Mini review
Regulation of immune cell homeostasis by type I interferons

https://doi.org/10.1016/j.cytogfr.2010.05.002Get rights and content

Abstract

Although initially identified and best characterized for their role in innate antiviral defence, type I interferons (IFN-I) are also known to have an important impact on the adaptive immune response. In part, this is linked to another long-recognised property of IFN-I, namely their ability to modify cellular proliferation and survival. Here, we review the influence of IFN-I on immune cell homeostasis, focusing on their effects on T cells and antigen-presenting cells.

Introduction

The immune system comprises a complex array of cell types that exist in a dynamic equilibrium. Under steady-state conditions, cell numbers remain roughly constant. Thus, while the life span and turnover of different cell types varies, there is an overall balance in which the loss of effete cells is offset by the input of new cells generated either by differentiation from precursors or by proliferation of mature cells. This balance is maintained based on a limiting availability of growth and survival factors that support these cells. Under conditions of immune activation, for example in response to infection, the equilibrium is altered, and lymphocyte numbers can increase dramatically. During such an event, signals for cell proliferation, differentiation and death are regulated to produce a large arsenal of effector lymphocytes that can combat the pathogen. Since the cost of maintaining such elevated cell numbers is high – in terms of energy expenditure and potentially damaging immune inflammation – cell numbers decline rapidly following clearance or control of the pathogen to again reach equilibrium, with cell numbers resembling pre-infection levels. However, important differences from the pre-immune state are apparent post-pathogen clearance. In particular, increased numbers of lymphocytes capable of recognising pathogen antigens persist and contribute to long-lasting immunological memory.

Hence, one key to proper functioning of the immune system is precise control of cell numbers through the regulation of cell production, proliferation and death. Given the crucial importance of rapid changes in cellularity in the face of infection, it is not surprising that immune cell turnover is influenced by factors that signal the presence of pathogens. Here we review the evidence that type I interferons (which include multiple IFN-αs, IFN-β and several closely related genes, hereafter referred to collectively as IFN-I), the production of which increases rapidly in response to both viral and bacterial infection, influence the function of the immune system through the regulation of cellular homeostasis. We focus primarily on two cell types: dendritic cells (DC), which are key antigen-presenting cells involved in the initiation of immune responses, and T cells, key effector and regulatory cells of the response.

Section snippets

Ontogeny of dendritic cells

Dendritic cells are hematopoietic cells that belong to the antigen-presenting cell (APC) family, which also includes B cells and macrophages. DC represent a heterogeneous cohort of cells located in both lymphoid and non-lymphoid tissues at sites that are optimal for capture of foreign antigens and contact with other immune cells. In mice lymphoid tissue DC include two main blood-borne subsets: plasmacytoid DC (pDC) and conventional DC (cDC), with the latter further subdivided into CD8+CD4CD11b

Dendritic cell homeostasis and survival

The homeostasis of steady-state lymphoid organ pDC and cDC in vivo is primarily regulated by Flt3-L which triggers proliferative DC precursors expressing Flt3 in BM, blood and lymphoid organs. In the steady-state, the various DC populations recycle with different kinetics, reflecting their diverse lifespan in vivo. The turnover of DC populations in vivo has typically been assessed using DNA precursors, particularly bromo-deoxyuridine (BrdU), which is incorporated into dividing DC progenitors;

Regulation of dendritic cell homeostasis and lifespan by type I IFN

Evidence from a variety of studies indicates that IFN-I can regulate the lifespan of various cell types. IFN-I not only affect cell growth and division but also influence cell survival. For example, IFN-I have been reported to trigger apoptosis of tumour cells as well as virus-infected cells [31]. IFN-I have also been found to affect the lifespan of non-malignant cells, such as T lymphocytes and other immune cells, thus contributing to the alterations in immune cell homeostasis that occur after

T cell development

T lymphocytes are produced from progenitors through a series of well-described maturation steps in the thymus [48]. The maturation process includes selection of T cells based on the specificity of their T cell antigen receptor (TCR), which in the predominant population of T cells comprises α- and β-chains in its antigen recognition unit (αβTCR). Selection is necessary because the coding sequence (and hence antigen-specificity) of the αβTCR is generated randomly through variable recombination of

T cell homeostasis and memory

Following export from the thymus, T lymphocytes adopt a pattern of migration in which they continuously recirculate between blood and lymph, exiting the blood stream into lymph nodes via high endothelial venules and returning via the thoracic duct. While traversing the secondary lymphoid organs (spleen, LN), T cells are able to scan the surface of APC for the presentation of foreign antigens. Until such time as they encounter antigens for which their TCR have sufficient affinity to trigger

Regulation of T cell homeostasis by IFN-I

IFN-I have been shown to impact T cell homeostasis at multiple levels. Given the key role of IFN-I in the innate response to infection, it is not surprising that these effects relate mainly to changes in T cell dynamics during the course of immune responses. Indeed, IFN-I have been shown to enhance the magnitude of primary and memory CD4+ and CD8+ T cell responses [40], [65], [66], [67], [68], [69].

One important factor in the immune-stimulatory activity of IFN-I is their ability to modify DC

Concluding remarks

In the past two decades, the mechanisms linking the innate response to infection and the generation of adaptive immune responses have become increasingly well understood. IFN-I are key cytokines involved in this process; they are induced rapidly after infection and are critical mediators of innate antiviral defense, yet also have a major impact on T and B cell-mediated immunity. Integral to their role in adaptive immunity is the impact that IFN-I have on DC homeostasis and function. DC are the

Acknowledgements

Supported by grants from the Italian Association for Cancer Research (AIRC) and by the European Community 7th Framework Program.

Fabrizio Mattei graduated from Università “La Sapienza” in Rome and completed his specialization in biotechnologies and oncology at the same university. In 2000, he became a fellow at the Edward Jenner Institute for Vaccine Research in David Tough's laboratory, where he acquired experience in the field of dendritic cell immunology. He is currently a senior researcher at the Istituto Superiore di Sanità in Rome where he has focused on the molecular interactions between dendritic cells and type I

References (107)

  • S. Bedoui et al.

    Characterization of an immediate splenic precursor of CD8+ dendritic cells capable of inducing antiviral T cell responses

    J Immunol

    (2009)
  • F. Ginhoux et al.

    The origin and development of nonlymphoid tissue CD103+ DCs

    J Exp Med

    (2009)
  • M.L. del Rio et al.

    CD103− and CD103+ bronchial lymph node dendritic cells are specialized in presenting and cross-presenting innocuous antigen to CD4+ and CD8+ T cells

    J Immunol

    (2007)
  • L. Wu et al.

    Heterogeneity of thymic dendritic cells

    Semin Immunol

    (2005)
  • M. Merad et al.

    Dendritic cell homeostasis

    Blood

    (2009)
  • K. Liu et al.

    Origin of dendritic cells in peripheral lymphoid organs of mice

    Nat Immunol

    (2007)
  • S.H. Naik et al.

    Development of plasmacytoid and conventional dendritic cell subtypes from single precursor cells derived in vitro and in vivo

    Nat Immunol

    (2007)
  • G.M. del Hoyo et al.

    Characterization of a common precursor population for dendritic cells

    Nature

    (2002)
  • J. Diao et al.

    Characterization of distinct conventional and plasmacytoid dendritic cell-committed precursors in murine bone marrow

    J Immunol

    (2004)
  • K. Liu et al.

    In vivo analysis of dendritic cell development and homeostasis

    Science

    (2009)
  • J. Diao et al.

    In situ replication of immediate dendritic cell (DC) precursors contributes to conventional DC homeostasis in lymphoid tissue

    J Immunol

    (2006)
  • S.H. Naik et al.

    Intrasplenic steady-state dendritic cell precursors that are distinct from monocytes

    Nat Immunol

    (2006)
  • J. Diao et al.

    Antigen transmission by replicating antigen-bearing dendritic cells

    J Immunol

    (2007)
  • K. Kabashima et al.

    Intrinsic lymphotoxin-beta receptor requirement for homeostasis of lymphoid tissue dendritic cells

    Immunity

    (2005)
  • S. Bedoui et al.

    Cross-presentation of viral and self antigens by skin-derived CD103+ dendritic cells

    Nat Immunol

    (2009)
  • L. Wu et al.

    Development of dendritic-cell lineages

    Immunity

    (2007)
  • A.T. Kamath et al.

    The development, maturation, and turnover rate of mouse spleen dendritic cell populations

    J Immunol

    (2000)
  • M. O’Keeffe et al.

    Mouse plasmacytoid cells: long-lived cells, heterogeneous in surface phenotype and function, that differentiate into CD8(+) dendritic cells only after microbial stimulus

    J Exp Med

    (2002)
  • A.T. Kamath et al.

    Developmental kinetics and lifespan of dendritic cells in mouse lymphoid organs

    Blood

    (2002)
  • C. Ruedl et al.

    Anatomical origin of dendritic cells determines their life span in peripheral lymph nodes

    J Immunol

    (2000)
  • M. Chen et al.

    Regulation of the lifespan in dendritic cell subsets

    Mol Immunol

    (2007)
  • D. Hildeman et al.

    Apoptosis and the homeostatic control of immune responses

    Curr Opin Immunol

    (2007)
  • F. Granucci et al.

    Inducible IL-2 production by dendritic cells revealed by global gene expression analysis

    Nat Immunol

    (2001)
  • A. Nopora et al.

    Bcl-2 controls dendritic cell longevity in vivo

    J Immunol

    (2002)
  • W.S. Hou et al.

    A Bcl-2-dependent molecular timer regulates the lifespan and immunogenicity of dendritic cells

    Nat Immunol

    (2004)
  • H. Hon et al.

    Bcl-xL is critical for dendritic cell survival in vivo

    J Immunol

    (2004)
  • M. Chen et al.

    Deficiency of Bim in dendritic cells contributes to overactivation of lymphocytes and autoimmunity

    Blood

    (2007)
  • T. Chino et al.

    Regulation of dendritic cell survival and cytokine production by osteoprotegerin

    J Leukoc Biol

    (2009)
  • K. Hochweller et al.

    Homeostasis of dendritic cells in lymphoid organs is controlled by regulation of their precursors via a feedback loop

    Blood

    (2009)
  • T. Birnberg et al.

    Lack of conventional dendritic cells is compatible with normal development and T cell homeostasis, but causes myeloid proliferative syndrome

    Immunity

    (2008)
  • M.J. Clemens

    Interferons and apoptosis

    J Interferon Cytokine Res

    (2003)
  • M.A. Essers et al.

    IFNalpha activates dormant haematopoietic stem cells in vivo

    Nature

    (2009)
  • T. Sato et al.

    Interferon regulatory factor-2 protects quiescent hematopoietic stem cells from type I interferon-dependent exhaustion

    Nat Med

    (2009)
  • G. Schiavoni et al.

    Cyclophosphamide induces type I interferon and augments the number of CD44(hi) T lymphocytes in mice: implications for strategies of chemoimmunotherapy of cancer

    Blood

    (2000)
  • F. Mattei et al.

    Type I IFN regulate DC turnover in vivo

    Eur J Immunol

    (2009)
  • M. Montoya et al.

    Rapid activation of spleen dendritic cell subsets following lymphocytic choriomeningitis virus infection of mice: analysis of the involvement of type 1 IFN

    J Immunol

    (2005)
  • G. Schiavoni et al.

    Type I IFN promote cross-presentation of apoptotic cell-derived antigen by CD8a+ DC and stimulate the cross-priming of CD8 T cells

    Eur J Immunol

    (2009)
  • K. Pokrovskaja et al.

    Alternative signaling pathways regulating type I interferon-induced apoptosis

    J Interferon Cytokine Res

    (2005)
  • J.H. Yen et al.

    Interferon beta induces mature dendritic cell apoptosis through caspase-11/caspase-3 activation

    Blood

    (2009)
  • S.M. Santini et al.

    Type I interferon as a powerful adjuvant for monocyte-derived dendritic cell development and activity in vitro and in Hu-PBL-SCID mice

    J Exp Med

    (2000)
  • F. Moschella et al.

    Gene expression profiling and functional activity of human dendritic cells induced with IFN-alpha-2b: implications for cancer immunotherapy

    Clin Cancer Res

    (2003)
  • C. Falschlehner et al.

    Following TRAIL's path in the immune system

    Immunology

    (2009)
  • M. Lehner et al.

    Type I interferons in combination with bacterial stimuli induce apoptosis of monocyte-derived dendritic cells

    Blood

    (2001)
  • B.L. McRae et al.

    Interferon-alpha and -beta inhibit the in vitro differentiation of immunocompetent human dendritic cells from CD14(+) precursors

    Blood

    (2000)
  • R.L. Paquette et al.

    Interferon-alpha and granulocyte-macrophage colony-stimulating factor differentiate peripheral blood monocytes into potent antigen-presenting cells

    J Leukoc Biol

    (1998)
  • T. Luft et al.

    Type I IFNs enhance the terminal differentiation of dendritic cells

    J Immunol

    (1998)
  • V.R. Cicinnati et al.

    Interferon-alpha differentially affects homeostasis of human plasmacytoid and myeloid dendritic cells

    J Interferon Cytokine Res

    (2009)
  • T.K. Starr et al.

    Positive and negative selection of T cells

    Annu Rev Immunol

    (2003)
  • J.H. Park et al.

    Signaling by intrathymic cytokines, not T cell antigen receptors, specifies CD8 lineage choice and promotes the differentiation of cytotoxic-lineage T cells

    Nat Immunol

    (2010)
  • R. Pacholczyk et al.

    The T-cell receptor repertoire of regulatory T cells

    Immunology

    (2008)
  • Cited by (33)

    • Type I interferons in pancreatic cancer and development of new therapeutic approaches

      2021, Critical Reviews in Oncology/Hematology
      Citation Excerpt :

      It is hypothesized that IFN-α/β removes resident mature DCs from lymphoid tissue and stimulate their replacement by new DCs that have captured TAAs (Mattei et al., 2009). On the other hand, antigen-bearing DCs exhibit enhanced survival, indicating that IFN-α/β affects the homeostasis of DCs differently depending on the maturation status (Mattei et al., 2010). IFN-α/β induced DCs also promote the recruitment of B- and T-cells into the tumor site by expressing several chemokines such as CXCL9, CXCL10, and CXCL11 (Padovan et al., 2002).

    • Nucleic Acid Induced Interferon and Inflammasome Responses in Regulating Host Defense to Gastrointestinal Viruses

      2019, International Review of Cell and Molecular Biology
      Citation Excerpt :

      These STAT heterodimers translocate to the nucleus to induce the expression of numerous interferon stimulated genes (ISG) that aid in restricting viral replication, inhibit protein synthesis and prime neighboring cells for a viral attack (Kisseleva et al., 2002; Wang et al., 2017b). In addition, antiviral IFNα/β signaling can induce apoptosis, thereby destroying the viral replicative niche (Balachandran et al., 2000; Mattei et al., 2010). Type III IFNs (IFNλ) exert antiviral effects analogous to the ones exerted by IFNα/β owing to the similar JAK/STAT signal transduction pathway and IFN response gene expression they can induce upon binding their cellular receptor.

    • Immune Signaling by RIG-I-like Receptors

      2011, Immunity
      Citation Excerpt :

      In addition to its role in driving innate immune defenses, IFN plays a major role in modulating the adaptive immune response. IFN is required to promote T cell survival and clonal expansion after antigen presentation (as reviewed in Mattei et al., 2010). Moreover, interferon potently induces the cytolytic activity of natural killer cells and cytotoxic lymphocytes (Biron et al., 1999) and plays an important role in promoting B cell differentiation and antibody production (Le Bon et al., 2001; Jego et al., 2003).

    View all citing articles on Scopus

    Fabrizio Mattei graduated from Università “La Sapienza” in Rome and completed his specialization in biotechnologies and oncology at the same university. In 2000, he became a fellow at the Edward Jenner Institute for Vaccine Research in David Tough's laboratory, where he acquired experience in the field of dendritic cell immunology. He is currently a senior researcher at the Istituto Superiore di Sanità in Rome where he has focused on the molecular interactions between dendritic cells and type I IFN signalling. His research is currently aimed at understanding the role of IFN signalling in the interface between tumor and immune system in both mouse and human models.

    Giovanna Schiavoni graduated from Università “La Sapienza” in Rome. From 1999 to 2000 she was a fellow at the Edward Jenner Institute for Vaccine Research in David Tough's laboratory, where she carried out studies on type I IFN effects on dendritic cells in vivo. She is currently a senior researcher at the Istituto Superiore di Sanità in Rome where her studies are focused on type I IFN signalling in dendritic cell development, lifespan and function. Her research is currently focused on understanding the role of IFN signalling in the development of anti-tumor immune responses in mouse models.

    David Tough is a Senior Scientific Investigator in the Immuno-Epigenetics Discovery Performance Unit (Epinova) within the Immuno-inflammation Centre of Excellence in Drug Discovery at GlaxoSmithKline. Dr. Tough received his Ph.D. degree in Immunology from The University of Manitoba and completed post-doctoral training at The Scripps Research Institute. Prior to joining GSK in 2006, he was a Senior Group Leader at The Edward Jenner Institute for Vaccine Research. He has published extensively in the area of immunological memory and on the role of cytokines in initiating immune responses and controlling lymphocyte homeostasis. He is an Associate Editor for the Journal of Immunology.

    View full text