Elsevier

Journal of Autoimmunity

Volume 66, January 2016, Pages 40-50
Journal of Autoimmunity

Human liver-resident CD56bright/CD16neg NK cells are retained within hepatic sinusoids via the engagement of CCR5 and CXCR6 pathways

https://doi.org/10.1016/j.jaut.2015.08.011Get rights and content

Highlights

  • Characterization of the mechanisms regulating the homing of NK cells to liver and the repertoire of receptors that distinguish liver-resident NK (lr-NK) cells from circulating counterparts (c-NK cells).

  • Nearly 50% of the entire liver NK cell population is composed of functionally relevant CD56bright lr-NK cells that localize within hepatic sinusoids.

  • CD56bright lr-NK cells express CD69, CCR5 and CXCR6 and this unique repertoire of chemokine receptors is functionally for their migration to hepatic sinusoids expressing their putative ligands.

  • The selective presence of these chemokines in sinusoidal spaces creates a unique tissue niche for lr-CD56bright NK cells that constitutively express CCR5 and CXCR6.

  • This study releases the phenotypic and functional differences that distinguish lr-Nk cells from c-NK cells.

Abstract

Rationale

The liver-specific natural killer (NK) cell population is critical for local innate immune responses, but the mechanisms that lead to their selective homing and the definition of their functionally relevance remain enigmatic.

Objectives

We took advantage of the availability of healthy human liver to rigorously define the mechanisms regulating the homing of NK cells to liver and the repertoire of receptors that distinguish liver-resident NK (lr-NK) cells from circulating counterparts.

Findings

Nearly 50% of the entire liver NK cell population is composed of functionally relevant CD56bright lr-NK cells that localize within hepatic sinusoids. CD56bright lr-NK cells express CD69, CCR5 and CXCR6 and this unique repertoire of chemokine receptors is functionally critical as it determines selective migration in response to the chemotactic stimuli exerted by CCL3, CCL5 and CXCL16. Here, we also show that hepatic sinusoids express CCL3pos Kupffer cells, CXCL16pos endothelial cells and CCL5pos T and NK lymphocytes. The selective presence of these chemokines in sinusoidal spaces creates a unique tissue niche for lr-CD56bright NK cells that constitutively express CCR5 and CXCR6. CD56bright lr-NK cells co-exist with CD56dim conventional NK (c-NK) cells that are, interestingly, transcriptionally and phenotypically similar to their peripheral circulating counterparts. Indeed, CD56dim c-NK cells lack expression of CD69, CCR5, and CXCR6 but express selectins, integrins and CX3CR1.

Conclusion

Our findings disclosing the phenotypic and functional differences between lr-Nk cells and c-NK cells are critical to distinguish liver-specific innate immune responses. Hence, any therapeutic attempts at modifying the large population of CD56bright lr-NK cells will require modification of hepatic CCR5 and CXCR6.

Introduction

Natural killer (NK) cells are important effectors of the innate immune system that can lyse tumor-transformed or virus-infected cells in the absence of prior antigen sensitization. NK cells are also endowed with immune-regulatory functions at tissue sites of inflammation through the establishment of cellular interactions and the production of pro-inflammatory cytokines including IFN-γ, TNF-α, CCL3 (Mip1-α), CCL4 (Mip1-β)and CCL5 (RANTES) [1], [2]. Human peripheral blood NK (PB-NK) cells are divided into two functionally distinct subsets characterized by a different distribution of CD56 and CD16 surface markers. CD56bright/CD16neg-low (CD56bright) NK cells account for 5–15% of all PB-NK cells and, while poorly cytotoxic, can produce large amounts of cytokines. CD56dim/CD16pos (CD56dim) NK cells represent the majority of PB-NK cells (up to 95%) and serve primarily as cytotoxic effectors [3]. In order to spare autologous cells from cytotoxicity and ensure tolerance to self, NK cells receive inhibitory signals from a large family of inhibitory NK cell receptors (iNKRs), that include Killer cell immunoglobulin-like receptors (KIRs) and C-type lectins recognizing specific alleles of self MHC-class-I molecules (MHC-I). NK cell effector-functions are generally induced by the engagement of another family of activating NK cell receptors (aNKRs) that binds their ligands expressed on stressed, infected or tumor-transformed target cells that either lack or have a decreased expression of self-MHC-I [1], [4], [5], [6].

NK cells can account up to 50% of the total lymphocyte population in the human liver, which is in contrast to their lower frequency in blood (5–15%) or other peripheral tissues such as lymph nodes. Hence, great efforts have been placed over the recent years to understand the role of hepatic NK cells in the pathogenesis of liver disorders including fibrosis, viral infections, tumors and autoimmune diseases [7], [8], [9]. In this regard, a distinct subset of hepatic NK cells endowed with adaptive immune properties and exhibiting antigen-specific recall responses to viruses and haptens has been recently described in mice [10], [11]. This memory-like response is exerted by a unique subset of CD49apos/DX5neg liver-resident NK (lr-NK) cells that are phenotypically and functionally distinct from peripheral blood CD49aneg/DX5pos conventional NK (c-NK) cells circulating throughout spleen and liver. Likewise, it has been reported that NK cells in human liver are different from their circulating counterparts [7], [12], [13], [14], [15], [16], thus suggesting that a unique lr-NK cell subset also exists in the human liver under homeostatic conditions. However, the overall frequency and the precise phenotype of human lr-NK cells are still being debated, while the distribution and the homing mechanisms regulating their retention in the liver are unknown. The present study characterizes CD56bright lr-NK cells selectively located within hepatic sinusoids and accounting for half of the entire hepatic NK cell population. We demonstrate that CD56bright lr-NK cells are phenotypically and transcriptionally distinct from PB-CD56bright NK cells and constitutively express high levels of markers associated with tissue residency including CD69, CCR5 and CXCR6. This unique phenotype is functionally relevant as CD56bright lr-NK cells migrate in response to CCL3, CCL5 and CXCL16, the CCR5 and CXCR6 ligands highly expressed within liver sinusoids on Kupffer cells, T and NK lymphocytes as well as endothelial cells.

Section snippets

Human donors

Human peripheral blood mononuclear cells (PBMCs) were isolated from buffy coats of healthy donors obtained in accordance with clinical protocols approved by the Institutional Review Board of Desio Hospital, Milan, Italy. Liver specimens were obtained from patients undergoing liver resection to remove liver metastases of colorectal carcinoma. Fragments of hepatic tissues used for our experiments were macroscopically and microscopically free of any diseases and considered healthy, as assessed by

CD56bright hepatic NK cells are enriched at high frequencies in the healthy human liver

Similar to their circulating counterparts, human hepatic NK cells can be distinguished into two CD56pos/CD16neg and CD56pos/CD16pos cell subsets under homeostatic conditions [3], [19]. However, the frequency of CD56pos/CD16neg hepatic NK cells is significantly higher compared to that of CD56pos/CD16neg PB-NK cells in matched donors [7], [22] (Fig. 1A and C). CD56pos/CD16neg PB-NK cells are conventionally defined as CD56bright NK cells due to the higher mean fluorescence intensity (MFI) of CD56

Discussion

The present study characterizes a novel subset of CD56bright lr-NK cells, whose transcriptional and phenotypic profiles differ from those of its circulating counterparts. We show that CD56bright lr-NK cells constitutively express markers of tissue-residency including CD69, CCR5 and CXCR6. The unique repertoire of chemokine receptors on CD56bright lr-NK cells is functionally relevant as this subset selectively migrates in response to the chemotactic stimuli given by CCR5 (i.e. CCL3 and CCL5) and

Author contributions

Conception and design: K.H. and D.M; Contributed unpublished, essential data, or reagents (recruited patients for liver specimens): M.D., M.C. and G.T.; Contributed unpublished, essential data, or reagents (recruited patients for liver-perfusate specimens): M.H. and A.B; Acquisition of data, analysis and interpretation of data: K.H., E.P., P.T., M.P., S.B., P.I., E.L., M.E.G., D.M. K.H. and D.M.; Drafting the article: K.H. and D.M.

Conflict of interest

The authors have declared that no conflict of interest exists.

Acknowledgments

We are deeply grateful to all human donors and patients participating to this study for their generosity and support. This work was supported by the Italian Ministry of Health (Bando Giovani Ricercatori, GR-2008-1135082 to D.M.), Italian Association for Cancer Research (AIRC IG 14687 to D.M.), the European Union (Marie Curie International Reintegration Grant 322093 to E.L.), the National Institutes of Health (DK39588 to M.E.G.) and by the intramural research program of Humanitas Clinical and

References (75)

  • T. Sathaliyawala et al.

    Distribution and compartmentalization of human circulating and tissue-resident memory T cell subsets

    Immunity

    (2013)
  • E.B. Alabraba et al.

    A new approach to isolation and culture of human Kupffer cells

    J. Immunol. Methods

    (2007)
  • I.D. Weiss et al.

    Ccr5 deficiency regulates the proliferation and trafficking of natural killer cells under physiological conditions

    Cytokine

    (2011)
  • A.G. Freud et al.

    A human CD34(+) subset resides in lymph nodes and differentiates into CD56bright natural killer cells

    Immunity

    (2005)
  • M.A. Cooper et al.

    NK cell and DC interactions

    Trends Immunol.

    (2004)
  • S. Nedvetzki et al.

    Reciprocal regulation of human natural killer cells and macrophages associated with distinct immune synapses

    Blood

    (2007)
  • C. Costantini et al.

    Human neutrophils interact with both 6-sulfo LacNAc+ DC and NK cells to amplify NK-derived IFN{gamma}: role of CD18, ICAM-1, and ICAM-3

    Blood

    (2011)
  • J. Yu et al.

    Location and cellular stages of natural killer cell development

    Trends Immunol.

    (2013)
  • J.W. Fathman et al.

    Identification of the earliest natural killer cell-committed progenitor in murine bone marrow

    Blood

    (2011)
  • H.R. Rodewald et al.

    A population of early fetal thymocytes expressing Fc gamma RII/III contains precursors of T lymphocytes and natural killer cells

    Cell

    (1992)
  • H. Chinen et al.

    Lamina propria c-kit+ immune precursors reside in human adult intestine and differentiate into natural killer cells

    Gastroenterology

    (2007)
  • A.A. Ansari

    Clinical features and pathobiology of Ebolavirus infection

    J. Autoimmun.

    (2014)
  • S. Berrih-Aknin

    Myasthenia Gravis: paradox versus paradigm in autoimmunity

    J. Autoimmun.

    (2014)
  • L. Steinman et al.

    From defining antigens to new therapies in multiple sclerosis: honoring the contributions of Ruth Arnon and Michael Sela

    J. Autoimmun.

    (2014)
  • E. Vivier et al.

    Innate or adaptive immunity? the example of natural killer cells

    Science

    (2011)
  • E. Lugli et al.

    NK cell subset redistribution during the course of viral infections

    Front. Immunol.

    (2014)
  • L.L. Lanier

    NK cell recognition

    Annu. Rev. Immunol.

    (2005)
  • A. Moretta et al.

    Activating receptors and coreceptors involved in human natural killer cell-mediated cytolysis

    Annu. Rev. Immunol.

    (2001)
  • E.O. Long et al.

    Controlling natural killer cell responses: integration of signals for activation and inhibition

    Annu. Rev. Immunol.

    (2013)
  • B. Oliviero et al.

    Natural killer cell functional dichotomy in chronic hepatitis B and chronic hepatitis C virus infections

    Gastroenterology

    (2009)
  • B. Rehermann

    Pathogenesis of chronic viral hepatitis: differential roles of T cells and NK cells

    Nat. Med.

    (2013)
  • H. Peng et al.

    Liver-resident NK cells confer adaptive immunity in skin-contact inflammation

    J. Clin. Invest.

    (2013)
  • S. Paust et al.

    Critical role for the chemokine receptor CXCR6 in NK cell-mediated antigen-specific memory of haptens and viruses

    Nat. Immunol.

    (2010)
  • Z. Zhang et al.

    Hypercytolytic activity of hepatic natural killer cells correlates with liver injury in chronic hepatitis B patients

    Hepatology

    (2011)
  • K. Ishiyama et al.

    Difference in cytotoxicity against hepatocellular carcinoma between liver and periphery natural killer cells in humans

    Hepatology

    (2006)
  • O. Wald et al.

    Involvement of the CXCL12/CXCR4 pathway in the advanced liver disease that is associated with hepatitis C virus or hepatitis B virus

    Eur. J. Immunol.

    (2004)
  • N. Marquardt et al.

    Cutting edge: identification and characterization of human intrahepatic CD49a+ NK cells

    J. Immunol.

    (2015 Mar 15)
  • Cited by (195)

    View all citing articles on Scopus
    View full text