Cancer Letters

Cancer Letters

Volume 427, 28 July 2018, Pages 63-73
Cancer Letters

Mini-review
Control of T cell effector functions by miRNAs

https://doi.org/10.1016/j.canlet.2018.04.011Get rights and content

Highlights

  • miRNAs as key post-transcriptional regulators of effector T cell differentiation.

  • miRNAs that act as regulators of CD4 T cell- (Th1, Th2, Th7, Tfh and Treg cells), CD8+ T cell- and NKT cell- differentiation.

  • Physiological relevance of miRNAs that affect simultaneously two or more effector T cell populations.

Abstract

The differentiation of effector T cells is a tightly regulated process that relies on the selective expression of lineage-defining master regulators that orchestrate unique transcriptional programs, including the production of distinct sets of effector cytokines. miRNAs are post-transcriptional regulators that are now viewed as critical players in these gene expression networks and help defining cell identity and function. This review summarises the role of individual miRNAs in the regulation of the differentiation of effector T cell subsets, including CD4+ T helper cells, cytotoxic CD8+ T cells and innate-like NKT cells. Moreover, we refer to miRNAs that have been identified to affect simultaneously two or more effector T cell populations, impacting on the balance between effector T cells in vivo, thus constituting potential biomarkers or targets for therapies aiming at boosting immunity or controlling autoimmunity.

Introduction

T lymphocytes develop in the thymus to acquire a T cell receptor (TCR) that undergoes key selection processes to attest productive somatic rearrangement (positive selection) while avoiding self-reactivity (negative selection). For most T cells, this is the result of molecular interactions between the TCR and peptide-major histocompatibility complex (MHC) complexes, although for a minor subset of Natural Killer (NK)-like, NKT cells, the selecting element is CD1d presenting glycolipids [1]. For conventional T cells, the outcome of the preferential TCR binding to MHC class I or II is the selection into the CD8+ or CD4+ T cell lineages, respectively. CD8+ T cells leave the thymus to become cytotoxic T lymphocytes (CTLs) upon cognate antigen recognition in the periphery. Most CD4+ T cells retain a naïve phenotype in the thymus, and will only differentiate into T helper (Th) cells upon activation in secondary lymphoid organs; however, 5–10% of CD4+ thymocytes undergo commitment to the regulatory T cell lineage, characterized by the expression of the master transcription factor (TF) Foxp3.

The peripheral differentiation of T cells relies on the selective expression of other lineage-defining master regulators that orchestrate unique transcriptional programs allowing the different T cell populations to secrete distinct sets of effector cytokines and other effector molecules [2]. In addition to transcriptional regulation, the establishment and maintenance of each effector T cell subset also involve the expression of epigenetic regulators such as microRNAs (miRNAs) that either promote or inhibit its differentiation and/or function. There has been growing evidence that miRNAs are an integral part of gene expression networks, determining cell identity and function by posttranscriptional repression of target mRNAs, including Th polarization, CD8+ T cell functions and NKT cell differentiation [[3], [4], [5], [6]].

miRNAs are small (∼22 nucleotides) noncoding RNAs which in mammals are transcribed by RNA polymerase II and processed by RNase III proteins Drosha and Dicer, interacting with an Argonaut (AGO) protein to form an effector complex called miRNA-induced silencing complex (miRISC) [7] miRISC is then guided by the miRNA to induce translational repression, deadenylation or decay of a target mRNA recognized by base pairing of the miRNA 5′ end “seed” domain (nucleotides 2 to 7) to usually the 3′ untranslated region (UTR) of the mRNA target [7]. miRNA-deficient T cells generated by specific genetic inactivation of either Drosha, DGCR8 (an essential cofactor of Drosha) or Dicer exhibit reduced proliferation and survival after in vitro stimulation, as well as an increase in effector Th cell differentiation and cytokine production, namely interferon-γ (IFN-γ) production, implying that miRNAs regulate T cell differentiation and might have an effect in the balance between different T cell populations, including, for example, Th1/Th17 versus regulatory T cells (Treg), or Th1 vs Th2 differentiation [[8], [9], [10]]. Ultimately, the balance between distinct T cell subsets impacts on the regulation of inflammation and autoimmunity. For example, whereas IFN-γ-producing Th1 and interleukin (IL)-17-producing Th17 are pro-inflammatory T cells that can be beneficial in a context of infection but detrimental in chronic inflammation and autoimmune disease [11], the anti-inflammatory functions of Treg cells have a protective role in chronic inflammation and autoimmunity [12]. Other T cell subsets, such as follicular helper T cells (Tfh) cells, are critical determinants of antigen-specific B cell immunity [13], whereas CD8+ T cells provide cellular immunity against intracellular pathogens and tumours [14]. It is thus of major importance to understand the miRNA-based mechanisms of gene regulation that underlie the differentiation, maintenance and functional plasticity of T cells, which may contribute to the development of novel diagnostic tools and immune therapies.

Several miRNAs, such as miR-181a and miR-214 have been implicated, respectively, in regulating thymic selection by T cells and in promoting T cell activation paying a general role in T cell function overall and acting upstream of T cell differentiation [[15], [16], [17], [18]]. Here we provide an updated [6,19,20], extended and integrated review on the multifaceted impact of miRNAs on T cell differentiation, including Th1, Th2, Th17, Treg, Tfh, CD8+ and NKT cells, and the balance between different effector or regulatory cell subtypes, thus highlighting the potential of using miRNA-based strategies to boost immunity or control autoimmunity.

Section snippets

Th1 cell differentiation and function

Th1 cells are CD4+ T cells characterized by the production of IFN-γ, having an important role in the host defense against intracellular pathogens and tumours, although dysregulated Th1 responses can promote autoimmunity [21]. The differentiation of Th1 cells is initiated by the IFN-γ activation of signal transducer and activator of transcription (STAT)1 signaling, resulting in the expression of the TF T-bet, which in turn induces the production of IFNγ and runt-related transcription factor 3

Th2 cell activities

Th2 cells differentiation from activated CD4+ T cells is driven by IL-4, which signals through the TF STAT6, which in turn upregulates GATA3, collaborating with STAT5 to drive the production of key Th2 cell lineage-defining cytokines, IL-4, IL-5 and IL-13 [34]. By supporting basophil, mast cell, and eosinophil survival, Th2 cells promote protective immune responses, including control of parasitic infections, but also contribute to chronic inflammatory diseases, such as allergy and asthma [35].

Th17 cell differentiation and functions

Th17 cells are characterized by the production of IL-17 A, IL-17 F, IL-21, IL-22 and, in humans, IL-26, and play a key role in the host defence against opportunistic fungi or extracellular bacteria. On the other hand, Th17 cells play a pathogenic role in various inflammatory disorders, such as psoriasis, IBD and MS [21]. Th17 cell differentiation is initiated by IL-6 and transforming growth factor (TGF)-β, leading to RAR-related orphan receptor (ROR)γt expression, the former through STAT3

Treg cell function

Treg cells supress effector T cell responses, limiting inflammation and preventing autoimmunity. They are characterized by the expression of the TF Foxp3, and cell-surface maker CD25, and the immunosuppressive cytokines TGF-β and IL-10. Treg develop in the thymus and emigrate to the periphery where they are critical to prevent tissue autoimmunity, as highlighted by the phenotype of IPEX (immune deregulation polyendocrinopathy enteropathy X-linked) patients [2,56]. However, Treg can also

Tfh cell differentiation and functions

Follicular helper T (Tfh) cells have been recently identified as a subset of effector helper T cells, characterized by expression of CXCR5 (often co-expressed with PD-1) that are necessary for initiation and maintenance of the germinal center (GC) reaction, being thus crucial for the development of antigen-specific B cell immunity [13]. BCL6, a transcriptional repressor, promotes the differentiation of Tfh cells, whereas BLIMP-1 antagonizes BCL6 and enforces the differentiation into non-Tfh

CD8+ T cell differentiation and functions

CD8+ T cells are essential players of adaptive immunity, contributing to host defence against intracellular pathogens and tumours [14]. After antigen recognition, activated CD8+ T cells undergo proliferative expansion and differentiate into cytotoxic T lymphocytes (CTLs) that are able to produce effector molecules, including IFN-γ and cytotoxic molecules such as perforin and GZMB [75] and kill target cells.

Concomitant with major changes in transcriptome and genetic remodelling of signature

miRNA-mediated regulation of NKT cell function

NKT cells are thymus-derived, innate-like T lymphocytes that recognize glycolipid antigens presented by the CD1d, a non-classical MHC class Ib molecule [92]. Invariant NKT (iNKT) cells are the major subset of NKT cells and express TCRs composed by a restricted set of β chains and an invariant V14/Jα18 chain in mice or V24/Jα18 in humans [93,94]. iNKT cells serve an important role in early host defence against invading pathogens by rapidly secreting effector cytokines such as IL-4 and IFN-γ that

Physiological relevance of miRNAs regulating multiple effector T cell populations

As described in the previous sections, miRNAs play an essential role as regulators of effector T cell functions, either by regulating relevant TFs implicated in their development or differentiation or the effector cytokines themselves. In addition to miR-150 and miR-155, which play pleiotropic roles in immune cells, several miRNAs have been identified that affect simultaneously two or more effector T cell populations. These miRNAs might contribute to the plasticity and balance between different

Concluding remarks

miRNA-mediated regulation of T cell differentiation is essential to the homeostasis of the immune system. By targeting multiple critical genes, some miRNAs can individually regulate several cellular processes or signaling pathways, thus affecting the differentiation of various T cell subsets in different tissues. However, it is also a reality that several miRNAs are tissue/cell type specific, acting synergistically to produce a given phenotype, with expression patterns reflecting underlying

Conflicts of interest

The authors declared no competing interests.

Acknowledgments

We thank our past colleagues, Nina Schmolka and Paula Vargas Romero, for discussions on this topic. Our research was funded by Fundação para a Ciência e a Tecnologia (PTDC/BEX-BCM/3592/2014 to A.Q.G.); and European Research Council (CoG_646701 to B.S.-S.). This publication was also funded by LISBOA-01-0145-FEDER-007391, project cofunded by FEDER, through POR Lisboa 2020 - Programa Operacional Regional de Lisboa, PORTUGAL 2020, and Fundação para a Ciência e a Tecnologia.

References (119)

  • M. Xu et al.

    MiR-155 contributes to Th17 cells differentiation in dextran sulfate sodium (DSS)-induced colitis mice via Jarid2

    Biochem. Biophys. Res. Commun.

    (2017)
  • X. Yang et al.

    MicroRNA-425 facilitates pathogenic Th17 cell differentiation by targeting forkhead box O1 (Foxo1) and is associated with inflammatory bowel disease

    Biochem. Biophys. Res. Commun.

    (2018)
  • L.F. Lu et al.

    Foxp3-dependent microRNA155 confers competitive fitness to regulatory T cells by targeting SOCS1 protein

    Immunity

    (2009)
  • H.Y. Yang et al.

    MicroRNA-17 modulates regulatory T cell function by targeting Co-regulators of the Foxp3 transcription factor

    Immunity

    (2016)
  • S. Kuchen et al.

    Regulation of microRNA expression and abundance during lymphopoiesis

    Immunity

    (2010)
  • R. Hu et al.

    miR-155 promotes T follicular helper cell accumulation during chronic, low-grade inflammation

    Immunity

    (2014)
  • T.B. Huffaker et al.

    Epistasis between microRNAs 155 and 146a during T cell-mediated antitumor immunity

    Cell Reports

    (2012)
  • J.C. Dudda et al.

    MicroRNA-155 is required for effector CD8+ T cell responses to virus infection and cancer

    Immunity

    (2013)
  • Z. Chen et al.

    miR-150 regulates memory CD8 T cell differentiation via c-Myb

    Cell Reports

    (2017)
  • A.A. Khan et al.

    MicroRNA-17∼92 regulates effector and memory CD8 T-cell fates by modulating proliferation in response to infections

    Blood

    (2013)
  • D.I. Godfrey et al.

    NKT cells: facts, functions and fallacies

    Immunol. Today

    (2000)
  • J. Henao-Mejia et al.

    The microRNA miR-181 is a critical cellular metabolic rheostat essential for NKT cell ontogenesis and lymphocyte development and homeostasis

    Immunity

    (2013)
  • D.G. Wei et al.

    Expansion and long-range differentiation of the NKT cell lineage in mice expressing CD1d exclusively on cortical thymocytes

    J. Exp. Med.

    (2005)
  • J. Zhu et al.

    Differentiation of effector CD4 T cell populations

    Annu. Rev. Immunol.

    (2010)
  • D. Baumjohann et al.

    The microRNA cluster miR-17 approximately 92 promotes TFH cell differentiation and represses subset-inappropriate gene expression

    Nat. Immunol.

    (2013)
  • Y. Liang et al.

    microRNAs function in CD8+T cell biology

    J. Leukoc. Biol.

    (2015)
  • L. Zhou et al.

    MicroRNAs are key regulators controlling iNKT and regulatory T-cell development and function

    Cell. Mol. Immunol.

    (2011)
  • L.T. Jeker et al.

    MicroRNA regulation of T-cell differentiation and function

    Immunol. Rev.

    (2013)
  • M. Ha et al.

    Regulation of microRNA biogenesis, Nature reviews

    Molecular cell biology

    (2014)
  • M.M. Chong et al.

    The RNAseIII enzyme Drosha is critical in T cells for preventing lethal inflammatory disease

    J. Exp. Med.

    (2008)
  • S.A. Muljo et al.

    Aberrant T cell differentiation in the absence of Dicer

    J. Exp. Med.

    (2005)
  • A. Jager et al.

    Effector and regulatory T-cell subsets in autoimmunity and tissue inflammation

    Scand. J. Immunol.

    (2010)
  • S. Sakaguchi et al.

    Dynamics of peripheral tolerance and immune regulation mediated by Treg

    Eur. J. Immunol.

    (2009)
  • S. Crotty

    Follicular helper CD4 T cells (TFH)

    Annu. Rev. Immunol.

    (2011)
  • P.J. Ebert et al.

    An endogenous positively selecting peptide enhances mature T cell responses and becomes an autoantigen in the absence of microRNA miR-181a

    Nat. Immunol.

    (2009)
  • P.T. Jindra et al.

    Costimulation-dependent expression of microRNA-214 increases the ability of T cells to proliferate by targeting Pten

    J. Immunol.

    (2010)
  • D. Baumjohann et al.

    MicroRNA-mediated regulation of T helper cell differentiation and plasticity, Nature reviews

    Immunology

    (2013)
  • T. Amado et al.

    Cross-regulation between cytokine and microRNA pathways in T cells

    Eur. J. Immunol.

    (2015)
  • C.B. Wilson et al.

    Epigenetic control of T-helper-cell differentiation, Nature reviews

    Immunology

    (2009)
  • K.M. Smith et al.

    miR-29ab1 deficiency identifies a negative feedback loop controlling Th1 bias that is dysregulated in multiple sclerosis

    J. Immunol.

    (2012)
  • F. Ma et al.

    The microRNA miR-29 controls innate and adaptive immune responses to intracellular bacterial infection by targeting interferon-gamma

    Nat. Immunol.

    (2011)
  • H. Guan et al.

    Inverse correlation of expression of microRNA-140-5p with progression of multiple sclerosis and differentiation of encephalitogenic T helper type 1 cells

    Immunology

    (2016)
  • P. Mohnle et al.

    MicroRNA-146a controls Th1-cell differentiation of human CD4+ T lymphocytes by targeting PRKCepsilon

    Eur. J. Immunol.

    (2015)
  • W. Wu et al.

    miR-10a inhibits dendritic cell activation and Th1/Th17 cell immune responses in IBD

    Gut

    (2015)
  • T. Satoorian et al.

    MicroRNA223 promotes pathogenic T-cell development and autoimmune inflammation in central nervous system in mice

    Immunology

    (2016)
  • A. Banerjee et al.

    Micro-RNA-155 inhibits IFN-gamma signaling in CD4+ T cells

    Eur. J. Immunol.

    (2010)
  • T. Wu et al.

    Cutting edge: miR-17-92 is required for both CD4 Th1 and T Follicular helper cell responses during viral infection

    J. Immunol.

    (2015)
  • S. Cho et al.

    A novel miR-24-TCF1 Axis in modulating effector T cell responses

    J. Immunol.

    (2017)
  • K.M. Ansel et al.

    Regulation of Th2 differentiation and Il4 locus accessibility

    Annu. Rev. Immunol.

    (2006)
  • J.V. Fahy

    Type 2 inflammation in asthma–present in most, absent in many, Nature reviews

    Immunology

    (2015)
  • Cited by (18)

    • MicroRNAs: Key modulators of inflammation-associated diseases

      2024, Seminars in Cell and Developmental Biology
    • Discordant immune response among treatment experienced patients infected with HIV-1: Crosstalk between MiRNAs expression and CD4+ T cells count

      2023, International Immunopharmacology
      Citation Excerpt :

      MiR-155 expression is rapidly induced by activation of T cells, and by directly targeting active CD4T cells that differentiate into Th1 cells, it reduces interferon-gamma. Thus, miR-155 may be responsible for the decreased expression of interferon-gamma receptors in CD4 T cells differentiating into Th1 [9]. MiR-155 has also other roles in mammalian safety for example, during thymic differentiation, increased miR-155 expression stimulates the proliferative potential of T regulatory cells by targeting SOCS1 (Suppressor of cytokine signaling 1).

    • Diabetes, infection risk and COVID-19

      2020, Molecular Metabolism
      Citation Excerpt :

      Thus, longitudinal studies might help clarify the discrepancies. Multiple studies have demonstrated that T2D is associated with overactivated T cells and the activation of inflammatory pathways [48–51]. Low-grade chronic inflammation in individuals with either T1D or T2D has been described [24,50,51].

    View all citing articles on Scopus
    View full text