Elsevier

Cytokine

Volume 74, Issue 1, July 2015, Pages 5-17
Cytokine

Review Article
T cell subsets and their signature cytokines in autoimmune and inflammatory diseases

https://doi.org/10.1016/j.cyto.2014.09.011Get rights and content

Highlights

  • T helper (Th) cells provide host defense, but can also promote autoimmune diseases.

  • The original description of Th subsets considered Th1 and Th2 cells.

  • New Th subsets have since been described including Th17, Th22, Th9, and Treg cells.

  • Th subsets have been defined based on their “signature” cytokine profiles.

  • New models of Th subset biology may have to incorporate T cell subset plasticity.

Abstract

CD4+ T helper (Th) cells are critical for proper immune cell homeostasis and host defense, but are also major contributors to pathology of autoimmune and inflammatory diseases. Since the discovery of the Th1/Th2 dichotomy, many additional Th subsets were discovered, each with a unique cytokine profile, functional properties, and presumed role in autoimmune tissue pathology. This includes Th1, Th2, Th17, Th22, Th9, and Treg cells which are characterized by specific cytokine profiles. Cytokines produced by these Th subsets play a critical role in immune cell differentiation, effector subset commitment, and in directing the effector response. Cytokines are often categorized into proinflammatory and anti-inflammatory cytokines and linked to Th subsets expressing them. This article reviews the different Th subsets in terms of cytokine profiles, how these cytokines influence and shape the immune response, and their relative roles in promoting pathology in autoimmune and inflammatory diseases. Furthermore, we will discuss whether Th cell pathogenicity can be defined solely based on their cytokine profiles and whether rigid definition of a Th cell subset by its cytokine profile is helpful.

Introduction

T helper (Th) cells are characterized by different cytokine profiles which are used to define their subsets. However, it is still an area of debate if pathogenic Th cells can be defined by simple cytokine profiles.

Over a quarter of a century ago, Mossman and Coffman made the seminal observation that long-term Th clones could be distinguished based on their cytokine profiles, which afforded Th subsets different functional properties [1]: Th1 cells, characterized by the secretion of Interferon-gamma (IFN-γ) and tumor necrosis factor alpha (TNF), and Th2 cells, which secrete interleukin (IL)-4, IL-5 and IL-13. This observation was novel and advanced the understanding of how the immune system adapts to specific pathogens and that Th subsets have unique roles in mediating protection. For example Th1 cells are responsible for cell-mediated immune responses, while Th2 are responsible for humoral-mediated immunity [2]. Interestingly, each of these Th subsets can promote immunopathology; for instance an excessive Th1 response will result in tissue damage, while excessive Th2 responses can result in atopy/hypersensitivity [2].

Since the discovery of the Th1/Th2 dichotomy, many additional Th subsets were discovered, each one with a unique cytokine profile, functional properties and presumed roles in autoimmune tissue pathology. These “new” Th subsets include IL-17 producing Th17 cells, regulatory Th cells (Tregs), and, recently, IL-9 producing Th9 cells and IL-22 producing Th22 cells. This article will review the different Th subsets in terms of cytokine profiles, how these cytokines influence and shape the immune response, and their relative roles in promoting pathology in autoimmune and inflammatory diseases. Furthermore, we will discuss whether Th cell pathogenicity can be defined solely based on their cytokine profiles and whether rigid definition of a Th cell subset by its cytokine profile is helpful.

Shown in Fig. 1 is an illustration of the pro-inflammatory and anti-inflammatory functions of the signature cytokines of each T cell subset.

Section snippets

Th1 cells

Th1 cells are the quintessential cell type involved in cell mediated inflammation and delayed-type hypersensitivity reactions. They are thought to be important for immunity to intracellular pathogens. Th1 cells are most often defined by their production of IL-2 and IFN-γ but have been reported to produce a number of cytokines including: TNF, lymphotoxin, and granulocyte-macrophage colony-stimulating factor (GM-CSF). Committed Th1 effectors express the transcription factor T-bet. Factors

Th2 cells

Th2 cells are recognized for their role in host defense against multi-cellular parasites and their involvement in allergies and atopic illnesses. To a large extent, Th2 cells function in epithelial tissues, most notably the intestinal tract and lungs. Perhaps as a direct result, Th2 differentiation and function are intimately regulated by innate and epithelial cell types that inhabit these tissues [51]. Recent work has identified the actions of innate cytokines IL-25, IL-33 and thymic stromal

Th17 cells

The IL-17 family of cytokines comprises potent inflammatory mediators involved in host defense against extracellular bacteria, fungi and other eukaryotic pathogens. IL-17 cytokines have been implicated in a broad spectrum of inflammatory conditions and autoimmune diseases [103]. Currently, there are six known IL-17 family members: IL-17A (commonly referred to as IL-17), IL-17B, IL-17C, IL-17D, IL-17E, and IL-17F [104]. Specialized CD4+ T helper cells (Th17) are the major source of IL-17 and

Th22 cells

Th22 cells are recent siblings of Th17 cells which produce predominantly the cytokine IL-22 and represent a separate Th subset with distinct gene expression and functions, and were initially associated with immunopathology of skin diseases [136], [137], [138]. IL-22 is a member of the IL-10 family of cytokines and it is produced by activated T cells, notably Th17 and Th22 cells, as well as by NK cells and γδ T cells, and it acts primarily on non-immune cells [139]. Recent evidence indicates

Th9 cells

IL-9 was first described as a T cell and mast cell growth factor and it is important in promoting mucus production and activation of mast cells as well as eosinophils [163]. It was initially viewed as a Th2 cell cytokine, however, more recently, IL-9 has been identified in a subset of T cells distinct from Th2 cells, now delineated as Th9 cells [164]. The development of Th9 cells requires the combination of TGF-β (which also promotes Tregs) and IL-4 (known to induce Th2 cells) [165].

Tregs

The concept of a specialized subset of T lymphocytes with suppressive function has been around since the early 1970s [172]. In the mid-1990s a novel subset of Th cells with “regulatory” function was identified and designated Tregs [173]. Tregs were later found to express the signature Foxp3 transcription factor, which is critical for their development, lineage commitment, and regulatory functions [174], [175]. Foxp3 expressing Treg subsets include thymically derived or natural Tregs (nTregs)

Discussion – or the enigma of the “Th1-like” Th17 cell

Here, we have summarized the most recent understanding of CD4+ Th cell subset “signature” cytokines in promoting autoimmune tissue pathology and/or mediating protection. Since a sustained and uncontrolled inflammatory response will be detrimental to the host, it must be self-limiting. As a result it seems that there is a switch point at which anti-inflammatory pathways are activated. Thus, instead of an elusive “pathogenic” T cell subset which secretes harmful cytokines, most Th cell subsets

Concluding remarks

The immune system seems to favor a balance between pathogenic and protective Th cells via dual roles for “subset-specific”, or “signature cytokines”, as well as allowing plasticity for subset differentiation and expression of “signature” cytokine(s) by other Th subsets. The observation that many Th subsets can convert into IFN-γ secreting Th1-like cells illustrates this fact since IFN-γ can be both pathogenic and protective. Targeting cytokines as therapy for autoimmune and/or inflammatory

Acknowledgments

This work was supported by Grants NS081237 (T.N.E.), NS52177 and G12MD007591 (T.G.F.) from the National Institute of Health, and Grants RG3499 and RG3701 from the National Multiple Sclerosis Society (T.G.F.) and a fellowship from the South Texas Center for Emerging Infectious Diseases at the University of Texas at San Antonio (I.R.).

References (227)

  • V. Dardalhon et al.

    Role of Th1 and Th17 cells in organ-specific autoimmunity

    J Autoimmun

    (2008)
  • G. Wildbaum et al.

    Antigen-specific CD25-Foxp3-FN-gamma(high) CD4+ T cells restrain the development of experimental allergic encephalomyelitis by suppressing Th17

    Am J Pathol

    (2010)
  • C. Lubeseder-Martellato

    Guanylate-binding protein-1 expression is selectively induced by inflammatory cytokines and is an activation marker of endothelial cells during inflammatory diseases

    Am J Pathol

    (2002)
  • D. Gomez

    Th1/Th2 cytokines in patients with systemic lupus erythematosus: is tumor necrosis factor alpha protective?

    Semin Arthritis Rheum

    (2004)
  • K. Hirahara

    Mechanisms underlying helper T-cell plasticity: implications for immune-mediated disease

    J Allergy Clin Immunol

    (2013)
  • M. Afanasyeva

    Experimental autoimmune myocarditis in A/J mice is an interleukin-4-dependent disease with a Th2 phenotype

    Am J Pathol

    (2001)
  • M. Rocken et al.

    IL-4-induced immune deviation as antigen-specific therapy for inflammatory autoimmune disease

    Immunol Today

    (1996)
  • V.K. Kuchroo

    B7-1 and B7-2 costimulatory molecules activate differentially the Th1/Th2 developmental pathways: application to autoimmune disease therapy

    Cell

    (1995)
  • E.K. Broberg et al.

    IL-4 is the key regulator in herpes simplex virus-based gene therapy of BALB/c experimental autoimmune encephalomyelitis

    Neurosci Lett

    (2004)
  • G.T. Tran

    IL-5 promotes induction of antigen-specific CD4+CD25+ T regulatory cells that suppress autoimmunity

    Blood

    (2012)
  • A. O’Garra et al.

    CD4+ T-cell subsets in autoimmunity

    Curr Opin Immunol

    (1997)
  • G. Zurawski et al.

    Interleukin 13, an interleukin 4-like cytokine that acts on monocytes and B cells, but not on T cells

    Immunol Today

    (1994)
  • J.B. Bice et al.

    Biologic targeted therapy in allergic asthma

    Ann Allergy Asthma Immunol: Off Publ Am College Allergy Asthma Immunol

    (2014)
  • J.R. Carneiro

    IL-2, IL-5, TNF-alpha and IFN-gamma mRNA expression in epidermal keratinocytes of systemic lupus erythematosus skin lesions

    Clinics

    (2011)
  • D. Cihakova

    Interleukin-13 protects against experimental autoimmune myocarditis by regulating macrophage differentiation

    Am J Pathol

    (2008)
  • H. Ochi

    Increased IL-13 but not IL-5 production by CD4-positive T cells and CD8-positive T cells in multiple sclerosis during relapse phase

    J Neurol Sci

    (2002)
  • T.R. Mosmann et al.

    Two types of murine helper T cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins

    J Immunol

    (1986)
  • A. Berger

    Th1 and Th2 responses: what are they?

    BMJ

    (2000)
  • I. Raphael et al.

    Stability of T-cell lineages in autoimmune diseases

    Expert Rev Clin Immunol

    (2012)
  • S. Skurkovich et al.

    Anticytokine therapy, especially anti-interferon-gamma, as a pathogenetic treatment in TH-1 autoimmune diseases

    Ann NY Acad Sci

    (2005)
  • S.L. Peng et al.

    T-bet regulates IgG class switching and pathogenic autoantibody production

    Proc Natl Acad Sci USA

    (2002)
  • U. Boehm et al.

    Cellular responses to interferon-gamma

    Annu Rev Immunol

    (1997)
  • K. Schroder et al.

    Interferon-gamma: an overview of signals, mechanisms and functions

    J Leukoc Biol

    (2004)
  • H.S. Panitch et al.

    Treatment of multiple sclerosis with gamma interferon: exacerbations associated with activation of the immune system

    Neurology

    (1987)
  • R.R. Voskuhl

    T helper 1 (Th1) functional phenotype of human myelin basic protein-specific T lymphocytes

    Autoimmunity

    (1993)
  • A.H. McDonald et al.

    Antigen-specific inhibition of immune interferon production by suppressor cells of autoimmune encephalomyelitis

    J Immunol

    (1988)
  • N. Ji et al.

    More than just a T-box: the role of T-bet as a possible biomarker and therapeutic target in autoimmune diseases

    Immunotherapy

    (2011)
  • T. Olsson

    Critical influences of the cytokine orchestration on the outcome of myelin antigen-specific T-cell autoimmunity in experimental autoimmune encephalomyelitis and multiple sclerosis

    Immunol Rev

    (1995)
  • D. Balomenos et al.

    Interferon-gamma is required for lupus-like disease and lymphoaccumulation in MRL-lpr mice

    J Clin Investig

    (1998)
  • A. Schwarting et al.

    IFN-gamma receptor signaling is essential for the initiation, acceleration, and destruction of autoimmune kidney disease in MRL-Fas(lpr) mice

    J Immunol

    (1998)
  • C. Haas et al.

    IFN-gamma receptor deletion prevents autoantibody production and glomerulonephritis in lupus-prone (NZB × NZW)F1 mice

    J Immunol

    (1998)
  • O.T. Preble et al.

    Systemic lupus erythematosus: presence in human serum of an unusual acid-labile leukocyte interferon

    Science

    (1982)
  • K.P. Machold et al.

    Interferon-gamma induced exacerbation of systemic lupus erythematosus

    J Rheumatol

    (1990)
  • I.A. Ferber

    Mice with a disrupted IFN-gamma gene are susceptible to the induction of experimental autoimmune encephalomyelitis (EAE)

    J Immunol

    (1996)
  • D.O. Willenborg et al.

    IFN-gamma plays a critical down-regulatory role in the induction and effector phase of myelin oligodendrocyte glycoprotein-induced autoimmune encephalomyelitis

    J Immunol

    (1996)
  • L. Flaishon

    Cutting edge: anti-inflammatory properties of low levels of IFN-gamma

    J Immunol

    (2002)
  • K. Haskins et al.

    Acceleration of diabetes in young NOD mice with a CD4+ islet-specific T cell clone

    Science

    (1990)
  • Y. Refaeli et al.

    Interferon gamma is required for activation-induced death of T lymphocytes

    J Exp Med

    (2002)
  • P. Bernabei

    Interferon-gamma receptor 2 expression as the deciding factor in human T, B, and myeloid cell proliferation or death

    J Leukoc Biol

    (2001)
  • A.L. Mellor et al.

    IDO expression by dendritic cells: tolerance and tryptophan catabolism

    Nat Rev Immunol

    (2004)
  • Cited by (0)

    View full text