Skip to main content

Functional integration of natural killer cells in a microfluidically perfused liver on-a-chip model

This article has been updated

Abstract

Objective

The liver acts as an innate immunity-dominant organ and natural killer (NK) cells, are the main lymphocyte population in the human liver. NK cells are in close interaction with other immune cells, acting as the first line of defense against pathogens, infections, and injury.

A previously developed, three-dimensional, perfused liver-on-a-chip comprised of human cells was used to integrate NK cells, representing pivotal immune cells during liver injury and regeneration. The objective of this study was to integrate functional NK cells in an in vitro model of the human liver and assess utilization of the model for NK cell-dependent studies of liver inflammation.

Results

NK cells from human blood and liver specimen were isolated by Percoll separation with subsequent magnetic cell separation (MACS), yielding highly purified blood and liver derived NK cells. After stimulation with toll-like-receptor (TLR) agonists (lipopolysaccharides, Pam3CSK4), isolated NK cells showed increased interferon (IFN)-gamma secretion. To study the role of NK cells in a complex hepatic environment, these cells were integrated in the vascular compartment of a microfluidically supported liver-on-a-chip model in close interaction with endothelial and resident macrophages. Successful, functional integration of NK cells was verified by immunofluorescence staining (NKp46), flow cytometry analysis and TLR agonist-dependent secretion of interleukin (IL)-6 and tumor necrosis factor (TNF)-alpha. Lastly, we observed that inflammatory activation of NK cells in the liver-on-a-chip led to a loss of vascular barrier integrity. Overall, our data shows the first successful, functional integration of NK cells in a liver-on-a-chip model that can be utilized to investigate NK cell-dependent effects on liver inflammation in vitro.

Peer Review reports

Introduction

The liver acts as an innate immunity-dominant organ and natural killer (NK) cells therefore provide the first line of defense against pathogens, infections or tumors [1, 2]. NK cells are the main lymphocyte population in the human liver accounting for up to 50%, commonly defined by expression of the surface marker CD (cluster of differentiation) 56 and lack of CD3 expression [3, 4]. During liver disease the hepatic NK cell number is modulated, possibly due to increased recruitment of circulating NK cells to the liver [5,6,7]. A diverse repertoire of NK cell surface receptors allows recognition and rapid response to cell damage and stress [8]. NK cells further coordinate early events of the innate immune response to injury through close interactions with other immune cells, such as T cells, dendritic cells or macrophages [8], and pro-inflammatory cytokine production, such as interferon gamma (IFN-gamma) [3, 8,9,10], tumor necrosis factor alpha (TNF-alpha) [3, 10], and granulocyte–macrophage colony-stimulating factor [3]. In addition, it has been shown that NK cells produce interleukin-6 (IL-6), TNF-alpha, and IL-1beta after stimulation with lipopolysaccharides (LPS), a common toll-like receptor (TLR) agonist [3]. Furthermore, a variety of surface receptors, such as purinergic receptors and tumor necrosis factor-related apoptosis-inducing ligand, control the cytotoxic activity of NK cells [9, 11] and facilitate NK cell maturation as well as function depend on their cellular localization site [12, 13]. Functionally, NK cells contribute through complex orchestrated pathways, e.g. cytokine or receptor mediated activation, changes of gene expression and interaction with other immune cells (e.g. macrophages), to clearance of damaged hepatocytes during hepatic injury and regeneration [14, 15]. By eliminating susceptible targets through multiple, non-redundant mechanisms, NK cells amplify or reduce the inflammatory response [12]. The close relationship and interaction of NK cells with other immune cells, such as Kupffer cells in the liver sinusoids, is mainly due to cytokine and chemokine secretion [4, 16, 17]. It has been shown that e.g. IL-12 and IL-18, secreted by Kupffer cells, are strong activators of NK cells and necessary for differentiation of NK cells [18,19,20].

Currently, investigation of NK cell function and hepatic immune cell interaction is limited by the insufficient physiological complexity of available in vitro models. One promising approach is the use of microphysiological systems that allow multicellular organ modelling in a dynamic microenvironment [21,22,23]. In this study, we expanded a previously established three-dimensional, microfluidically perfused liver-on-a-chip model comprised of human cells (macrophages, endothelial cells, hepatocytes, stellate cells) represents a functional unit of the liver sinusoid and was previously described [24,25,26]. One study, dealing with taurolidine-induced liver damage, provided comparable results generated from the liver-on-a-chip and an animal model [27]. The overall goal was to integrate human NK cells and show their recruitment and functional response to inflammatory triggers within the liver-on-a-chip. Our study substantiates the use of the developed liver-on-a-chip model for studies of NK cells and paves the way for further immunological in vitro studies.

Main text

Isolation of NK cells from human blood and liver specimen

Initially, NK cells were freshly isolated from either human blood or human liver tissue. Specifically, blood-derived NK cell populations were isolated from peripheral blood mononuclear cells (PBMC) using a previously described Percoll separation procedure, followed by PE electromagnetic beads and LS columns sorting, according to manufacturer’s instructions (Miltenyi Biotec, Auburn, CA, USA) [11]. Liver tissue-derived NK cells were isolated from cold-stored, tumor-free liver specimen immediately after surgical resection. Liver specimen were cut into small pieces, passed through a 200 gauge stainless mesh and separated similarly to blood-derived NK cells [28, 29]. The amount and purity of NK cells was assessed by flow cytometry analysis using CD56 (positive selection) and CD3 (negative selection). CD56+CD3 NK cell populations from blood or liver tissue were used for further experiments immediately after isolation.

For both isolation procedures, purity of NK cell populations were 89.3 ± 4.9% and 92.7 ± 4.6%, respectively (Fig. 1A), comparable to previous reports [29].

Fig. 1
figure 1

FACS analysis for CD56+CD3 NK cells in the NK cell suspension revealed high percentage of isolated NK cells derived from human blood and human liver specimen (A). Representative immunofluorescence images of NK cells (B) stained with cytotoxicity and maturation marker NKp46 in monoculture of isolated blood NK cells (green = NKp46, blue = DAPI). IFN-gamma secretion assessed by Cytometric Bead Array (CBA) assay (BD Biosciences) of isolated NK cells after 16 h of TLR-receptor agonist stimulation (LPS and Pam3CSK4) was significantly increased compared to controls (C). Figures AC present data of at least three independent experiments, p-values are assessed by Mann Whitney test

NKp46, a marker of NK cell cytotoxicity and maturation [30, 31], was expressed on isolated NK cells, which was verified by immunofluorescence staining (Fig. 1B and Additional file 1: Fig. S1). To activate NK cells in vitro, these cells can be stimulated with a macrophage-derived cytokine mixture (e.g. IL-12/18) or TLR agonists [3, 9]. To activate isolated NK cells, they were challenged with TLR-agonists (LPS and Pam3CSK4), which were previously used to induce inflammation in the liver-on-a-chip model. As functional readout IFN-gamma was used, as one of the most important cytokines secreted by NK cells. IFN-gamma was measured in the supernatant of cell cultures using the cytometric bead array (CBA; BD Biosciences) according to the manufacturer’s protocol. After stimulation of isolated NK cells with LPS (100 ng/ml) und Pam3CSK4 (250 ng/ml) for 16 h, IFN-gamma secretion was significantly increased in comparison to controls reflecting the activation and functionality of isolated NK cells (Fig. 1C).

Integration of NK cells in the liver-on-a-chip model

Human organ on-a-chip models enable the investigation of physiological and pathophysiological processes in vitro in a standardized, multicellular microenvironment. These complex in vitro models offer advantages compared to common in vitro and animal models through scalability and replication of organ-specific microenvironments ex vivo. Furthermore, biochip-embedded models obtain an optimal supply with oxygen and nutrients due to microfluidic media perfusion of the chip [26]. This allows more physiological, multicellular tissue culture improving cellular communication and functional polarization [21]. Conventional cell cultures may overcome some species-dependent barriers, but are limited to reflect physiological conditions in respect to e.g. complex cellular cross-communications [21]. Furthermore, improved culture conditions in organ-on-a-chip models also allow integration of circulating immune cells. Although conventional cell cultures will still remain the mainstay of cellular in vitro analysis, the organ-a-chip technology will help to investigate complex cellular interactions in more physiological conditions [21].

The underlying biochip-based model represents a three-dimensional functional unit of the liver with a permeable membrane serving as a cell culture surface and artificial barrier, allowing microfluidic perfusion and physiological arrangement of cellular structures in vitro. This liver-on-a-chip model used for our analysis was previously established and characterized, showing functional improvements compared to conventional in vitro culture [24, 27]. The so called multi-organ-tissue-flow (MOTiF) biochip, which is manufactured using a cyclic olefin copolymer (COC), was purchased from ChipShop GmbH (Jena, Germany) as previously described [25, 26]. Briefly, the biochip design with two channels and one 12 µm thick PET membrane (pore diameter 8 µm with a density of 2 × 105 pores/cm2) allows precise control of flow conditions maintained through perfusion of media through silicon tubes connected to a peristaltic pump. To reduce bubble formation in the biochip, ramping structures were introduced and chips were hydrophilized by oxygen plasma treatment. Top and bottom side of the chip and channel structures were sealed by a 140 µm thick COC film. Prior to seeding of all cells, biochips were equilibrated overnight by media perfusion of the upper (vascular/endothelial layer) and lower (epithelial layer) channel of the chip for 24 h. The human liver-on-a-chip was assembled by seeding of endothelial cells (human umbilical vein endothelial cells) as well as resident primary macrophages (vascular layer, upper channel) and hepatocytes (HepaRG) as well as stellate cells (LX2; epithelial layer, lower channel) as previously described [27]. In human livers, NK cells are located next to macrophages, lymphocytes and myeloid cells in the endothelial layer, leading to close interactions of these immune cells [4, 32]. To mimic the physiological state, isolated blood derived NK cells were added to the vascular layer in a ratio of one to two, NK cells to Kupffer cells, respectively. The upper, vascular chamber was continuously perfused with media mimicking the blood flow, whereas the bottom, epithelial chamber was kept static with daily media exchange.

Perfusion and stimulation of liver-on-a-chip model comprising NK cells

The main goal of this project was, to integrate functional NK cells into the existing liver-on-a-chip model. Therefore, freshly isolated blood NK cells were seeded as described into the liver-on-a-chip model. After 48 h of perfusion, the presence of NK cells in the endothelial cell layer was verified by immunofluorescence staining, analysis of the maturation and cytotoxicity marker NKp46 (Fig. 2A, B), and flow cytometry analysis. Hence, cells of the vascular layer were detached from the membrane and the NK cell population was analyzed by flow cytometry using CD3 (Horizon VE450), CD16 (PE), and CD56 (PE-Cy7). The NK cell population was defined by expression of NK cell surface markers CD56 and absence of CD3 (Fig. 2C). To prove that NK cells were still responsive to inflammatory triggers, the liver-on-a-chip model was cultured for a maximum 48 h with and without TLR-agonists (LPS/Pam3CSK4) stimulation. Under physiological conditions, the liver is constantly exposed to antigens, pathogens, and tumor cells, entering the liver via the blood flow to the liver sinusoids. The immune and endothelial cells are first exposed to these agents, and thus build the first line of defense [32, 33]. Comparable to the NK cells stimulation in monocultures, the hepatic injury was induced by TLR stimulation through continuous perfusion of the upper chamber consisting of endothelial cells, NK cells and macrophages. Interestingly, a trend of increased expression of NKp46 after TLR stimulation compared to control was observed (p = 0.09, Fig. 3A), indicating an increased number and associated cytotoxic activity of NK cells in the model. Furthermore, the pro-inflammatory cytokines IL-6 (p = 0.005) and TNF-alpha (p = 0.03) were significantly increased after TLR stimulation compared to controls (Fig. 3B, C), potentially due to secretion by NK cells and macrophages. Nevertheless, IFN-gamma secretion was under the lower detection limit of the assay in the liver-on-a-chip-model after 16 and 24 h of TLR agonist stimulation, respectively. These low IFN-gamma levels may be a result of NK cell differentiation or protective feedback mechanisms from other cell types in the liver-on-a-chip model leading to different NK cell function and cytokine secretion [1, 34]. TLR-stimulation of the liver-on-a-chip model further led to a disruption of the endothelial barrier assessed by the significantly reduced expression of VE-cadherin compared to the control condition (p = 0.05, Fig. 3D, E).

Fig. 2
figure 2

Representative immunofluorescence image (A) of the endothelial layer including NK cells and without NK cells (B) stained with cytotoxicity and maturation marker NKp46 (green = NKp46, blue = DAPI, orange = VE-cadherin). Detection of CD56+/CD3 NK cells by FACS isolated from the endothelial layer of the liver-on-a-chip model after 48 h of perfusion (C)

Fig. 3
figure 3

Stimulation with TLR-receptor agonists (LPS and Pam3CSK4) revealed a trend to higher expression of the maturation and cytotoxicity marker NKp46 on the endothelial layer of the liver-on-a-chip model (A). Increased TNF-alpha (B) and IL-6 (C) secretion of liver-on-a-chip including NK cells after 24 h perfusion with TLR-receptor agonists. Liver injury, reflected by endothelial cell layer disruption, assessed by immunofluorescence of VE-cadherin, was significantly increased after TLR-receptor agonist stimulation than in controls (D). Representative immunofluorescence image (E) showing disruption of the endothelial layer after application of the TLR receptor agonists. Figures AD present data of at least three independent experiments, p-values are assessed by Mann Whitney test

In conclusion, our data highlights the first successful approach of integrated viable und functional NK cells isolated from human blood in a liver-on-a-chip model. NK cells and other immune cells, e.g. macrophages, have close interactions during inflammatory and immunological processes in the liver. The aim of this investigation was to establish an in vitro model including NK cells representing a functional hepatic unit as close as possible to the in vivo situation. Our study highlights the potential of organ-on-a-chip models to specifically analyze the contribution of isolated immune cells under physiological conditions, which is currently only possible using animal models or human studies. Further functional analysis and cell interactions of NK cells in the liver-on-a-chip model need to be conducted to provide a more comprehensive analysis of pathophysiological mechanisms. Future applications could be to investigate NK cells driven immunological processes during hepatic inflammation (sterile or bacterial induced) with the advantage of a controlled and standardized in vitro model comprising of human cells.

Limitations

NK cells were successfully isolated from human blood and human livers, but so far only human blood NK cells and not liver NK cells were integrated into the existing liver-on-a-chip model. Previous reports showed functional differences between liver-resident NK cell populations and recruited NK cells from the blood invading the liver [7, 35]. Follow-up studies are necessary to determine the differences between different NK-cell populations in our liver-on-a-chip model. So far, we could show, that the transfer of freshly isolated blood-derived NK-cells into the liver-on-a-chip-model was possible, but further functional investigations and differentiation or maturation processes of NK cells [36] were not investigated. In addition, studies without resident macrophages and only NK cells should elucidate the specific contribution of NK cells in the liver during inflammation. These issues are possible cornerstones of future studies using this presented model.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Change history

  • 11 December 2023

    “Open Access funding enabled and organized by Projekt DEAL” removed from the funding.

Abbreviations

CD:

Cluster of differentiation

COC:

Cyclic olefin copolymers

FACS:

Fluorescence-activated cell sorting

IL:

Interleukin

LPS:

Lipopolysaccharides

MOTiF:

Multi-organ-tissue-flow

NK:

Natural killer

PBMCs:

Peripheral blood mononuclear cells

TNF:

Tumor necrosis factor

TLR:

Toll-like receptor

References

  1. Sun H, Sun C, Tian Z, Xiao W. NK cells in immunotolerant organs. Cell Mol Immunol. 2013;10(3):202–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Seki S, Habu Y, Kawamura T, Takeda K, Dobashi H, Ohkawa T, Hiraide H. The liver as a crucial organ in the first line of host defense: the roles of Kupffer cells, natural killer (NK) cells and NK1.1 Ag+ T cells in T helper 1 immune responses. Immunol Rev. 2000;174:35–46.

    Article  CAS  PubMed  Google Scholar 

  3. Mestre-Duran C, Martin-Cortazar C, Garcia-Solis B, Pernas A, Pertinez L, Galan V, Sisinni L, Clares-Villa L, Navarro-Zapata A, Al-Akioui K, et al. Ruxolitinib does not completely abrogate the functional capabilities of TLR4/9 ligand-activated NK cells. Front Immunol. 2022;13:1045316.

    Article  CAS  PubMed  Google Scholar 

  4. Suh YG, Jeong WI. Hepatic stellate cells and innate immunity in alcoholic liver disease. World J Gastroenterol. 2011;17(20):2543–51.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Yamagiwa S, Kamimura H, Ichida T. Natural killer cell receptors and their ligands in liver diseases. Med Mol Morphol. 2009;42(1):1–8.

    Article  CAS  PubMed  Google Scholar 

  6. Krueger PD, Lassen MG, Qiao H, Hahn YS. Regulation of NK cell repertoire and function in the liver. Crit Rev Immunol. 2011;31(1):43–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Carrega P, Ferlazzo G. Natural killer cell distribution and trafficking in human tissues. Front Immunol. 2012;3:347.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. Functions of natural killer cells. Nat Immunol. 2008;9(5):503–10.

    Article  CAS  PubMed  Google Scholar 

  9. Fahrner R, Trochsler M, Corazza N, Graubardt N, Keogh A, Candinas D, Brunner T, Stroka D, Beldi G. Tumor necrosis factor-related apoptosis-inducing ligand on NK cells protects from hepatic ischemia-reperfusion injury. Transplantation. 2014;97(11):1102–9.

    Article  CAS  PubMed  Google Scholar 

  10. Palano MT, Cucchiara M, Gallazzi M, Riccio F, Mortara L, Gensini GF, Spinetti G, Ambrosio G, Bruno A. When a friend becomes your enemy: natural killer cells in atherosclerosis and atherosclerosis-associated risk factors. Front Immunol. 2021;12:798155.

    Article  CAS  PubMed  Google Scholar 

  11. Graubardt N, Fahrner R, Trochsler M, Keogh A, Breu K, Furer C, Stroka D, Robson SC, Slack E, Candinas D, et al. Promotion of liver regeneration by natural killer cells in a murine model is dependent on extracellular adenosine triphosphate phosphohydrolysis. Hepatology. 2013;57(5):1969–79.

    Article  CAS  PubMed  Google Scholar 

  12. Di Santo JP. Natural killer cell developmental pathways: a question of balance. Annu Rev Immunol. 2006;24:257–86.

    Article  PubMed  Google Scholar 

  13. Di Santo JP. Natural killer cells: diversity in search of a niche. Nat Immunol. 2008;9(5):473–5.

    Article  PubMed  Google Scholar 

  14. Campbell JS, Prichard L, Schaper F, Schmitz J, Stephenson-Famy A, Rosenfeld ME, Argast GM, Heinrich PC, Fausto N. Expression of suppressors of cytokine signaling during liver regeneration. J Clin Investig. 2001;107(10):1285–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Vivier E, Raulet DH, Moretta A, Caligiuri MA, Zitvogel L, Lanier LL, Yokoyama WM, Ugolini S. Innate or adaptive immunity? The example of natural killer cells. Science. 2011;331(6013):44–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Fausto N. Liver regeneration. J Hepatol. 2000;32(1 Suppl):19–31.

    Article  CAS  PubMed  Google Scholar 

  17. Schwabe RF, Brenner DA. Mechanisms of liver injury. I. TNF-alpha-induced liver injury: role of IKK, JNK, and ROS pathways. Am J Physiol Gastrointest liver Physiol. 2006;290(4):G583-589.

    Article  CAS  PubMed  Google Scholar 

  18. Tu Z, Bozorgzadeh A, Pierce RH, Kurtis J, Crispe IN, Orloff MS. TLR-dependent cross talk between human kupffer cells and NK cells. J Exp Med. 2008;205(1):233–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Aarsund M, Segers FM, Wu Y, Inngjerdingen M. Comparison of characteristics and tumor targeting properties of extracellular vesicles derived from primary NK cells or NK-cell lines stimulated with IL-15 or IL-12/15/18. Cancer Immunol, Immunother CII. 2022;71(9):2227–38.

    Article  CAS  PubMed  Google Scholar 

  20. Oka N, Markova T, Tsuzuki K, Li W, El-Darawish Y, Pencheva-Demireva M, Yamanishi K, Yamanishi H, Sakagami M, Tanaka Y, et al. IL-12 regulates the expansion, phenotype, and function of murine NK cells activated by IL-15 and IL-18. Cancer Immunol, Immunother CII. 2020;69(9):1699–712.

    Article  CAS  PubMed  Google Scholar 

  21. Groger M, Rennert K, Giszas B, Weiss E, Dinger J, Funke H, Kiehntopf M, Peters FT, Lupp A, Bauer M, et al. Monocyte-induced recovery of inflammation-associated hepatocellular dysfunction in a biochip-based human liver model. Sci Rep. 2016;6:21868.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Walther BK, Rajeeva Pandian NK, Gold KA, Kiliç ES, Sama V, Gu J, Gaharwar AK, Guiseppi-Elie A, Cooke JP, Jain A. Mechanotransduction-on-chip: vessel-chip model of endothelial YAP mechanobiology reveals matrix stiffness impedes shear response. Lab Chip. 2021;21(9):1738–51. https://doi.org/10.1039/d0lc01283a.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Milani N, Parrott N, Ortiz Franyuti D, Godoy P, Galetin A, Gertz M, Fowler S. Application of a gut-liver-on-a-chip device and mechanistic modelling to the quantitative in vitro pharmacokinetic study of mycophenolate mofetil. Lab Chip. 2022;22:2853.

    Article  CAS  PubMed  Google Scholar 

  24. Raasch M, Rennert K, Jahn T, Peters S, Henkel T, Huber O, Schulz I, Becker H, Lorkowski S, Funke H, et al. Microfluidically supported biochip design for culture of endothelial cell layers with improved perfusion conditions. Biofabrication. 2015;7(1):015013.

    Article  PubMed  Google Scholar 

  25. Rennert K, Steinborn S, Groger M, Ungerbock B, Jank AM, Ehgartner J, Nietzsche S, Dinger J, Kiehntopf M, Funke H, et al. A microfluidically perfused three dimensional human liver model. Biomaterials. 2015;71:119–31.

    Article  CAS  PubMed  Google Scholar 

  26. Fahrner R, Moller A, Press AT, Kortgen A, Kiehntopf M, Rauchfuss F, Settmacher U, Mosig AS. Short-term treatment with taurolidine is associated with liver injury. BMC Pharmacol Toxicol. 2017;18(1):61.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Doherty DG, Norris S, Madrigal-Estebas L, McEntee G, Traynor O, Hegarty JE, O’Farrelly C. The human liver contains multiple populations of NK cells, T cells, and CD3+CD56+ natural T cells with distinct cytotoxic activities and Th1, Th2, and Th0 cytokine secretion patterns. J Immunol. 1999;163(4):2314–21.

    Article  CAS  PubMed  Google Scholar 

  28. Morsy MA, Norman PJ, Mitry R, Rela M, Heaton ND, Vaughan RW. Isolation purification and flow cytometric analysis of human intrahepatic lymphocytes using an improved technique. Lab Investig J Tech Methods Pathol. 2005;85(2):285–96.

    Article  Google Scholar 

  29. Gotthardt D, Prchal-Murphy M, Seillet C, Glasner A, Mandelboim O, Carotta S, Sexl V, Putz EM. NK cell development in bone marrow and liver: site matters. Genes Immun. 2014;15(8):584–7.

    Article  CAS  PubMed  Google Scholar 

  30. Hudspeth K, Donadon M, Cimino M, Pontarini E, Tentorio P, Preti M, Hong M, Bertoletti A, Bicciato S, Invernizzi P, et al. Human liver-resident CD56(bright)/CD16(neg) NK cells are retained within hepatic sinusoids via the engagement of CCR5 and CXCR6 pathways. J Autoimmun. 2016;66:40–50.

    Article  CAS  PubMed  Google Scholar 

  31. Mosig AS. Organ-on-chip models: new opportunities for biomedical research. Future Sci OA. 2017;3(2):FSO130.

    Article  PubMed  Google Scholar 

  32. Peng H, Wisse E, Tian Z. Liver natural killer cells: subsets and roles in liver immunity. Cell Mol Immunol. 2016;13(3):328–36.

    Article  CAS  PubMed  Google Scholar 

  33. Fahrner R, Dondorf F, Ardelt M, Settmacher U, Rauchfuss F. Role of NK, NKT cells and macrophages in liver transplantation. World J Gastroenterol. 2016;22(27):6135–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Gao B, Radaeva S, Park O. Liver natural killer and natural killer T cells: immunobiology and emerging roles in liver diseases. J Leukoc Biol. 2009;86(3):513–28. https://doi.org/10.1189/JLB.0309135.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Gregoire C, Chasson L, Luci C, Tomasello E, Geissmann F, Vivier E, Walzer T. The trafficking of natural killer cells. Immunol Rev. 2007;220:169–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Peng H, Tian Z. Re-examining the origin and function of liver-resident NK cells. Trends Immunol. 2015;36(5):293–9.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to acknowledge the technical assistance of Swen Carlstedt, Melanie Ulrich, and Nora Mosig (Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Germany).

Funding

This study obtained funding from the Center for Sepsis Control and Care (CSCC), Jena University Hospital and by the Federal Ministry for Education and Research (Bundesministerium für Bildung und Forschung, BMBF) for salary, material and institutional equipment.

Author information

Authors and Affiliations

Authors

Contributions

Study conception and design: RF, MG, ASM. Acquisition of data: RF, MG. Analysis and interpretation of data: RF, MG, US, ASM. Drafting of manuscript: RF. Critical revision of manuscript: MG, US, ASM. All authors have read and approved the final version of this manuscript.

Corresponding author

Correspondence to René Fahrner.

Ethics declarations

Ethical approval and consent to participate

All research protocols were carried out in accordance with the National Institutes of Health guidelines and approval of the ethical committee approval was given from the University of Jena, Germany (No. 4282–12/14) and donors gave informed consent for umbilical cord donation for the endothelial cell isolation.

Consent for publication

Not applicable for this study.

Competing interests

The authors declare that they have no conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Figure S1.

Representation immunofluorescence images of NK cells in low magnification stained with cytotoxicity and maturation marker NKp46 in monoculture of isolation blood NK cells (green=NKp46, blue=DAPI, nuclei).

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fahrner, R., Gröger, M., Settmacher, U. et al. Functional integration of natural killer cells in a microfluidically perfused liver on-a-chip model. BMC Res Notes 16, 285 (2023). https://doi.org/10.1186/s13104-023-06575-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13104-023-06575-w

Keywords