Diversity of peripheral blood human NK cells identified by single-cell RNA sequencing.

Human natural killer (NK) cells in peripheral blood perform many functions, and classification of specific subsets has been a longstanding goal. We report single-cell RNA sequencing of NK cells, comparing gene expression in unstimulated and interleukin (IL)-2-activated cells from healthy cytomegalovirus (CMV)-negative donors. Three NK cell subsets resembled well-described populations; CD56brightCD16-, CD56dimCD16+CD57-, and CD56dimCD16+CD57+. CD56dimCD16+CD57- cells subdivided to include a population with higher chemokine mRNA and increased frequency of killer-cell immunoglobulin-like receptor expression. Three novel human blood NK cell populations were identified: a population of type I interferon-responding NK cells that were CD56neg; a population exhibiting a cytokine-induced memory-like phenotype, including increased granzyme B mRNA in response to IL-2; and finally, a small population, with low ribosomal expression, downregulation of oxidative phosphorylation, and high levels of immediate early response genes indicative of cellular activation. Analysis of CMV+ donors established that CMV altered the proportion of NK cells in each subset, especially an increase in adaptive NK cells, as well as gene regulation within each subset. Together, these data establish an unexpected diversity in blood NK cells and provide a new framework for analyzing NK cell responses in health and disease.

[1]  J. Verbsky,et al.  Heterogeneity of human bone marrow and blood natural killer cells defined by single-cell transcriptome , 2019, Nature Communications.

[2]  S. Linnarsson,et al.  Remodeling of secretory lysosomes during education tunes functional potential in NK cells , 2019, Nature Communications.

[3]  R. Johnson,et al.  Gene Regulatory Programs Conferring Phenotypic Identities to Human NK Cells , 2019, Cell.

[4]  J. Orange,et al.  Emerging insights into human health and NK cell biology from the study of NK cell deficiencies , 2018, Immunological reviews.

[5]  The Gene Ontology Consortium,et al.  The Gene Ontology Resource: 20 years and still GOing strong , 2018, Nucleic Acids Res..

[6]  Eric Vivier,et al.  High-Dimensional Single-Cell Analysis Identifies Organ-Specific Signatures and Conserved NK Cell Subsets in Humans and Mice , 2018, Immunity.

[7]  B. Suarez-Alvarez,et al.  Acute myeloid leukemia and NK cells: two warriors confront each other , 2018, Oncoimmunology.

[8]  É. Vivier,et al.  Natural killer cells and other innate lymphoid cells in cancer , 2018, Nature Reviews Immunology.

[9]  F. Klawonn,et al.  Proteome analysis of human CD56neg NK cells reveals a homogeneous phenotype surprisingly similar to CD56dim NK cells , 2018, European journal of immunology.

[10]  T. Michel,et al.  Revisiting the Functional Impact of NK Cells. , 2018, Trends in immunology.

[11]  Gregory A. Wyant,et al.  NUFIP1 is a ribosome receptor for starvation-induced ribophagy , 2018, Science.

[12]  Paul Hoffman,et al.  Integrating single-cell transcriptomic data across different conditions, technologies, and species , 2018, Nature Biotechnology.

[13]  E. Sahai,et al.  NK Cells Stimulate Recruitment of cDC1 into the Tumor Microenvironment Promoting Cancer Immune Control , 2018, Cell.

[14]  M. Caligiuri,et al.  The Broad Spectrum of Human Natural Killer Cell Diversity. , 2017, Immunity.

[15]  S. Dawson,et al.  CMTM6 maintains the expression of PD-L1 and regulates anti-tumour immunity , 2017, Nature.

[16]  H. Horlings,et al.  Identification of CMTM6 and CMTM4 as PD-L1 protein regulators , 2017, Nature.

[17]  J. Strominger,et al.  Transcriptome analysis reveals similarities between human blood CD3− CD56bright cells and mouse CD127+ innate lymphoid cells , 2017, Scientific Reports.

[18]  E. Riley,et al.  NK Cells: Uncertain Allies against Malaria , 2017, Front. Immunol..

[19]  C. Watzl,et al.  LFA-1 Activation in NK Cells and Their Subsets: Influence of Receptors, Maturation, and Cytokine Stimulation , 2017, The Journal of Immunology.

[20]  C. Mody,et al.  Granule-Dependent Natural Killer Cell Cytotoxicity to Fungal Pathogens , 2017, Front. Immunol..

[21]  L. Lanier,et al.  Tracking the fate of antigen-specific versus cytokine-activated natural killer cells after cytomegalovirus infection , 2016, The Journal of experimental medicine.

[22]  F. Claas,et al.  Cytokine-induced memory-like natural killer cells exhibit enhanced responses against myeloid leukemia , 2016, Science Translational Medicine.

[23]  L. Lanier,et al.  NK cells and type 1 innate lymphoid cells: partners in host defense , 2016, Nature Immunology.

[24]  T. Michel,et al.  Human CD56bright NK Cells: An Update , 2016, The Journal of Immunology.

[25]  Åsa K. Björklund,et al.  The heterogeneity of human CD127+ innate lymphoid cells revealed by single-cell RNA sequencing , 2016, Nature Immunology.

[26]  L. Lanier,et al.  Natural killer cell memory in infection, inflammation and cancer , 2016, Nature Reviews Immunology.

[27]  H. Ljunggren,et al.  Cytomegalovirus infection drives adaptive epigenetic diversification of NK cells with altered signaling and effector function. , 2015, Immunity.

[28]  David K. Finlay,et al.  mTORC1-Dependent Metabolic Reprogramming Is a Prerequisite for NK Cell Effector Function , 2014, The Journal of Immunology.

[29]  C. Bottomley,et al.  Rapid NK cell differentiation in a population with near-universal human cytomegalovirus infection is attenuated by NKG2C deletions. , 2014, Blood.

[30]  A. Pera,et al.  Shaping of NK cell subsets by aging. , 2014, Current opinion in immunology.

[31]  A. Moffett,et al.  Uterine NK cells: active regulators at the maternal-fetal interface. , 2014, The Journal of clinical investigation.

[32]  J. Orange,et al.  Cell biological steps and checkpoints in accessing NK cell cytotoxicity , 2014, Immunology and cell biology.

[33]  Gary E. Swan,et al.  Genetic and Environmental Determinants of Human NK Cell Diversity Revealed by Mass Cytometry , 2013, Science Translational Medicine.

[34]  Ludo Waltman,et al.  A smart local moving algorithm for large-scale modularity-based community detection , 2013, The European Physical Journal B.

[35]  W. Yokoyama,et al.  Natural killer cells in human autoimmune disorders , 2013, Arthritis Research & Therapy.

[36]  C. Infante-Duarte,et al.  The Role of Natural Killer Cells in Multiple Sclerosis and Their Therapeutic Implications , 2012, Front. Immunol..

[37]  T. Fehniger,et al.  Cytokine activation induces human memory-like NK cells. , 2012, Blood.

[38]  L. Lanier,et al.  Cytomegalovirus reactivation after allogeneic transplantation promotes a lasting increase in educated NKG2C+ natural killer cells with potent function. , 2012, Blood.

[39]  T. Asselah,et al.  CMV drives clonal expansion of NKG2C+ NK cells expressing self‐specific KIRs in chronic hepatitis patients , 2012, European journal of immunology.

[40]  Eleanor M. Riley,et al.  Activation of Natural Killer Cells during Microbial Infections , 2011, Front. Immun..

[41]  P. J. Norris,et al.  Expansion of a unique CD57+NKG2Chi natural killer cell subset during acute human cytomegalovirus infection , 2011, Proceedings of the National Academy of Sciences.

[42]  J. Madsen,et al.  Natural killer cells in rejection and tolerance of solid organ allografts , 2011, Current opinion in organ transplantation.

[43]  H. Ljunggren,et al.  Rapid expansion and long-term persistence of elevated NK cell numbers in humans infected with hantavirus , 2011, The Journal of experimental medicine.

[44]  M. Caligiuri,et al.  PRDM1/Blimp-1 Controls Effector Cytokine Production in Human NK Cells , 2010, The Journal of Immunology.

[45]  H. Pircher,et al.  CD57 defines a functionally distinct population of mature NK cells in the human CD56dimCD16+ NK-cell subset. , 2010, Blood.

[46]  H. Ljunggren,et al.  Expression patterns of NKG2A, KIR, and CD57 define a process of CD56dim NK-cell differentiation uncoupled from NK-cell education. , 2010, Blood.

[47]  H. Ljunggren,et al.  CD56 negative NK cells: origin, function, and role in chronic viral disease. , 2010, Trends in immunology.

[48]  Daniel J Klionsky,et al.  Mammalian autophagy: core molecular machinery and signaling regulation. , 2010, Current opinion in cell biology.

[49]  M. Caligiuri,et al.  CD94 surface density identifies a functional intermediary between the CD56bright and CD56dim human NK-cell subsets. , 2010, Blood.

[50]  H. Ljunggren,et al.  Expansion of Functionally Skewed CD56-Negative NK Cells in Chronic Hepatitis C Virus Infection: Correlation with Outcome of Pegylated IFN-α and Ribavirin Treatment1 , 2009, The Journal of Immunology.

[51]  P. Eid,et al.  PolyI:C plus IL‐2 or IL‐12 induce IFN‐γ production by human NK cells via autocrine IFN‐β , 2009, European journal of immunology.

[52]  Liping Yang,et al.  Cytokine-induced memory-like natural killer cells , 2009, Proceedings of the National Academy of Sciences.

[53]  Joseph C. Sun,et al.  Adaptive Immune Features of Natural Killer Cells , 2009, Nature.

[54]  M. Caligiuri,et al.  Human natural killer cells. , 2008, Blood.

[55]  M. Sohrmann,et al.  Mature ribosomes are selectively degraded upon starvation by an autophagy pathway requiring the Ubp3p/Bre5p ubiquitin protease , 2008, Nature Cell Biology.

[56]  J. Hanna,et al.  Novel Insights on Human NK Cells’ Immunological Modalities Revealed by Gene Expression Profiling1 , 2004, The Journal of Immunology.

[57]  N. Malats,et al.  Imprint of human cytomegalovirus infection on the NK cell receptor repertoire. , 2004, Blood.

[58]  Michael T. Wilson,et al.  The response of natural killer T cells to glycolipid antigens is characterized by surface receptor down-modulation and expansion , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[59]  W. Leonard,et al.  IL-2 negatively regulates IL-7 receptor α chain expression in activated T lymphocytes , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[60]  M. Ashburner,et al.  Gene Ontology: tool for the unification of biology , 2000, Nature Genetics.

[61]  J V Giorgi,et al.  Natural killer cell immunodeficiency in HIV disease is manifest by profoundly decreased numbers of CD16+CD56+ cells and expansion of a population of CD16dimCD56- cells with low lytic activity. , 1995, Journal of acquired immune deficiency syndromes and human retrovirology : official publication of the International Retrovirology Association.

[62]  E. Gianchecchi,et al.  NK cells in autoimmune diseases: Linking innate and adaptive immune responses. , 2018, Autoimmunity reviews.