CD161-PC7, CD4-APC, CD8-APC-AF700, CD3-APC/Cy7, HLA-DR-PacBlue, and CD45 RA-BV 510™, while the second consisted of CXCR3-Alexa Fluor 488, CD25-РЕ, CXCR5-РЕ/Dazzle™ 594, CCR4-PerСP/Сy5.5, CCR6-РЕ/Cy7, CD4-АPC, CD8 АPC-AF700, CD3-АPC/Cy7, CCR7-BV 421, and CD45 RA-BV 510. The flow cytometry data were analyzed by using Kaluza software v2.3. A statistical analysis was performed with Statistica 7.0 and GraphPad Prism 8 software packages. RESULTS OF THE STUDY Primarily, we found that patients with sarcoidosis had decreased absolute numbers of Treg cells in circulation. We noted that the level of CCR7-expressing Tregs decreased in patients with sarcoidosis vs. the control group (65.55% (60.08; 70.60) vs. 76.93% (69.59; 79.86) with p < 0.001). We noticed that the relative numbers of CD45RA-CCR7+ Tregs decreased in patients with sarcoidosis (27.11% vs. 35.43%, p < 0.001), while the frequency of CD45 RA-CCR7- and CD45RA+ CCR7- Tregs increased compared to the control group (33.3% vs. 22.73% and 0.76% vs. 0.51% with p < 0.001 and p = 0.028, respectively). CXCR3-expressing Treg cell subsets-Th1-like CCR60078CXCR3+ Tregs and Th17.1-like CCR6+ CXCR3+ Tregs-significantly increased in patients with sarcoidosis vs. the control group (14.4% vs. 10.5% with p < 0.01 and 27.9% vs. 22.8% with p < 0.01, respectively). Furthermore, the levels of peripheral blood EM Th17-like Tregs significantly decreased in the sarcoidosis group vs. the control group (36.38% vs. 46.70% with p < 0.001). Finally, we found that CXCR5 expression was increased in CM Tregs cell subsets in patients with sarcoidosis. CONCLUSIONS Our data indicated a decrease in circulating Tregs absolute numbers and several alterations in Treg cell subsets. Moreover, our results highlight the presence of increased levels of CM CXCR5+ follicular Tregs in the periphery that could be linked with the imbalance of follicular Th cell subsets and alterations in B cell, based on the immune response. The balance between the two functionally distinct Treg cell populations-Th1-like and Th17-like Tregs-could be used in sarcoidosis diagnosis and the determination of prognosis and disease outcomes. Furthermore, we want to declare that analysis of Treg numbers of phenotypes could fully characterize their functional activity in peripherally inflamed tissues.
Collapse
Affiliation(s)
- Igor Kudryavtsev
- Department of Immunology, Institution of Experimental Medicine, 197376 St. Petersburg, Russia
| | - Yulia Zinchenko
- Phthisiopulmonology Department, St. Petersburg Research Institute of Phthisiopulmonology, 194064 St. Petersburg, Russia
| | - Anna Starshinova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia
| | - Maria Serebriakova
- Department of Immunology, Institution of Experimental Medicine, 197376 St. Petersburg, Russia
| | - Anna Malkova
- Laboratory of the Mosaic of Autoimmunity, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Tatiana Akisheva
- Department of Immunology, Institution of Experimental Medicine, 197376 St. Petersburg, Russia
| | - Dmitriy Kudlay
- Department of Pharmacology, Sechenov First Moscow State Medical University, 119992 Moscow, Russia
- Institute of Immunology, 115552 Moscow, Russia
| | - Anzhela Glushkova
- Bekhterev National Research Medical Center for Psychiatry and Neurology, 19201 St. Petersburg, Russia
| | - Piotr Yablonskiy
- Phthisiopulmonology Department, St. Petersburg Research Institute of Phthisiopulmonology, 194064 St. Petersburg, Russia
- Laboratory of the Mosaic of Autoimmunity, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Yehuda Shoenfeld
- Laboratory of the Mosaic of Autoimmunity, St. Petersburg State University, 199034 St. Petersburg, Russia
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer 5265601, Israel
| |
Collapse
|
15
|
Hara A, Chihara N, Akatani R, Nishigori R, Tsuji A, Yoshimura H, Kawamoto M, Otsuka Y, Kageyama Y, Kondo T, Leypoldt F, Wandinger KP, Matsumoto R. Circulating plasmablasts and follicular helper T-cell subsets are associated with antibody-positive autoimmune epilepsy. Front Immunol 2022; 13:1048428. [PMID: 36569937 PMCID: PMC9773883 DOI: 10.3389/fimmu.2022.1048428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/31/2022] [Indexed: 12/13/2022] Open
Abstract
Autoimmune epilepsy (AE) is an inflammatory disease of the central nervous system with symptoms that have seizures that are refractory to antiepileptic drugs. Since the diagnosis of AE tends to rely on a limited number of anti-neuronal antibody tests, a more comprehensive analysis of the immune background could achieve better diagnostic accuracy. This study aimed to compare the characteristics of anti-neuronal antibody-positive autoimmune epilepsy (AE/Ab(+)) and antibody-negative suspected autoimmune epilepsy (AE/Ab(-)) groups. A total of 23 patients who met the diagnostic criteria for autoimmune encephalitis with seizures and 11 healthy controls (HC) were enrolled. All patients were comprehensively analyzed for anti-neuronal antibodies; 13 patients were identified in the AE/Ab(+) group and 10 in the AE/Ab(-) group. Differences in clinical characteristics, including laboratory and imaging findings, were evaluated between the groups. In addition, the immunophenotype of peripheral blood mononuclear cells (PBMCs) and CSF mononuclear cells, particularly B cells and circulating Tfh (cTfh) subsets, and multiplex assays of serum and CSF were analyzed using flow cytometry. Patients with AE/Ab(+) did not show any differences in clinical parameters compared to patients with AE/Ab(-). However, the frequency of plasmablasts within PBMCs and CSF in patients with AE/Ab(+) was higher than that in patients with AE/Ab(-) and HC, and the frequency of cTfh17 cells and inducible T-cell co-stimulator (ICOS) expressing cTfh17 cells within cTfh subsets was higher than that in patients with AE/Ab(-). Furthermore, the frequency of ICOShighcTfh17 cells was positively correlated with that of the unswitched memory B cells. We also found that IL-12, IL-23, IL-6, IL-17A, and IFN-γ levels were elevated in the serum and IL-17A and IL-6 levels were elevated in the CSF of patients with AE/Ab(+). Our findings indicate that patients with AE/Ab(+) showed increased differentiation of B cells and cTfh subsets associated with antibody production. The elevated frequency of plasmablasts and ICOS expressing cTfh17 shift in PBMCs may be indicative of the presence of antibodies in patients with AE.
Collapse
Affiliation(s)
- Atsushi Hara
- Division of Neurology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Norio Chihara
- Division of Neurology, Kobe University Graduate School of Medicine, Kobe, Japan,*Correspondence: Norio Chihara, ; Riki Matsumoto,
| | - Ritsu Akatani
- Division of Neurology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ryusei Nishigori
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Asato Tsuji
- Division of Neurology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hajime Yoshimura
- Department of Neurology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Michi Kawamoto
- Department of Neurology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Yoshihisa Otsuka
- Department of Neurology, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Japan
| | - Yasufumi Kageyama
- Department of Neurology, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Japan
| | - Takayuki Kondo
- Department of Neurology, Kansai Medical University Medical Center, Moriguchi, Japan
| | - Frank Leypoldt
- Neuroimmunology, Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Kiel, Germany,Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Klaus-Peter Wandinger
- Neuroimmunology, Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Riki Matsumoto
- Division of Neurology, Kobe University Graduate School of Medicine, Kobe, Japan,*Correspondence: Norio Chihara, ; Riki Matsumoto,
| |
Collapse
|
16
|
Duan Z, Jia A, Cui W, Feng J. Correlation between neutrophil-to-lymphocyte ratio and severity of myasthenia gravis in adults: A retrospective study. J Clin Neurosci 2022; 106:117-121. [DOI: 10.1016/j.jocn.2022.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 10/01/2022] [Accepted: 10/14/2022] [Indexed: 11/15/2022]
|
17
|
Shesternya PA, Savchenko AA, Gritsenko OD, Vasileva AO, Kudryavtsev IV, Masterova AA, Isakov DV, Borisov AG. Features of Peripheral Blood Th-Cell Subset Composition and Serum Cytokine Level in Patients with Activity-Driven Ankylosing Spondylitis. Pharmaceuticals (Basel) 2022; 15:ph15111370. [PMID: 36355542 PMCID: PMC9695783 DOI: 10.3390/ph15111370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
Th cells may exhibit pathological activity depending on the regulatory and functional signals sensed under a wide range of immunopathological conditions, including ankylosing spondylitis (AS). The relationship between Th cells and cytokines is important for diagnoses and for determining treatment. Accordingly, the aim of this study was to investigate the relationship between Th-cell subset composition and serum cytokine profile for patients with activity-driven AS. In our study, patients were divided into two groups according to disease activity: low-activity AS (ASDAS-CRP < 2.1) and high-activity AS (ASDAS-CRP > 2.1). The peripheral blood Th cell subset composition was studied by flow cytometry. Using multiplex analysis, serum cytokine levels were quantified and investigated. It was found that only patients with high-activity AS had reduced central memory (CM) Th1 cells (p = 0.035) but elevated numbers of CM (p = 0.014) and effector memory (EM) Th2 cells (p < 0.001). However, no activity-driven change in the Th17 cell subset composition was observed in AS patients. Moreover, low-AS activity patients had increased numbers of Tfh17 EM cells (p < 0.001), whereas high-AS activity was associated with elevated Tfh2 EM level (p = 0.031). The serum cytokine profiles in AS patients demonstrated that cues stimulating cellular immunity were increased, but patients with high-AS activity reveled increased IL-5 level (p = 0.017). Analyzing the data obtained from AS patients allowed us to conclude that Th cell subset differentiation was mainly affected during the CM stage and characterized the IL-23/IL-17 regulatory axis, whereas increased humoral immunity was observed in the high-AS activity group.
Collapse
Affiliation(s)
- Pavel A. Shesternya
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Ministry of Healthcare, 660022 Krasnoyarsk, Russia
- Correspondence:
| | - Andrei A. Savchenko
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Ministry of Healthcare, 660022 Krasnoyarsk, Russia
- Federal Research Center “Krasnoyarsk Science Center”, Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia
| | - Olga D. Gritsenko
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Ministry of Healthcare, 660022 Krasnoyarsk, Russia
| | - Alexandra O. Vasileva
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Ministry of Healthcare, 660022 Krasnoyarsk, Russia
| | | | - Alena A. Masterova
- Federal Research Center “Krasnoyarsk Science Center”, Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia
| | - Dmitry V. Isakov
- Academician I.P. Pavlov First St. Petersburg State Medical University, Ministry of Healthcare, 197022 St. Peterburg, Russia
| | - Alexandr G. Borisov
- Federal Research Center “Krasnoyarsk Science Center”, Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia
| |
Collapse
|
18
|
An Q, Zhao J, Zhu X, Yang B, Wu Z, Su Y, Zhang L, Xu K, Ma D. Exploiting the role of T cells in the pathogenesis of Sjögren's syndrome for therapeutic treatment. Front Immunol 2022; 13:995895. [PMID: 36389806 PMCID: PMC9650646 DOI: 10.3389/fimmu.2022.995895] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 10/17/2022] [Indexed: 08/19/2023] Open
Abstract
Sjögrens syndrome (SS) is caused by autoantibodies that attack proprioceptive salivary and lacrimal gland tissues. Damage to the glands leads to dry mouth and eyes and affects multiple systems and organs. In severe cases, SS is life-threatening because it can lead to interstitial lung disease, renal insufficiency, and lymphoma. Histological examination of the labial minor salivary glands of patients with SS reveals focal lymphocyte aggregation of T and B cells. More studies have been conducted on the role of B cells in the pathogenesis of SS, whereas the role of T cells has only recently attracted the attention of researchers. This review focusses on the role of various populations of T cells in the pathogenesis of SS and the progress made in research to therapeutically targeting T cells for the treatment of patients with SS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Dan Ma
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
19
|
Nelke C, Pawlitzki M, Schroeter CB, Huntemann N, Räuber S, Dobelmann V, Preusse C, Roos A, Allenbach Y, Benveniste O, Wiendl H, Lundberg IE, Stenzel W, Meuth SG, Ruck T. High-Dimensional Cytometry Dissects Immunological Fingerprints of Idiopathic Inflammatory Myopathies. Cells 2022; 11:3330. [PMID: 36291195 PMCID: PMC9601098 DOI: 10.3390/cells11203330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/05/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022] Open
Abstract
Chronic inflammation of skeletal muscle is the common feature of idiopathic inflammatory myopathies (IIM). Given the rarity of the disease and potential difficulty of routinely obtaining target tissue, i.e., standardized skeletal muscle, our understanding of immune signatures of the IIM spectrum remains incomplete. Further insight into the immune topography of IIM is needed to determine specific treatment targets according to clinical and immunological phenotypes. Thus, we used high-dimensional flow cytometry to investigate the immune phenotypes of anti-synthetase syndrome (ASyS), dermatomyositis (DM) and inclusion-body myositis (IBM) patients as representative entities of the IIM spectrum and compared them to healthy controls. We studied the CD8, CD4 and B cell compartments in the blood aiming to provide a contemporary overview of the immune topography of the IIM spectrum. ASyS was characterized by altered CD4 composition and expanded T follicular helper cells supporting B cell-mediated autoimmunity. For DM, unsupervised clustering identified expansion of distinct B cell subtypes highly expressing immunoglobulin G4 (IgG4) and CD38. Lastly, terminally differentiated, cytotoxic CD8 T cells distinguish IBM from other IIM. Interestingly, these terminally differentiated CD8 T cells highly expressed the integrin CD18 mediating cellular adhesion and infiltration. The distinct immune cell topography of IIM might provide the framework for targeted treatment approaches potentially improving therapeutic outcomes.
Collapse
Affiliation(s)
- Christopher Nelke
- Department of Neurology, Medical Faculty, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, 48149 Münster, Germany
| | - Marc Pawlitzki
- Department of Neurology, Medical Faculty, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, 48149 Münster, Germany
| | - Christina B. Schroeter
- Department of Neurology, Medical Faculty, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany
| | - Niklas Huntemann
- Department of Neurology, Medical Faculty, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany
| | - Saskia Räuber
- Department of Neurology, Medical Faculty, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany
| | - Vera Dobelmann
- Department of Neurology, Medical Faculty, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany
| | - Corinna Preusse
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Andreas Roos
- Department of Neuropediatrics, University of Duisburg-Essen, 45147 Essen, Germany
| | - Yves Allenbach
- Service de Médecine Interne et Immunologie Clinique, University Hospital Pitié Salpêtrière, 75013 Paris, France
| | - Olivier Benveniste
- Service de Médecine Interne et Immunologie Clinique, University Hospital Pitié Salpêtrière, 75013 Paris, France
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, 48149 Münster, Germany
| | - Ingrid E. Lundberg
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, and Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Werner Stenzel
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Sven G. Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany
| |
Collapse
|
20
|
Glazanova TV, Shilova ER. Immune system disturbances after a new coronavirus infection COVID-19. JOURNAL INFECTOLOGY 2022. [DOI: 10.22625/2072-6732-2022-14-4-26-37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
During the pandemic, a large number of works devoted to COVID infection have appeared, which have made it possible to understand the pathogenetic features of the disease and to accumulate significant clinical experience. However, the question remains about the degree of participation of humoral and cellular (primarily T-cell) immunity in the mechanisms of immune defense and resistance to COVID-19, the individual features of the immune response in different subjects. Post-COVID syndrome is currently a separate diagnosis included in the ICD-10 International Classification of Diseases, but the long-term effects of the SARS-CoV-2 on the immune system are not yet well established. At the same time, a long-term increased activity of the immune system can contribute to the development of autoimmune reactions. The review of the literature presents the results of studies, mainly devoted to immune system disorders after COVID infection. The changes in subpopulations of T-lymphocytes, B-lymphocytes, their functional properties, the complement system and other factors of humoral immunity, as well as the production of a number of cytokines are described. Data on immune disorders in post-COVID syndrome and during the convalescence period are presented in detail. Since COVID-19 is an infection that has a significant impact on the hematopoietic system and hemostasis, special attention is paid to the category of subjects with an increased risk of severe complications. Among the latter are elderly patients, persons suffering from diabetes mellitus, oncological and oncohematological patients, in particular, with hematopoietic and lymphoid tissue neoplasia, such as chronic lymphocytic leukemia, lymphoma, multiple myeloma. The review pays special attention to the peculiarities of the course of COVID-19 and the response of the immune system to vaccination in patients with oncohematological diseases. Deciphering the significance of individual links of cellular and humoral immunity in patients who have undergone COVID-19 is an important issue in creating effective vaccines and improving therapeutic methods.
Collapse
Affiliation(s)
| | - E. R. Shilova
- Russian Research institute of hematology and transfusiology
| |
Collapse
|
21
|
Yaping W, Zhe W, Zhuling C, Ruolei L, Pengyu F, Lili G, Cheng J, Bo Z, Liuyin L, Guangdong H, Yaoling W, Niuniu H, Rui L. The soldiers needed to be awakened: Tumor-infiltrating immune cells. Front Genet 2022; 13:988703. [PMID: 36246629 PMCID: PMC9558824 DOI: 10.3389/fgene.2022.988703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/29/2022] [Indexed: 11/18/2022] Open
Abstract
In the tumor microenvironment, tumor-infiltrating immune cells (TIICs) are a key component. Different types of TIICs play distinct roles. CD8+ T cells and natural killer (NK) cells could secrete soluble factors to hinder tumor cell growth, whereas regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) release inhibitory factors to promote tumor growth and progression. In the meantime, a growing body of evidence illustrates that the balance between pro- and anti-tumor responses of TIICs is associated with the prognosis in the tumor microenvironment. Therefore, in order to boost anti-tumor response and improve the clinical outcome of tumor patients, a variety of anti-tumor strategies for targeting TIICs based on their respective functions have been developed and obtained good treatment benefits, including mainly immune checkpoint blockade (ICB), adoptive cell therapies (ACT), chimeric antigen receptor (CAR) T cells, and various monoclonal antibodies. In recent years, the tumor-specific features of immune cells are further investigated by various methods, such as using single-cell RNA sequencing (scRNA-seq), and the results indicate that these cells have diverse phenotypes in different types of tumors and emerge inconsistent therapeutic responses. Hence, we concluded the recent advances in tumor-infiltrating immune cells, including functions, prognostic values, and various immunotherapy strategies for each immune cell in different tumors.
Collapse
Affiliation(s)
- Wang Yaping
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Wang Zhe
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Chu Zhuling
- Department of General Surgery, Eastern Theater Air Force Hospital of PLA, Nanjing, China
| | - Li Ruolei
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Fan Pengyu
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Guo Lili
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Ji Cheng
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhang Bo
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Liu Liuyin
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Hou Guangdong
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Wang Yaoling
- Department of Geriatrics, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hou Niuniu
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- Department of General Surgery, Eastern Theater Air Force Hospital of PLA, Nanjing, China
- *Correspondence: Hou Niuniu, ; Ling Rui,
| | - Ling Rui
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Hou Niuniu, ; Ling Rui,
| |
Collapse
|
22
|
Jenkins D, Phalke S, Bell R, Lessard S, Gupta S, Youssef M, Tam K, Nocon A, Rivera-Correa J, Wright T, Sculco T, Otero M, Pernis AB, Sculco P. Adaptive immune responses in patients requiring revision after total knee arthroplasty. J Orthop Res 2022; 41:984-993. [PMID: 36121317 DOI: 10.1002/jor.25445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/25/2022] [Accepted: 09/14/2022] [Indexed: 02/04/2023]
Abstract
Dissatisfaction occurs in nearly 20% of patients after total knee arthroplasty (TKA); however, there remains only limited understanding of the biologic mechanisms that may contribute to suboptimal postoperative outcomes requiring revision surgery. Expansion of effector T and B cells, could promote an abnormal healing response via local or peripheral immune system mechanisms and contribute to inferior outcomes necessitating revision TKA. In this pilot study, we hypothesized that patients suffering from complications of arthrofibrosis or instability may exhibit differences in adaptive immune function. Patients (n = 31) undergoing revision TKA for an indication of arthrofibrosis or instability were prospectively enrolled. Whole blood and synovial fluid (SF) from the operative knee were collected at time of surgery. Peripheral blood mononuclear cells were isolated and analyzed by flow cytometry. Serum and SF were assessed for immunoglobulin levels by Luminex and antiphospholipid antibodies by enzyme-linked immunoassay. No significant differences were observed in peripheral blood T/B cell populations or serum immunoglobulins levels between groups. SF analysis demonstrated significant differences between the two groups, with higher levels of immunoglobulin G1 (IgG1) (p = 0.0184), IgG3 (p = 0.0084) and antiphosphatidyl serine IgG (p = 0.034) in arthrofibrosis relative to instability patients. Increased levels of both IgG subclasses and antiphospholipid antibodies in the SF suggest that intra-articular T-B cell interactions, potentially triggered by exposure to apoptotic components generated during post-op healing, could be functioning as a source of immune complexes that fuel fibrous tissue growth in arthrofibrotic patients.
Collapse
Affiliation(s)
- Daniel Jenkins
- HSS Research Institute, Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York City, New York, USA
| | - Swati Phalke
- HSS Research Institute, Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York City, New York, USA
| | - Richard Bell
- HSS Research Institute, Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York City, New York, USA
- HSS Research Institute, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York City, New York, USA
| | - Samantha Lessard
- HSS Research Institute, Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York City, New York, USA
| | - Sanjay Gupta
- HSS Research Institute, Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York City, New York, USA
| | - Mark Youssef
- Department of Orthopedic Surgery, Stavros Niarchos Foundation Complex Joint Reconstruction Center, Hospital for Special Surgery, New York City, New York, USA
| | - Kathleen Tam
- Department of Orthopedic Surgery, Stavros Niarchos Foundation Complex Joint Reconstruction Center, Hospital for Special Surgery, New York City, New York, USA
| | - Allina Nocon
- Department of Orthopedic Surgery, Stavros Niarchos Foundation Complex Joint Reconstruction Center, Hospital for Special Surgery, New York City, New York, USA
| | - Juan Rivera-Correa
- HSS Research Institute, Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York City, New York, USA
| | - Timothy Wright
- Department of Biomechanics, Hospital for Special Surgery, New York City, New York, USA
| | - Thomas Sculco
- Department of Orthopedic Surgery, Stavros Niarchos Foundation Complex Joint Reconstruction Center, Hospital for Special Surgery, New York City, New York, USA
| | - Miguel Otero
- HSS Research Institute, Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York City, New York, USA
- HSS Research Institute, Orthopedic Soft Tissue Research Program, Weill Cornell Medical College, New York City, New York, USA
| | - Alessandra B Pernis
- HSS Research Institute, Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York City, New York, USA
- HSS Research Institute, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York City, New York, USA
- Department of Medicine, Weill Cornell Medicine, New York City, New York, USA
- Department of Medicine, Immunology & Microbial Pathogenesis, Weill Cornell Medicine, New York City, New York, USA
| | - Peter Sculco
- Department of Orthopedic Surgery, Stavros Niarchos Foundation Complex Joint Reconstruction Center, Hospital for Special Surgery, New York City, New York, USA
| |
Collapse
|
23
|
Al Moussawy M, Abdelsamed HA. Non-cytotoxic functions of CD8 T cells: “repentance of a serial killer”. Front Immunol 2022; 13:1001129. [PMID: 36172358 PMCID: PMC9511018 DOI: 10.3389/fimmu.2022.1001129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/25/2022] [Indexed: 12/01/2022] Open
Abstract
Cytotoxic CD8 T cells (CTLs) are classically described as the “serial killers” of the immune system, where they play a pivotal role in protective immunity against a wide spectrum of pathogens and tumors. Ironically, they are critical drivers of transplant rejection and autoimmune diseases, a scenario very similar to the famous novel “The strange case of Dr. Jekyll and Mr. Hyde”. Until recently, it has not been well-appreciated whether CTLs can also acquire non-cytotoxic functions in health and disease. Several investigations into this question revealed their non-cytotoxic functions through interactions with various immune and non-immune cells. In this review, we will establish a new classification for CD8 T cell functions including cytotoxic and non-cytotoxic. Further, we will discuss this novel concept and speculate on how these functions could contribute to homeostasis of the immune system as well as immunological responses in transplantation, cancer, and autoimmune diseases.
Collapse
Affiliation(s)
- Mouhamad Al Moussawy
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Starzl Transplantation Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Hossam A. Abdelsamed
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Starzl Transplantation Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Pittsburgh Liver Research Center, School of Medicine, Pittsburgh, PA, United States
- *Correspondence: Hossam A. Abdelsamed,
| |
Collapse
|
24
|
Wang B, Wang M, Ao D, Wei X. CXCL13-CXCR5 axis: Regulation in inflammatory diseases and cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188799. [PMID: 36103908 DOI: 10.1016/j.bbcan.2022.188799] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 01/10/2023]
Abstract
Chemokine C-X-C motif ligand 13 (CXCL13), originally identified as a B-cell chemokine, plays an important role in the immune system. The interaction between CXCL13 and its receptor, the G-protein coupled receptor (GPCR) CXCR5, builds a signaling network that regulates not only normal organisms but also the development of many diseases. However, the precise action mechanism remains unclear. In this review, we discussed the functional mechanisms of the CXCL13-CXCR5 axis under normal conditions, with special focus on its association with diseases. For certain refractory diseases, we emphasize the diagnostic and therapeutic role of CXCL13-CXCR5 axis.
Collapse
Affiliation(s)
- Binhan Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Manni Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Danyi Ao
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
25
|
George PJ, Marches R, Nehar-Belaid D, Banchereau J, Lustigman S. The Th1/Tfh-like biased responses elicited by the rASP-1 innate adjuvant are dependent on TRIF and Type I IFN receptor pathways. Front Immunol 2022; 13:961094. [PMID: 36119026 PMCID: PMC9478378 DOI: 10.3389/fimmu.2022.961094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/12/2022] [Indexed: 11/23/2022] Open
Abstract
Ov-ASP-1 (rASP-1), a parasite-derived protein secreted by the helminth Onchocerca volvulus, is an adjuvant which enhances the potency of the influenza trivalent vaccine (IIV3), even when used with 40-fold less IIV3. This study is aimed to provide a deeper insight into the molecular networks that underline the adjuvanticity of rASP-1. Here we show that rASP-1 stimulates mouse CD11c+ bone marrow-derived dendritic (BMDCs) to secrete elevated levels of IL-12p40, TNF-α, IP-10 and IFN-β in a TRIF-dependent but MyD88-independent manner. rASP-1-activated BMDCs promoted the differentiation of naïve CD4+ T cells into Th1 cells (IFN-γ+) that was TRIF- and type I interferon receptor (IFNAR)-dependent, and into Tfh-like cells (IL21+) and Tfh1 (IFN-γ+ IL21+) that were TRIF-, MyD88- and IFNAR-dependent. rASP-1-activated BMDCs promoted the differentiation of naïve CD4+ T cells into Th17 (IL-17+) cells only when the MyD88 pathway was inhibited. Importantly, rASP-1-activated human blood cDCs expressed upregulated genes that are associated with DC maturation, type I IFN and type II IFN signaling, as well as TLR4-TRIF dependent signaling. These activated cDCs promoted the differentiation of naïve human CD4+ T cells into Th1, Tfh-like and Th17 cells. Our data thus confirms that the rASP-1 is a potent innate adjuvant that polarizes the adaptive T cell responses to Th1/Tfh1 in both mouse and human DCs. Notably, the rASP-1-adjuvanted IIV3 vaccine elicited protection of mice from a lethal H1N1 infection that is also dependent on the TLR4-TRIF axis and IFNAR signaling pathway, as well as on its ability to induce anti-IIV3 antibody production.
Collapse
Affiliation(s)
- Parakkal Jovvian George
- Laboratory Molecular Parasitology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, United States
| | - Radu Marches
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| | | | - Jacques Banchereau
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| | - Sara Lustigman
- Laboratory Molecular Parasitology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, United States
| |
Collapse
|
26
|
Starshinova A, Malkova А, Kudryavtsev I, Kudlay D, Zinchenko Y, Yablonskiy P. Tuberculosis and autoimmunity: Common features. Tuberculosis (Edinb) 2022; 134:102202. [DOI: 10.1016/j.tube.2022.102202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 03/27/2022] [Indexed: 11/25/2022]
|
27
|
Peña-Romero AC, Orenes-Piñero E. Dual Effect of Immune Cells within Tumour Microenvironment: Pro- and Anti-Tumour Effects and Their Triggers. Cancers (Basel) 2022; 14:1681. [PMID: 35406451 PMCID: PMC8996887 DOI: 10.3390/cancers14071681] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 02/04/2023] Open
Abstract
Our body is constantly exposed to pathogens or external threats, but with the immune response that our body can develop, we can fight off and defeat possible attacks or infections. Nevertheless, sometimes this threat comes from an internal factor. Situations such as the existence of a tumour also cause our immune system (IS) to be put on alert. Indeed, the link between immunology and cancer is evident these days, with IS being used as one of the important targets for treating cancer. Our IS is able to eliminate those abnormal or damaged cells found in our body, preventing the uncontrolled proliferation of tumour cells that can lead to cancer. However, in several cases, tumour cells can escape from the IS. It has been observed that immune cells, the extracellular matrix, blood vessels, fat cells and various molecules could support tumour growth and development. Thus, the developing tumour receives structural support, irrigation and energy, among other resources, making its survival and progression possible. All these components that accompany and help the tumour to survive and to grow are called the tumour microenvironment (TME). Given the importance of its presence in the tumour development process, this review will focus on one of the components of the TME: immune cells. Immune cells can support anti-tumour immune response protecting us against tumour cells; nevertheless, they can also behave as pro-tumoural cells, thus promoting tumour progression and survival. In this review, the anti-tumour and pro-tumour immunity of several immune cells will be discussed. In addition, the TME influence on this dual effect will be also analysed.
Collapse
Affiliation(s)
| | - Esteban Orenes-Piñero
- Department of Biochemistry and Molecular Biology-A, University of Murcia, 30120 Murcia, Spain;
| |
Collapse
|
28
|
Bolouri N, Akhtari M, Farhadi E, Mansouri R, Faezi ST, Jamshidi A, Mahmoudi M. Role of the innate and adaptive immune responses in the pathogenesis of systemic lupus erythematosus. Inflamm Res 2022; 71:537-554. [PMID: 35298669 DOI: 10.1007/s00011-022-01554-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 02/08/2022] [Accepted: 02/16/2022] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE), the most common form of lupus, is a multisystemic rheumatic disease with different clinical features that generally affect women of childbearing age. The common symptoms of SLE are very similar to other autoimmune and non-autoimmune disorders, thereby it is known as a thousand faces disease. In this article, we are going to discuss some of the most updated information about immune system-related factors, cells, and cytokines involved in SLE pathogenesis. METHODS Different electronic databases, especially PubMed/MEDLINE, Scopus, and Google Scholar, were searched to review and analyze relevant literature on the role of innate and adaptive immune cells and cytokines in the pathogenesis of SLE. A search for relevant literature was accomplished using various keywords including systemic lupus erythematosus, apoptosis, autoantibodies, immunopathogenesis of SLE, adaptive and innate immune cells, inflammatory cytokines, hormones, etc. RESULTS AND CONCLUSION: The most important characteristic of SLE is the production of antibodies against different nuclear autoantigens like double-strand DNA and RNA. The depositions of the immune complexes (ICs) that are generated between autoantibodies and autoantigens, along with aberrant clearance of them, can lead to permanent inflammation and contribute to tissue or organ damage. Related mechanisms underlying the initiation and development of SLE have not been clarified yet. Although, defects in immune tolerance, enhanced antigenic load, hyperactivity of T cells, and inappropriate regulation of B cells contribute to the pathogenic autoantibodies generation. Besides, sex hormones that influence the immune system seem to act as triggers or protectors of SLE development.
Collapse
Affiliation(s)
- Nasim Bolouri
- Immunology Department, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Rheumatology Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Akhtari
- Rheumatology Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Inflammation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Farhadi
- Rheumatology Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran. .,Inflammation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Reza Mansouri
- Immunology Department, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyedeh Tahereh Faezi
- Rheumatology Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Jamshidi
- Rheumatology Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Inflammation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Malkova A, Kudryavtsev I, Starshinova A, Kudlay D, Zinchenko Y, Glushkova A, Yablonskiy P, Shoenfeld Y. Post COVID-19 Syndrome in Patients with Asymptomatic/Mild Form. Pathogens 2021; 10:1408. [PMID: 34832564 PMCID: PMC8620929 DOI: 10.3390/pathogens10111408] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 12/30/2022] Open
Abstract
Post COVID-19 Syndrome (PCS) is a complex of various symptoms developing a month or more after the acute phase of the disease. The cases of PCS development among patients with asymptomatic/mild forms are frequently reported; however, the pathogenesis of PCS in this group of patients is still not completely clear. The publications about COVID-19 which were published in online databases from December 2019 to September 2021 are analyzed in this review. According to the analysis, PCS develops on average in 30-60% of patients, mainly among women. Fatigue, shortness of breath, cough, and anosmia were reported as the most common symptoms. The possible association between the described PCS symptoms and brain damage was revealed. We assume the possibility of an alternative course of COVID-19, which develops in genetically predisposed individuals with a stronger immune response, in which it predominantly affects the cells of the nervous system, possibly with the presence of an autoimmune component, which might have similarity with chronic fatigue syndrome or autoimmune disautonomia. Thus, the gender (female) and the presence of anosmia during an asymptomatic or mild course of the disease can be predictive factors for the development of PCS, which can be caused by autoimmune damage to neurons, glia, and cerebral vessels.
Collapse
Affiliation(s)
- Annа Malkova
- Medical Department, St-Petersburg State University, 199034 Saint-Petersburg, Russia; (P.Y.); (Y.S.)
| | - Igor Kudryavtsev
- Department of Immunology, Institution of Experimental Medicine, 197376 Saint-Petersburg, Russia;
| | - Anna Starshinova
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia;
| | - Dmitry Kudlay
- Medical Department, I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia;
- NRC Institute of Immunology FMBA of Russia, 115478 Moscow, Russia
| | - Yulia Zinchenko
- St. Petersburg Research Institute of Phthisiopulmonology, 199034 Saint-Petersburg, Russia;
| | - Anzhela Glushkova
- V.M. Bekhterev National Research Medical Center for Psychiatry and Neurology, 192019 Saint-Petersburg, Russia;
| | - Piotr Yablonskiy
- Medical Department, St-Petersburg State University, 199034 Saint-Petersburg, Russia; (P.Y.); (Y.S.)
- St. Petersburg Research Institute of Phthisiopulmonology, 199034 Saint-Petersburg, Russia;
| | - Yehuda Shoenfeld
- Medical Department, St-Petersburg State University, 199034 Saint-Petersburg, Russia; (P.Y.); (Y.S.)
- Ariel University, Kiryat HaMada 3, Ariel 40700, Israel
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer 5265601, Israel
| |
Collapse
|
30
|
Golovkin A, Kalinina O, Bezrukikh V, Aquino A, Zaikova E, Karonova T, Melnik O, Vasilieva E, Kudryavtsev I. Imbalanced Immune Response of T-Cell and B-Cell Subsets in Patients with Moderate and Severe COVID-19. Viruses 2021; 13:1966. [PMID: 34696395 PMCID: PMC8538447 DOI: 10.3390/v13101966] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 12/12/2022] Open
Abstract
Background: The immunological changes associated with COVID-19 are largely unknown. Methods: Patients with COVID-19 showing moderate (n = 18; SpO2 > 93%, respiratory rate > 22 per minute, CRP > 10 mg/L) and severe (n = 23; SpO2 < 93%, respiratory rate >30 per minute, PaO2/FiO2 ≤ 300 mmHg, permanent oxygen therapy, qSOFA > 2) infection, and 37 healthy donors (HD) were enrolled. Circulating T- and B-cell subsets were analyzed by flow cytometry. Results: CD4+Th cells were skewed toward Th2-like phenotypes within CD45RA+CD62L− (CM) and CD45RA–CD62L− (EM) cells in patients with severe COVID-19, while CM CCR6+ Th17-like cells were decreased if compared with HD. Within CM Th17-like cells “classical” Th17-like cells were increased and Th17.1-like cells were decreased in severe COVID-19 cases. Circulating CM follicular Th-like (Tfh) cells were decreased in all COVID-19 patients, and Tfh17-like cells represented the most predominant subset in severe COVID-19 cases. Both groups of patients showed increased levels of IgD-CD38++ B cells, while the levels of IgD+CD38− and IgD–CD38− were decreased. The frequency of IgD+CD27+ and IgD–CD27+ B cells was significantly reduced in severe COVID-19 cases. Conclusions: We showed an imbalance within almost all circulating memory Th subsets during acute COVID-19 and showed that altered Tfh polarization led to a dysregulated humoral immune response.
Collapse
Affiliation(s)
- Alexey Golovkin
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (O.K.); (V.B.); (A.A.); (E.Z.); (T.K.); (O.M.); (E.V.); (I.K.)
| | - Olga Kalinina
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (O.K.); (V.B.); (A.A.); (E.Z.); (T.K.); (O.M.); (E.V.); (I.K.)
| | - Vadim Bezrukikh
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (O.K.); (V.B.); (A.A.); (E.Z.); (T.K.); (O.M.); (E.V.); (I.K.)
| | - Arthur Aquino
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (O.K.); (V.B.); (A.A.); (E.Z.); (T.K.); (O.M.); (E.V.); (I.K.)
| | - Ekaterina Zaikova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (O.K.); (V.B.); (A.A.); (E.Z.); (T.K.); (O.M.); (E.V.); (I.K.)
| | - Tatyana Karonova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (O.K.); (V.B.); (A.A.); (E.Z.); (T.K.); (O.M.); (E.V.); (I.K.)
| | - Olesya Melnik
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (O.K.); (V.B.); (A.A.); (E.Z.); (T.K.); (O.M.); (E.V.); (I.K.)
| | - Elena Vasilieva
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (O.K.); (V.B.); (A.A.); (E.Z.); (T.K.); (O.M.); (E.V.); (I.K.)
| | - Igor Kudryavtsev
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (O.K.); (V.B.); (A.A.); (E.Z.); (T.K.); (O.M.); (E.V.); (I.K.)
- Institute of Experimental Medicine, 197376 St. Petersburg, Russia
| |
Collapse
|
31
|
Geng L, Zhao J, Deng Y, Molano I, Xu X, Xu L, Ruiz P, Li Q, Feng X, Zhang M, Tan W, Kamen DL, Bae SC, Gilkeson GS, Sun L, Tsao BP. Human SLE variant NCF1-R90H promotes kidney damage and murine lupus through enhanced Tfh2 responses induced by defective efferocytosis of macrophages. Ann Rheum Dis 2021; 81:255-267. [PMID: 34556485 DOI: 10.1136/annrheumdis-2021-220793] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 09/01/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVES We previously identified a hypomorphic variant, p.Arg90His (p.R90H) of neutrophil cytosolic factor 1 (NCF1, a regulatory subunit of phagocyte nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 complex), as an putative causal variant for systemic lupus erythematosus (SLE), and established a knock-in (KI) H90 variant in the C57BL/6 background to study how this variant promotes lupus development. METHODS Wild type (WT) and KI littermates were assessed for immune profiles and lupus-like features. Disease activity and renal damage of patients with SLE were assessed by systemic lupus erythematosus disease activity index (SLEDAI) and renal items of systemic lupus international collaborating clinics (SLICC), respectively. RESULTS Compared with WT littermates, 5-week-old homozygous KI mice had reduced oxidative burst, splenomegaly, elevated type I interferon (IFN-I) scores, increased ratios of splenic follicular T helper 2 (Tfh2) to either T follicular regulatory (Tfr) or Tfh1 cells, increased ANA+ follicular, germinal centre and plasma cells without spontaneous kidney disease up to 1 year of age. Pristane treatment exacerbated the immune dysregulation and induced IFN-I-dependent kidney disease in 36-week-old H90 KI female mice. Decreased efferocytosis of macrophages derived from KI mice and patients with homozygous H90 SLE promoted elevated ratios of Tfh2/Tfr and Tfh2/Tfh1 as well as dysregulated humoral responses due to reduced voltage-gated proton channel 1 (Hv1)-dependent acidification of phagosome pH to neutralise the decreased electrogenic effect of the H90 variant, resulting in impaired maturation and phagosome proteolysis, and increased autoantibody production and kidney damage in mice and patients with SLE of multiple ancestries. CONCLUSIONS A lupus causal variant, NCF1-H90, reduces macrophage efferocytosis, enhances Tfh2 responses and promotes autoantibody production and kidney damage in both mice and patients with SLE.
Collapse
Affiliation(s)
- Linyu Geng
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA.,Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jian Zhao
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Yun Deng
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ivan Molano
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Xue Xu
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Lingxiao Xu
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Phillip Ruiz
- Department of Surgery, University of Miami School of Medicine, Miami, Florida, USA
| | - Quanzhen Li
- Department of Immunology and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Xuebing Feng
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Miaojia Zhang
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenfeng Tan
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Diane L Kamen
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Sang-Cheol Bae
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases and Hanyang University Institute for Rheumatology, Seoul, The Republic of Korea
| | - Gary S Gilkeson
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA.,Ralph H Johnson VA Medical Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Lingyun Sun
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Betty P Tsao
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
32
|
Beurier P, Ricard L, Eshagh D, Malard F, Siblany L, Fain O, Mohty M, Gaugler B, Mekinian A. TFH cells in systemic sclerosis. J Transl Med 2021; 19:375. [PMID: 34461933 PMCID: PMC8407089 DOI: 10.1186/s12967-021-03049-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/20/2021] [Indexed: 12/17/2022] Open
Abstract
Systemic sclerosis is an autoimmune disease characterized by excessive dermal fibrosis with progression to internal organs, vascular impairment and immune dysregulation evidenced by the infiltration of inflammatory cells in affected tissues and the production of auto antibodies. While the pathogenesis remains unclear, several data highlight that T and B cells deregulation is implicated in the disease pathogenesis. Over the last decade, aberrant responses of circulating T follicular helper cells, a subset of CD4 T cells which are able to localise predominantly in the B cell follicles through a high level of chemokine receptor CXCR5 expression are described in pathogenesis of several autoimmune diseases and chronic graft-versus-host-disease. In the present review, we summarized the observed alteration of number and frequency of circulating T follicular helper cells in systemic sclerosis. We described their role in aberrant B cell activation and differentiation though interleukine-21 secretion. We also clarified T follicular helper-like cells involvement in fibrogenesis in both human and mouse model. Finally, because T follicular helper cells are involved in both fibrosis and autoimmune abnormalities in systemic sclerosis patients, we presented the different strategies could be used to target T follicular helper cells in systemic sclerosis, the therapeutic trials currently being carried out and the future perspectives from other auto-immune diseases and graft-versus-host-disease models.
Collapse
Affiliation(s)
- Pauline Beurier
- INSERM UMRs 938, Centre de Recherche Saint-Antoine, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Sorbonne Université, 75012, Paris, France.,Sorbonne Université, Paris, France
| | - Laure Ricard
- INSERM UMRs 938, Centre de Recherche Saint-Antoine, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Sorbonne Université, 75012, Paris, France.,Sorbonne Université, Paris, France.,Service D'Hématologie Clinique, AP-HP, Hôpital Saint-Antoine, 75012, Paris, France
| | - Deborah Eshagh
- INSERM UMRs 938, Centre de Recherche Saint-Antoine, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Sorbonne Université, 75012, Paris, France.,Sorbonne Université, Paris, France
| | - Florent Malard
- INSERM UMRs 938, Centre de Recherche Saint-Antoine, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Sorbonne Université, 75012, Paris, France.,Sorbonne Université, Paris, France.,Service D'Hématologie Clinique, AP-HP, Hôpital Saint-Antoine, 75012, Paris, France
| | - Lama Siblany
- INSERM UMRs 938, Centre de Recherche Saint-Antoine, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Sorbonne Université, 75012, Paris, France.,Sorbonne Université, Paris, France
| | - Olivier Fain
- INSERM UMRs 938, Centre de Recherche Saint-Antoine, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Sorbonne Université, 75012, Paris, France.,Sorbonne Université, Paris, France
| | - Mohamad Mohty
- INSERM UMRs 938, Centre de Recherche Saint-Antoine, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Sorbonne Université, 75012, Paris, France.,Sorbonne Université, Paris, France.,Service D'Hématologie Clinique, AP-HP, Hôpital Saint-Antoine, 75012, Paris, France
| | - Béatrice Gaugler
- INSERM UMRs 938, Centre de Recherche Saint-Antoine, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Sorbonne Université, 75012, Paris, France
| | - Arsène Mekinian
- INSERM UMRs 938, Centre de Recherche Saint-Antoine, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Sorbonne Université, 75012, Paris, France. .,Sorbonne Université, Paris, France. .,Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DMU 3iD), AP-HP, Hôpital Saint-Antoine, 75012, Paris, France.
| |
Collapse
|
33
|
Bemark M, Angeletti D. Know your enemy or find your friend?-Induction of IgA at mucosal surfaces. Immunol Rev 2021; 303:83-102. [PMID: 34331314 PMCID: PMC7612940 DOI: 10.1111/imr.13014] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/15/2022]
Abstract
Most antibodies produced in the body are of the IgA class. The dominant cell population producing them are plasma cells within the lamina propria of the gastrointestinal tract, but many IgA-producing cells are also found in the airways, within mammary tissues, the urogenital tract and inside the bone marrow. Most IgA antibodies are transported into the lumen by epithelial cells as part of the mucosal secretions, but they are also present in serum and other body fluids. A large part of the commensal microbiota in the gut is covered with IgA antibodies, and it has been demonstrated that this plays a role in maintaining a healthy balance between the host and the bacteria. However, IgA antibodies also play important roles in neutralizing pathogens in the gastrointestinal tract and the upper airways. The distinction between the two roles of IgA - protective and balance-maintaining - not only has implications on function but also on how the production is regulated. Here, we discuss these issues with a special focus on gut and airways.
Collapse
Affiliation(s)
- Mats Bemark
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Immunology and Transfusion Medicine, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Davide Angeletti
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
34
|
Shen C, Xue X, Zhang X, Wu L, Duan X, Su C. Dexamethasone reduces autoantibody levels in MRL/lpr mice by inhibiting Tfh cell responses. J Cell Mol Med 2021; 25:8329-8337. [PMID: 34318604 PMCID: PMC8419171 DOI: 10.1111/jcmm.16785] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/15/2021] [Accepted: 06/25/2021] [Indexed: 12/14/2022] Open
Abstract
Previous studies have shown that dexamethasone (Dex) reduces the levels of anti‐nuclear (ANA) and anti‐dsDNA antibodies in MRL/lpr mice (a mouse model of SLE). However, the effect of Dex on T follicular helper (Tfh) cells is less documented. Here, using the MRL/lpr mouse model, we investigated the influence of Dex on Tfh cells and potential underlying mechanisms. The data showed that the proportion of Tfh cells, identified as CD4+CXCR5+ICOS+, CD4+CXCR5+PD‐1+ or CD4+BCL‐6+ cells, markedly decreased after treatment with the Dex, in both Balb/c mice and MRL/lpr mice. Dex significantly inhibited IL‐21 expression at both the mRNA and the protein levels. Dex also significantly reduced the proportion of germinal centre B cells and decreased serum IgG, IgG2a/b and IgA levels. Moreover, a positive correlation between the proportion of Tfh cells (CD4+CXCR5+ICOS+, CD4+CXCR5+PD‐1+ or CD4+BCL‐6+) and autoantibodies was observed. Dex significantly increased the Prdm1 and Stat5b mRNA expression and decreased the Bcl‐6 and c‐Maf mRNA expression of CD4+T cells. In brief, Dex inhibited the Tfh development, which relies on many other transcription factors in addition to Bcl‐6. Our data indicate that Dex can be used as a Tfh cell inhibitor in SLE.
Collapse
Affiliation(s)
- Chunxiu Shen
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Xiaonan Xue
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Xiaoyu Zhang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Lihua Wu
- Department of Nephrology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xiangguo Duan
- Department of Laboratory Surgery, General Hospital of Ningxia Medical University, Yinchuan, China.,College of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Chunxia Su
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
35
|
Kurata I, Mikami N, Ohyama A, Osada A, Kondo Y, Tsuboi H, Sumida T, Matsumoto I. Impaired function of PD-1 + follicular regulatory T cells in systemic lupus erythematosus. Clin Exp Immunol 2021; 206:28-35. [PMID: 34240405 DOI: 10.1111/cei.13643] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/25/2021] [Accepted: 07/02/2021] [Indexed: 12/01/2022] Open
Abstract
Aberrant autoantibody production is characteristic of systemic lupus erythematosus (SLE), but follicular regulatory T (TFR) cells can potentially suppress this abnormality. We investigate functional changes in TFR cells from SLE patients. Circulating TFR cells were collected from 19 SLE patients and 14 healthy controls (HC) to compare molecular expression and in-vitro suppressive capacity of follicular helper T (TFH) cell proliferation. To reveal the stability of forkhead box protein 3 (FoxP3) in TFR, pyrosequencing of conserved non-coding sequence (CNS) 2 at the FoxP3 gene locus was performed. We then tested interleukin (IL)-2 in SLE-TFR cells to check restoration of suppressor function. Programmed cell death 1 (PD-1) expression in SLE-TFR cells was positively correlated with anti-DNA antibody levels and disease activity. These cells had impaired suppressive function for TFH cells with decreased expression of suppression mediators FoxP3, cytotoxic T lymphocyte antigen 4 (CTLA-4) and IL-2 receptor alpha (IL-2Rα). Pyrosequencing identified hyper-methylation in CNS2 region of SLE-TFR cells comparing to HC. With in-vitro IL-2 stimulation, PD-1 expression of TFR cells significantly decreased, together with increased expression of FoxP3 and CTLA-4, especially at a low dose. Thus, SLE-TFR cells have functionally defective to TFH suppression, but low-dose IL-2 therapy might be useful to restore this ability.
Collapse
Affiliation(s)
- Izumi Kurata
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba-city, Japan
| | - Natsuko Mikami
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba-city, Japan
| | - Ayako Ohyama
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba-city, Japan
| | - Atsumu Osada
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba-city, Japan
| | - Yuya Kondo
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba-city, Japan
| | - Hiroto Tsuboi
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba-city, Japan
| | - Takayuki Sumida
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba-city, Japan
| | - Isao Matsumoto
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba-city, Japan
| |
Collapse
|
36
|
Ma X, Nakayamada S. Multi-Source Pathways of T Follicular Helper Cell Differentiation. Front Immunol 2021; 12:621105. [PMID: 33717120 PMCID: PMC7947315 DOI: 10.3389/fimmu.2021.621105] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 01/14/2021] [Indexed: 12/31/2022] Open
Abstract
T follicular helper (Tfh) cells participate in humoral immune by promoting inflammation and aiding B cells survival, proliferation, maturation, and generation autoantibodies. The plasticity of Tfh cells enables the immune system to adjust the direction of differentiation according to the degree of the immune response, regulate the germinal center (GC) response and maintain homeostasis. Tfh differentiation involves several signaling factors, including multiple cytokines, receptors, transcription factors and genes. The signal transducer and activator of transcription (STAT) family signaling pathways are crucial for Tfh formation. However, because of the multi-factorial and multi-stage features of Tfh differentiation, every STAT member plays a role in Tfh differentiation, but is not completely depended on. With the gradual recognition of different Tfh subsets (Tfh1, Tfh2, Tfh17), the process of Tfh differentiation can no longer be explained by straight-line derivation models. In this review, we summarize the roles of different STATs in mediating Tfh subsets, analyze the contributions of mutual restraint and cooperation among cytokine-STAT signals to terminal Tfh differentiation, and clarify the multi-source pathways of Tfh differentiation with a three-dimensional illustration.
Collapse
Affiliation(s)
- Xiaoxue Ma
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, China.,Department of Microbiology & Immunology and Pediatrics, Dalhousie University, Halifax, NS, Canada
| | - Shingo Nakayamada
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| |
Collapse
|
37
|
van Beers JJ, Damoiseaux JG. Immune Monitoring upon Treatment with Biologics in Sjögren's Syndrome: The What, Where, When, and How. Biomolecules 2021; 11:116. [PMID: 33467204 PMCID: PMC7830440 DOI: 10.3390/biom11010116] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/14/2021] [Accepted: 01/14/2021] [Indexed: 12/25/2022] Open
Abstract
Over the years, a wide variety of therapeutic antibodies has been successfully introduced in the auto-immunology clinic, and many more are on the way. Many of these treatments address either a pathogenic circulating molecule or a cell-bound molecule. Whereas addressing the former target results in neutralization of the soluble factor and binding to the latter target either inhibits cellular function or induces selective cell death. If this targeted molecule or cell is part of the immune system, this therapy evokes a state of immunodeficiency with infections as a possible consequence. Therefore, immune monitoring is needed to prevent such adverse side effects of immunotherapy. In this paper, different immunotherapies used in Sjögren's syndrome, as well as different approaches to monitoring the immune system, are discussed.
Collapse
Affiliation(s)
- Joyce J.B.C. van Beers
- Central Diagnostic Laboratory Maastricht University Medical Center, Laboratory Specialist in Medical Immunology and Clinical Chemistry, 6202 AZ Maastricht, The Netherlands
| | - Jan G.M.C. Damoiseaux
- Central Diagnostic Laboratory Maastricht University Medical Center, Laboratory Specialist in Medical Immunology, 6202 AZ Maastricht, The Netherlands;
| |
Collapse
|
38
|
Barcelos F, Martins C, Madeira N, Ângelo-Dias M, Cardigos J, Alves N, Vaz-Patto J, Cunha-Branco J, Borrego LM. Lymphocyte subpopulations in Sjögren's syndrome are distinct in anti-SSA-positive patients and related to disease activity. Clin Rheumatol 2021; 40:2791-2804. [PMID: 33443605 DOI: 10.1007/s10067-020-05537-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/28/2020] [Accepted: 12/02/2020] [Indexed: 12/28/2022]
Abstract
OBJECTIVES Sjögren's syndrome (SjS) patients exhibit great phenotypical heterogeneity, reinforced by the positiveness of anti-SSA antibody. We aimed to evaluate lymphocyte subpopulations in SSA-positive (SSA+SjS) and SSA-negative (SSA-SjS) SjS patients, Sicca patients, and healthy controls (HC), and to investigate associations between lymphocyte subpopulations and disease activity in SjS. METHODS According to the fulfilment of the ACR/EULAR 2016 classification criteria, patients were included as SjS or as Sicca. HC were selected from the Ophthalmology outpatient clinic. Lymphocyte subpopulations were characterized by flow cytometry. Statistical analysis was performed with GraphPad PrismTM, with statistical significance concluded if p < 0.05. RESULTS We included 53 SjS patients (38 SSA+ and 15 SSA-), 72 Sicca, and 24 HC. SSA+SjS patients presented increased IL-21+CD4+ and CD8+ T cells compared to Sicca and HC, whereas compared to SSA-SjS patients, only IL-21+CD4+ T cell percentages were increased and Tfh17 percentages and numbers were decreased. Compared to Sicca and HC, SSA+SjS patients had higher levels of CD24HiCD38Hi B cells, naïve B cells, and IgM-/+CD38++ plasmablasts, and lower levels of memory B cells, including CD24HiCD27+ B cells. SSA+SjS patients with clinically active disease had positive correlations between ESSDAI and IL-21+CD4+ (p = 0.038, r = 0.456) and IL-21+CD8+ T cells (p = 0.046, r = 0.451). CONCLUSIONS In SjS, a distinct lymphocyte subset distribution profile seems to be associated with positive anti-SSA. Moreover, the association between ESSDAI and IL-21+CD4+ and IL-21+CD8+ (follicular) T cells in SSA+SjS patients suggests the involvement of these cells in disease pathogenesis and activity, and possibly their utility for the prognosis and assessment of response to therapy. Key Points • SSA+SjS patients have a pronounced naïve/memory B cell imbalance. • SSA+SjS patients have more active disease associated with IL-21+CD4+ and IL-21+CD8+ follicular T cell expansion. • IL-21+CD4+ and IL-21+CD8+ T cell quantification may be useful for the prognosis and assessment of response to therapy.
Collapse
Affiliation(s)
- Filipe Barcelos
- CEDOC, Chronic Diseases Research Center, Immunology, NOVA Medical School
- FCM, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056, Lisbon, Portugal. .,Comprehensive Health Research Centre (CHRC), NOVA Medical School / FCM, Universidade Nova de Lisboa, Lisbon, Portugal. .,Rheumatology Department, Instituto Português de Reumatologia, Lisbon, Portugal. .,Rheumatology Department, Hospital CUF Descobertas, Lisbon, Portugal.
| | - Catarina Martins
- CEDOC, Chronic Diseases Research Center, Immunology, NOVA Medical School
- FCM, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056, Lisbon, Portugal.,Comprehensive Health Research Centre (CHRC), NOVA Medical School / FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Nathalie Madeira
- Rheumatology Department, Instituto Português de Reumatologia, Lisbon, Portugal
| | - Miguel Ângelo-Dias
- CEDOC, Chronic Diseases Research Center, Immunology, NOVA Medical School
- FCM, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056, Lisbon, Portugal
| | - Joana Cardigos
- Ophthalmology Department, Centro Hospitalar de Lisboa Central, Hospital de Santo António dos Capuchos, Lisbon, Portugal
| | - Nuno Alves
- Ophthalmology Department, Centro Hospitalar de Lisboa Central, Hospital de Santo António dos Capuchos, Lisbon, Portugal.,Ophthalmology Department, Hospital CUF Descobertas, Lisbon, Portugal
| | - José Vaz-Patto
- Rheumatology Department, Instituto Português de Reumatologia, Lisbon, Portugal
| | - Jaime Cunha-Branco
- CEDOC, Chronic Diseases Research Center, Immunology, NOVA Medical School
- FCM, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056, Lisbon, Portugal.,Comprehensive Health Research Centre (CHRC), NOVA Medical School / FCM, Universidade Nova de Lisboa, Lisbon, Portugal.,Rheumatology Department, Hospital CUF Descobertas, Lisbon, Portugal.,Rheumatology Department, Centro Hospitalar de Lisboa Ocidental, Hospital de Egas Moniz, Lisbon, Portugal
| | - Luís-Miguel Borrego
- CEDOC, Chronic Diseases Research Center, Immunology, NOVA Medical School
- FCM, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056, Lisbon, Portugal.,Comprehensive Health Research Centre (CHRC), NOVA Medical School / FCM, Universidade Nova de Lisboa, Lisbon, Portugal.,Immunoalergy Department, Hospital da Luz Lisboa, Lisbon, Portugal
| |
Collapse
|
39
|
Foley CL, Al Remeithi SS, Towe CT, Dauber A, Backeljauw PF, Tyzinski L, Kumar AR, Hwa V. Developmental Adaptive Immune Defects Associated with STAT5B Deficiency in Three Young Siblings. J Clin Immunol 2021; 41:136-146. [PMID: 33090292 PMCID: PMC7854992 DOI: 10.1007/s10875-020-00884-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/02/2020] [Indexed: 01/06/2023]
Abstract
Patients with rare homozygous mutations in signal transducer and activator of transcription 5B (STAT5B) develop immunodeficiency resulting in chronic eczema, chronic infections, autoimmunity, and chronic lung disease. STAT5B-deficient patients are typically diagnosed in the teenage years, limiting our understanding of the development of associated phenotypic immune abnormalities. We report the first detailed chronological account of post-natal immune dysfunction associated with STAT5B deficiency in humans. Annual immunophenotyping of three siblings carrying a novel homozygous nonsense mutation in STAT5B was carried out over 4 years between the ages of 7 months to 8 years. All three siblings demonstrated consistent B cell hyperactivity including elevated IgE levels and autoantibody production, associated with diagnoses of atopy and autoimmunity. Total T cell levels in each sibling remained normal, with regulatory T cells decreasing in the oldest sibling. Interestingly, a skewing toward memory T cells was identified, with the greatest changes in CD8+ effector memory T cells. These results suggest an importance of STAT5B in B cell function and naïve versus memory T cell survival. Progressive dysregulation of FOXP3+ regulatory T cells and CD8+ memory T cell subsets reveal a crucial role of STAT5B in T cell homeostasis. The early diagnosis and focused immune evaluations of these three young STAT5B-deficient siblings support an important role of STAT5B in adaptive immune development and function.
Collapse
Affiliation(s)
- Corinne L Foley
- Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Cincinnati Center for Growth Disorders, Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sareea S Al Remeithi
- Division of Endocrinology, Department of PediatricsSheikh Khalifa Medical City, Abu Dhabi, UAE
| | - Christopher T Towe
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Andrew Dauber
- Division of Endocrinology, Children's National Hospital, Department of Pediatrics, George Washington School of Medicine and Health Sciences, Washington, DC, USA
| | - Philippe F Backeljauw
- Cincinnati Center for Growth Disorders, Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Leah Tyzinski
- Cincinnati Center for Growth Disorders, Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ashish R Kumar
- Bone Marrow Transplantation & Immune Deficiency, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Vivian Hwa
- Cincinnati Center for Growth Disorders, Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|