Early Differentiation of Human CD11c+NK Cells with γδ T Cell Activation Properties Is Promoted by Dialyzable Leukocyte Extracts

Dalia Ramírez-Ramírez, Eduardo Vadillo, Lourdes Andrea Arriaga-Pizano, Héctor Mayani, Sergio Estrada-Parra, Marco Antonio Velasco-Velázquez, Sonia Mayra Pérez-Tapia, Rosana Pelayo, Dalia Ramírez-Ramírez, Eduardo Vadillo, Lourdes Andrea Arriaga-Pizano, Héctor Mayani, Sergio Estrada-Parra, Marco Antonio Velasco-Velázquez, Sonia Mayra Pérez-Tapia, Rosana Pelayo

Abstract

Reconstitution of the hematopoietic system during immune responses and immunological and neoplastic diseases or upon transplantation depends on the emergent differentiation of hematopoietic stem/progenitor cells within the bone marrow. Although in the last decade the use of dialyzable leukocyte extracts (DLE) as supportive therapy in both infectious and malignant settings has increased, its activity on the earliest stages of human hematopoietic development remains poorly understood. Here, we have examined the ability of DLE to promote replenishment of functional lymphoid lineages from CD34+ cells. Our findings suggest that DLE increases their differentiation toward a conspicuous CD56+CD16+CD11c+ NK-like cell population endowed with properties such as IFNy production, tumor cell cytotoxicity, and the capability of inducing γδ T lymphocyte proliferation. Of note, long-term coculture controlled systems showed the bystander effect of DLE-stromal cells by providing NK progenitors with signals to overproduce this cell subset. Thus, by direct effect on progenitor cells and through activation and remodeling of the supporting hematopoietic microenvironment, DLE may contribute a robust innate immune response by promoting the emerging lymphopoiesis of functional CD11c+ NK cells in a partially TLR-related manner. Unraveling the identity and mechanisms of the involved DLE components may be fundamental to advance the NK cell-based therapy field.

Figures

Figure 1
Figure 1
Dialyzable leukocyte extracts (DLE) promote proliferation of primitive hematopoietic CD34+ cells from neonatal and adult sources. CD34+ cells from adult bone marrow (ABM) and umbilical cord blood (UCB) were enriched and cultured for 72 hours with DLE Transferon and BrdU. Cells were stained for the identification of CD34+ cell progenitors followed by intracellular staining of BrdU. Multiparametric flow cytometry was performed in a FACSCanto II and the cell frequencies of BrdU+ cells calculated. Data are representative of 3 independent experiments. P < 0.05; ∗∗P < 0.01.
Figure 2
Figure 2
CD56+CD11c+ NK-like cells expeditiously develop from early lymphoid progenitors upon DLE stimulation. Upon 24 hours of DLE Transferon stimulation, the whole fraction of CD34+ cells (a and b) or purified primitive cells, including hematopoietic stem cells (HSC), multipotent progenitors (MPP), and early lymphoid progenitors (ELP) (a), were cocultured with MS-5 stromal cells for 30 days under lymphoid lineage conditions. CD56+CD11c+ NK-like cell production was investigated by flow cytometry and reported as cell frequencies (a) or yield per input progenitor (b and c). For comparison, unstimulated cultures were considered as control conditions. DLE, dialyzable leukocyte extracts Transferon. ∗∗P < 0.01.
Figure 3
Figure 3
Stromal cell activation by DLE increases the emergent innate immune cell differentiation. MS-5 stromal cells were exposed or not to DLE Transferon 24 hours before coculturing with CD34+ hematopoietic stem and progenitor cells (HSPC) that were pretreated 24 hours with DLE Transferon (a). All conditions were placed in lymphoid lineage cocultures for 30 days followed by flow cytometry analyses of CD56+ NK cells and CD11c+ DC production. Yields per input progenitor were tabulated to show the positive contribution of activated stromal cells to innate differentiation (b). DLE, dialyzable leukocyte extracts Transferon. P < 0.05; ∗∗P < 0.01; ∗∗∗P < 0.001.
Figure 4
Figure 4
Preferential production of functional CD11c+CD16+ NK-like cells promoted by hematopoietic-stromal cell communication responding to DLE. MS-5 stromal cells were exposed or not to DLE Transferon 24 hours before coculturing with DLE Transferon-pretreated CD34+ hematopoietic stem and progenitor cells (HSPC), as described in Figure 3(a). All conditions were placed in lymphoid lineage cocultures for 30 days followed by flow cytometry analyses, where the indicated gates were used to discriminate CD56+CD11c+ and CD56+CD16+CD11c+NK cell frequencies (a). Yields per input progenitor were tabulated to record the significant variations by stromal or hematopoietic cells exposure to DLE Transferon (b). ∗∗∗P < 0.001.
Figure 5
Figure 5
Effector functions of CD11c+ NK cells differentiated from DLE-induced lymphoid progenitor cells include tumor cytotoxicity and IFN-gamma production. NK cytotoxicity function was evaluated on CFSE+ K562 tumor cells as target of NK-like cells derived from DLE stimulation (DLE-NK) (a). We used 7-aminoactinomycin D (7-AAD) to evidence the frequency of CFSE+7-AAD+ killed target cells. Production of IFN-gamma by CD11c−CD56+ and CD11c+CD56+ NK cells was detected by intracellular staining (b).
Figure 6
Figure 6
Activation of γδ T cells as a result of DLE-associated CD11c+ NK cell emergent production. γδ T cells from healthy donors were cocultured with the DLE-derived CD11c+ NK cell at a ratio of 2 : 1 CD56+CD11c+ NK : γδ T cell (a). Both NK and γδ T cell populations were highly purified by flow cytometry sorting before setting the proliferation assay (b). γδ T lymphocyte proliferation was assessed at 72 hours by dilution of Cell Trace Violet (CTV) dye (c).
Figure 7
Figure 7
Emergent CD11c+ NK cell production contributing the adjuvant immune-surveillance effects of DLE Transferon: a proposed model. Dialyzable leukocyte extracts Transferon promote the early differentiation of functional CD11c+ NK cells endowed with the capabilities of tumor cell cytotoxicity, IFNy production, and γδ T lymphocyte proliferation induction, which may in turn contribute to innate immune responses against virus-infected or tumor cells. The potential involved mechanisms require further investigation.

References

    1. Kobayashi H., Suda T., Takubo K. How hematopoietic stem/progenitors and their niche sense and respond to infectious stress. Experimental Hematology. 2016;44(2):92–100. doi: 10.1016/j.exphem.2015.11.008.
    1. Baldridge M. T., King K. Y., Goodell M. A. Inflammatory signals regulate hematopoietic stem cells. Trends in Immunology. 2011;32(2):57–65. doi: 10.1016/j.it.2010.12.003.
    1. Vadillo E., Dorantes-Acosta E., Pelayo R. Regulation of hematopoietic stem/progenitor cell development by inflammation cues. In: Pérez-Martínez L., Pedraza-Alva G., Osorio E. F., editors. Molecular Aspects of Inflammation. 2014. pp. 71–87. (Res Signpost 37/661 (2)).
    1. Vadillo E., Pelayo R. Adult NK lineage development in humans is strengthened upon emergency. Inflammation and Cell Signaling. 2014;1(3, article e269) doi: 10.14800/ics.269.
    1. Welner R. S., Pelayo R., Kincade P. W. Evolving views on the genealogy of B cells. Nature Reviews Immunology. 2008;8(2):95–106. doi: 10.1038/nri2234.
    1. Yáñez A., Goodridge H. S., Gozalbo D., Gil M. L. TLRs control hematopoiesis during infection. European Journal of Immunology. 2013;43(10):2526–2533. doi: 10.1002/eji.201343833.
    1. Nagai Y., Garrett K. P., Ohta S., et al. Toll-like receptors on hematopoietic progenitor cells stimulate innate immune system replenishment. Immunity. 2006;24(6):801–812. doi: 10.1016/j.immuni.2006.04.008.
    1. Welner R. S., Pelayo R., Nagai Y., et al. Lymphoid precursors are directed to produce dendritic cells as a result of TLR9 ligation during herpes infection. Blood. 2008;112(9):3753–3761. doi: 10.1182/blood-2008-04-151506.
    1. Vadillo E., Dorantes-Acosta E., Arriaga-Pizano L., et al. Adult, but not neonatal, human lymphoid progenitors respond to TLR9 ligation by producing functional NK-like cells. Experimental Hematology. 2014;42(7):562–573. doi: 10.1016/j.exphem.2014.03.008.
    1. Vadillo E., Pelayo R. Toll-like receptors in development and function of the hematopoietic system. Revista de Investigación Clínica. 2012;64(5):461–476.
    1. Vilchis-Ordoñez A., Contreras-Quiroz A., Vadillo E., et al. Bone marrow cells in acute lymphoblastic leukemia create a proinflammatory microenvironment influencing normal hematopoietic differentiation fates. BioMed Research International. 2015;2015:14. doi: 10.1155/2015/386165.386165
    1. Kobayashi H., Kobayashi C. I., Nakamura-Ishizu A., et al. Bacterial c-di-GMP affects hematopoietic Stem/progenitors and their niches through STING. Cell Reports. 2015;11(1):71–84. doi: 10.1016/j.celrep.2015.02.066.
    1. Viza D., Fudenberg H. H., Palareti A., Ablashi D., De Vinci C., Pizza G. Transfer factor: an overlooked potential for the prevention and treatment of infectious diseases. Folia Biologica. 2013;59(2):53–67.
    1. Medina-Rivero E., Merchand-Reyes G., Pavón L., et al. Batch-to-batch reproducibility of Transferon™. Journal of Pharmaceutical and Biomedical Analysis. 2014;88:289–294. doi: 10.1016/j.jpba.2013.09.004.
    1. García-Hernández U., Robledo-Ávila F. H., Álvarez-Jiménez V. D., et al. Dialyzable leukocyte extracts activate TLR-2 on monocytes. Natural Product Communications. 2014;9(6):853–856.
    1. Estrada-Parra S., Nagaya A., Serrano E., et al. Comparative study of transfer factor and acyclovir in the treatment of herpes zoster. International Journal of Immunopharmacology. 1998;20(10):521–535. doi: 10.1016/s0192-0561(98)00031-9.
    1. Ojeda M. O., van't Veer C., Fernández-Ortega C. B., Araña Rosainz M. D. J., Buurman W. A. Dialyzable leukocyte extract differentially regulates the production of TNFα, IL-6, and IL-8 in bacterial component-activated leukocytes and endothelial cells. Inflammation Research. 2005;54(2):74–81. doi: 10.1007/s00011-004-1326-5.
    1. Robles-Contreras A., Vizuet L., Rivera E., et al. Down regulation of IL-8 and IL-6 in human limbal epithelial cells cultured with human dialyzable leukocyte extracts. Revista Alergia Mexico. 2011;58(3):147–154.
    1. Berrón-Pérez R., Chávez-Sánchez R., Estrada-García I., et al. Indications, usage, and dosage of the transfer factor, S. Espinosa-Padilla, R. Cortez-Gómez, E. Serrano-Miranda, R. Ondarza-Aguilera, S. M. Pérez-Tapia, B. Pineda Olvera, M. C. Jiménez-Martínez, A. Portugués, A. Rodríguez, L. Cano, P. U. Pacheco, J. Barrientos, R. Chacón- Salinas, J. Serafín, P. Mendez, A. Monges, E. Cervantes and S. Estrada-Parra. Revista Alergia Mexico. 2007;54(4):134–139.
    1. Fabre R. A., Pérez-Tapia S. M., Aguilar L. D., et al. Transfer factors as immunotherapy and supplement of chemotherapy in experimental pulmonary tuberculosis. Clinical and Experimental Immunology. 2004;136(2):215–223. doi: 10.1111/j.1365-2249.2004.02454.x.
    1. Flores Sandoval G., Gómez Vera J., Orea Solano M., et al. Factor de transferencia como inmunomodulador específico en el tratamiento de la dematitis atópica moderada a severa. Revista Alergia México. 2005;52(6):215–220.
    1. Salinas-Jazmín N., Estrada-Parra S., Becerril-García M. A., et al. Herpes murine model as a biological assay to test dialyzable leukocyte extracts activity. Journal of Immunology Research. 2015;2015:9. doi: 10.1155/2015/146305.146305
    1. Bryceson Y. T., Fauriat C., Nunes J. M., et al. Functional analysis of human NK cells by flow cytometry. Methods in Molecular Biology. 2010;612:335–352. doi: 10.1007/978-1-60761-362-6_23.
    1. Di Santo J. P. Natural killer cells: diversity in search of a niche. Nature Immunology. 2008;9(5):473–475. doi: 10.1038/ni.f.201.
    1. de Maria A., Bozzano F., Cantoni C., Moretta L. Revisiting human natural killer cell subset function revealed cytolytic CD56dimCD16+ NK cells as rapid producers of abundant IFN-γ on activation. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(2):728–732. doi: 10.1073/pnas.1012356108.
    1. Della Chiesa M., Marcenaro E., Sivori S., Carlomagno S., Pesce S., Moretta A. Human NK cell response to pathogens. Seminars in Immunology. 2014;26(2):152–160. doi: 10.1016/j.smim.2014.02.001.
    1. Burt B. M., Plitas G., Stableford J. A., et al. CD11c identifies a subset of murine liver natural killer cells that responds to adenoviral hepatitis. Journal of Leukocyte Biology. 2008;84(4):1039–1046. doi: 10.1189/jlb.0408256.
    1. Pillarisetty V. G., Katz S. C., Bleier J. I., Shah A. B., DeMatteo R. P. Natural killer dendritic cells have both antigen presenting and lytic function and in response to CpG produce IFN-γ via autocrine IL-12. Journal of Immunology. 2005;174(5):2612–2618. doi: 10.4049/jimmunol.174.5.2612.
    1. Plitas G., Chaudhry U. I., Kingham T. P., Raab J. R., DeMatteo R. P. NK dendritic cells are innate immune responders to Listeria monocytogenes infection. The Journal of Immunology. 2007;178(7):4411–4416. doi: 10.4049/jimmunol.178.7.4411.
    1. Chaudhry U. I., Plitas G., Burt B. M., Kingham T. P., Raab J. R., DeMatteo R. P. NK dendritic cells expanded in IL-15 exhibit antitumor responses in vivo. Journal of Immunology. 2007;179(7):4654–4660. doi: 10.4049/jimmunol.179.7.4654.
    1. Chaudhry U. I., Kingham T. P., Plitas G., Katz S. C., Raab J. R., DeMatteo R. P. Combined stimulation with interleukin-18 and CpG induces murine natural killer dendritic cells to produce IFN-γ and inhibit tumor growth. Cancer Research. 2006;66(21):10497–10504. doi: 10.1158/0008-5472.can-06-1908.
    1. Chastain E. M. L., Getts D. R., Miller S. D. Deficient natural killer dendritic cell responses underlay the induction of Theiler's virus-induced autoimmunity. mBio. 2015;6(4) doi: 10.1128/mbio.01175-15.e01175-15
    1. Blasius A. L., Barchet W., Cella M., Colonna M. Development and function of murine B220+CD11c+NK1.1+ cells identify them as a subset of NK cells. The Journal of Experimental Medicine. 2007;204(11):2561–2568. doi: 10.1084/jem.20070991.
    1. Chan C. W., Crafton E., Fan H.-N., et al. Interferon-producing killer dendritic cells provide a link between innate and adaptive immunity. Nature Medicine. 2006;12(2):207–213. doi: 10.1038/nm1352.
    1. Taieb J., Chaput N., Ménard C., et al. A novel dendritic cell subset involved in tumor immunosurveillance. Nature Medicine. 2006;12(2):214–219. doi: 10.1038/nm1356.
    1. Welner R. S., Pelayo R., Garrett K. P., et al. Interferon-producing killer dendritic cells (IKDCs) arise via a unique differentiation pathway from primitive c-kitHiCD62L+ lymphoid progenitors. Blood. 2007;109(11):4825–4931. doi: 10.1182/blood-2006-08-043810.
    1. GeurtsvanKessel C. H., Bergen I. M., Muskens F., et al. Both conventional and interferon killer dendritic cells have antigen-presenting capacity during influenza virus infection. PLoS ONE. 2009;4(9) doi: 10.1371/journal.pone.0007187.e7187
    1. Aranami T., Miyake S., Yamamura T. Differential expression of CD11c by peripheral blood NK cells reflects temporal activity of multiple sclerosis. The Journal of Immunology. 2006;177(8):5659–5667. doi: 10.4049/jimmunol.177.8.5659.
    1. Nagasawa T. CXCL12/SDF-1 and CXCR4. Frontiers in Immunology. 2015;6, article 301 doi: 10.3389/fimmu.2015.00301.
    1. Purizaca J., Meza I., Pelayo R. Early lymphoid development and microenvironmental cues in B-cell acute lymphoblastic leukemia. Archives of Medical Research. 2012;43(2):89–101. doi: 10.1016/j.arcmed.2012.03.005.
    1. Noda M., Omatsu Y., Sugiyama T., Oishi S., Fujii N., Nagasawa T. CXCL12-CXCR4 chemokine signaling is essential for NK-cell development in adult mice. Blood. 2011;117(2):451–458. doi: 10.1182/blood-2010-04-277897.
    1. Li W., Okuda A., Yamamoto H., et al. Regulation of development of CD56brightCD11c+ NK-Like cells with helper function by IL-18. PLoS ONE. 2013;8(12) doi: 10.1371/journal.pone.0082586.e82586
    1. Tsuda J., Li W., Yamanishi H., et al. Involvement of CD56brightCD11c+ cells in IL-18-mediated expansion of human γδ T cells. Journal of Immunology. 2011;186(4):2003–2012.
    1. Ribot J. C., DeBarros A., Silva-Santos B. Searching for ‘signal 2’: costimulation requirements of γδ T cells. Cellular and Molecular Life Sciences. 2011;68(14):2345–2355. doi: 10.1007/s00018-011-0698-2.
    1. Wu Y.-L., Ding Y.-P., Tanaka Y., et al. γδ T cells and their potential for immunotherapy. International Journal of Biological Sciences. 2014;10(2):119–135. doi: 10.7150/ijbs.7823.
    1. Todaro M., D'Asaro M., Caccamo N., et al. Efficient killing of human colon cancer stem cells by γδ T lymphocytes. The Journal of Immunology. 2009;182(11):7287–7296. doi: 10.4049/jimmunol.0804288.
    1. Sugie T., Murata-Hirai K., Iwasaki M., et al. Zoledronic acid-induced expansion of γδ T cells from early-stage breast cancer patients: effect of IL-18 on helper NK cells. Cancer Immunology, Immunotherapy. 2013;62(4):677–687. doi: 10.1007/s00262-012-1368-4.
    1. Fujishima N., Hirokawa M., Fujishima M., et al. Skewed T cell receptor repertoire of Vδ1+ γδ T lymphocytes after human allogeneic haematopoietic stem cell transplantation and the potential role for Epstein-Barr virus-infected B cells in clonal restriction. Clinical & Experimental Immunology. 2007;149(1):70–79. doi: 10.1111/j.1365-2249.2007.03388.x.

Source: PubMed

3
Subscribe