Development and maturation of natural killer cells

https://doi.org/10.1016/j.coi.2016.01.007Get rights and content

Highlights

  • How do NK cells diverge from other innate lymphocyte cell fates?

  • Transcription factors key in early NK cell development include Nfil3, Ets family.

  • Transcription factors critical for NK cell maturation include Id2, T-bet, Eomes.

  • Activating cytokines, STATs control NK cell cytotoxicity and IFN-γ production.

  • Apoptosis and autophagy regulate development of memory NK cells.

Natural killer (NK) cells are innate lymphocytes that are critical for host protection against pathogens and cancer due to their ability to rapidly release inflammatory cytokines and kill infected or transformed cells. In the 40 years since their initial discovery, much has been learned about how this important cellular lineage develops and functions. We now know that NK cells are the founding members of an expanded family of lymphocyte known as innate lymphoid cells (ILC). Furthermore, we have recently discovered that NK cells can possess features of adaptive immunity such as antigen specificity and long-lived memory responses. Here we will review our current understanding of the molecular mechanisms driving development of NK cells from the common lymphoid progenitor (CLP) to mature NK cells, and from activated effectors to long-lived memory NK cells.

Introduction

NK cells, like B and T cells, are a lymphocyte lineage derived from the CLP [1], and like B cells, are thought to develop primarily in the bone marrow [2], although other sites of development, such as the liver and thymus, have also been proposed (reviewed in [3]). However, unlike the antigen receptors of B and T cells, NK cell receptors are germ line encoded and do not require gene rearrangement by RAG recombinase [4], though recent work has suggested that RAG plays an unexpected cell-intrinsic role in NK cell development [5••]. NK cells also undergo an ‘education’ process during development where they acquire the ability to recognize lack of self MHC class I, or ‘missing-self’, a feature that facilitates their surveillance of target cells that have down-regulated MHC class I during infection or malignancy [6]. NK cells rely on both cytokines and transcription factors to promote and control their development. Cytokine signaling from interleukin (IL)-15 is critical for the development of NK cells and is required throughout their lifetime [7, 8]. Transcription factors such as Nfil3 and PU.1 are necessary for development of early NK cell progenitors [9, 10, 11, 12], whereas Id2, Tox, and others are important later in development [13, 14, 15]. Eomes and T-bet are among factors that then control the final stages of NK cell maturation [16, 17]. In the periphery, the activation and differentiation of NK cells are regulated by a plethora of transcription factors mediating distinct effector functions. This review will outline current knowledge about the stages of NK cell development and the factors driving each stage.

Section snippets

Stages of NK cell development and differentiation

The CLP is characterized by expression of IL-7Rα (CD127), c-kit (CD117), Sca-1, and Flt-3 (CD135), as well as the lack of common lineage markers such as CD3, CD4, CD8, CD19, Ter119, Gr-1 and NK1.1 (Figure 1) [1]. From the CLP, cells develop into NK cell precursors (NKP), which are defined by expression of the IL-15 receptor β chain (CD122), and lack of common lineage markers, including the NK cell markers NK1.1 and DX5 (CD49b) (Figure 1) [2]. This NKP population has been further refined based

Transcriptional control of early NK cell development

Lineage commitment to either an adaptive or innate lymphocyte cell fate is determined by a complex network of transcription factors (Figure 2). For example, Notch signaling through the ligands Jagged1 and Jagged2 preferentially drives NK cell development from the CLP [28, 29, 30], whereas delta-like ligands (DLL) promote T cell development [31]. Moreover, thymocytes can be diverted into an NK cell-like fate if the Notch1-dependent transcription factor Bcl11b is ablated during T cell development

Transcription factors governing NK cell maturation

In addition to the early role for Id family transcription factors in suppressing the adaptive lymphocyte fate while promoting innate lymphocyte development, these factors are also important later in the development of NK cells. Id2-deficient mice have a cell-intrinsic lack of peripheral NK cells [13] that was found to be due to an arrest at the iNK stage [14], indicating that Id2 is important in the transition from immature to mature NK cell. Both Id2 and Id3 are expressed in NKP, and Id2

Regulation of effector NK cell responses and memory formation

The STAT family of transcription factors contains members that are phosphorylated downstream of pro-inflammatory cytokine receptors and form homo-dimers or hetero-dimers that translocate to the nucleus to induce gene transcription (reviewed in [80]). During viral infection, type I IFNs and downstream STAT1 have been shown to enhance NK cell cytotoxicity (Figure 3) [81, 82], and shield activated NK cells from cell death via an NKG2D-dependent fratricide mechanism [83]. IL-12 and downstream

Concluding remarks

As is evident by the number of groundbreaking studies discussed in this review, the development of NK cells is a highly dynamic process and new discoveries occur each week. Still, there is so much we do not yet understand. Future work will elucidate the mechanisms regulating the various factors described above, how the factors interact with each other, and how they might be involved in disease processes.

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

We thank members of the Sun lab for helpful discussions. In particular we thank Aimee Beaulieu, Clair Geary, and Tim O'Sullivan, who read and commented on this review. We apologize to those whose work we were unable to discuss due to space limitations. TLG is supported by a fellowship from the National Institute of Allergy and Infectious Diseases (F31 AI114019). JCS is supported by the Searle Scholars Program, the Cancer Research Institute, and National Institutes of Health grants AI085034 and

References (96)

  • S.L. DeHart et al.

    Jagged2 promotes the development of natural killer cells and the establishment of functional natural killer cell lines

    Blood

    (2005)
  • A.C. Jaleco et al.

    Differential effects of Notch ligands Delta-1 and Jagged-1 in human lymphoid differentiation

    J Exp Med

    (2001)
  • S.M. Lehar et al.

    Notch ligands Delta 1 and Jagged1 transmit distinct signals to T-cell precursors

    Blood

    (2005)
  • T. Ikawa et al.

    An essential developmental checkpoint for production of the T cell lineage

    Science

    (2010)
  • P. Li et al.

    Reprogramming of T cells to natural killer-like cells upon Bcl11b deletion

    Science

    (2010)
  • X. Yu et al.

    The basic leucine zipper transcription factor NFIL3 directs the development of a common innate lymphoid cell precursor

    eLife

    (2014)
  • S. Mitsui et al.

    Antagonistic role of E4BP4 and PAR proteins in the circadian oscillatory mechanism

    Genes Dev

    (2001)
  • A. Marçais et al.

    The metabolic checkpoint kinase mTOR is essential for IL-15 signaling during the development and activation of NK cells

    Nat Immunol

    (2014)
  • Y. Deng et al.

    Transcription factor Foxo1 is a negative regulator of natural killer cell maturation and function

    Immunity

    (2015)
  • N.D. Huntington et al.

    Interleukin 15-mediated survival of natural killer cells is determined by interactions among Bim, Noxa and Mcl-1

    Nat Immunol

    (2007)
  • S. Taki et al.

    IFN regulatory factor-2 deficiency revealed a novel checkpoint critical for the generation of peripheral NK cells

    J Immunol

    (2005)
  • S.-I. Ohno et al.

    Runx proteins are involved in regulation of CD122, Ly49 family and IFN-gamma expression during NK cell differentiation

    Int Immunol

    (2008)
  • J.C. Sun et al.

    Proinflammatory cytokine signaling required for the generation of natural killer cell memory

    J Exp Med

    (2012)
  • S. Madera et al.

    Cutting edge: stage-specific requirement of IL-18 for antiviral NK cell expansion

    J Immunol

    (2015)
  • T. Nabekura et al.

    IL-33 receptor ST2 amplifies the expansion of NK cells and enhances host defense during mouse cytomegalovirus infection

    J Immunol

    (2015)
  • W.E. Seaman et al.

    Natural killer cells, bone, and the bone marrow: studies in estrogen-treated mice and in congenitally osteopetrotic (mi/mi) mice

    J Immunol

    (1979)
  • A. Ito et al.

    Inhibitory effect on natural killer activity of microphthalmia transcription factor encoded by the mutant mi allele of mice

    Blood

    (2001)
  • T.R. Kataoka et al.

    Reduced expression of IL-12 receptor beta2 and IL-18 receptor alpha genes in natural killer cells and macrophages derived from B6-mi/mi mice

    Lab Invest

    (2005)
  • D.G.T. Hesslein et al.

    Transcriptional control of natural killer cell development and function

    Adv Immunol

    (2011)
  • E.E. Rosmaraki et al.

    Identification of committed NK cell progenitors in adult murine bone marrow

    Eur J Immunol

    (2001)
  • L.L. Lanier et al.

    Natural killer cells: definition of a cell type rather than a function

    J Immunol

    (1986)
  • M.T. Orr et al.

    Natural killer cell education and tolerance

    Cell

    (2010)
  • W.M. Yokoyama et al.

    The dynamic life of natural killer cells

    Annu Rev Immunol

    (2004)
  • D.M. Gascoyne et al.

    The basic leucine zipper transcription factor E4BP4 is essential for natural killer cell development

    Nat Immunol

    (2009)
  • S. Kamizono et al.

    Nfil3/E4bp4 is required for the development and maturation of NK cells in vivo

    J Exp Med

    (2009)
  • M. Kashiwada et al.

    IL-4-induced transcription factor NFIL3/E4BP4 controls IgE class switching

    Proc Natl Acad Sci U S A

    (2010)
  • F. Colucci et al.

    Differential requirement for the transcription factor PU.1 in the generation of natural killer cells versus B and T cells

    Blood

    (2001)
  • M.D. Boos et al.

    Mature natural killer cell and lymphoid tissue-inducing cell development requires Id2-mediated suppression of E protein activity

    J Exp Med

    (2007)
  • P. Aliahmad et al.

    Shared dependence on the DNA-binding factor TOX for the development of lymphoid tissue-inducer cell and NK cell lineages

    Nat Immunol

    (2010)
  • M.J. Townsend et al.

    T-bet regulates the terminal maturation and homeostasis of NK and Valpha14i NKT cells

    Immunity

    (2004)
  • Q. Yang et al.

    TCF-1 upregulation identifies early innate lymphoid progenitors in the bone marrow

    Nat Immunol

    (2015)
  • S. Kim et al.

    In vivo developmental stages in murine natural killer cell maturation

    Nat Immunol

    (2002)
  • Y. Hayakawa et al.

    CD27 dissects mature NK cells into two subsets with distinct responsiveness and migratory capacity

    J Immunol

    (2006)
  • L. Chiossone et al.

    Maturation of mouse NK cells is a 4-stage developmental program

    Blood

    (2009)
  • G. Min-Oo et al.

    Natural killer cells: walking three paths down memory lane

    Trends Immunol

    (2013)
  • S.M. Kaech et al.

    Heterogeneity and cell-fate decisions in effector and memory CD8+ T cell differentiation during viral infection

    Immunity

    (2007)
  • I. Maillard et al.

    Regulation of lymphoid development, differentiation, and function by the Notch pathway

    Annu Rev Immunol

    (2005)
  • L. Li et al.

    An early T cell lineage commitment checkpoint dependent on the transcription factor Bcl11b

    Science

    (2010)
  • Cited by (103)

    • Innate lymphoid cells: potential targets for cancer therapeutics

      2023, Trends in Cancer
      Citation Excerpt :

      Until recently, ILCs have been represented by natural killer (NK) cells [12] and LTi cells [13], but this family has now been expanded by the discovery of additional subsets [14]. NK cells have been extensively characterized for their capacity to kill tumor cells and virus-infected cells in the absence of a previous exposure to an antigen [15–17], while LTi cells are recruited to embryonic anlagen where they are crucial for the development of secondary lymphoid tissues (see Glossary). Collectively, ILCs can now be divided into five distinct subsets: NK cells, ILC1s, ILC2s, ILC3s, and LTi cells, which have been defined based on the identification of their lineage-specific progenitor populations [14] (Figure 1).

    • The immune system

      2022, Clinical Immunology
    View all citing articles on Scopus
    View full text