Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 118(22)2021 06 01.
Article in English | MEDLINE | ID: mdl-34050021

ABSTRACT

Natural killer (NK) cells are major antileukemic immune effectors. Leukemic blasts have a negative impact on NK cell function and promote the emergence of phenotypically and functionally impaired NK cells. In the current work, we highlight an accumulation of CD56-CD16+ unconventional NK cells in acute myeloid leukemia (AML), an aberrant subset initially described as being elevated in patients chronically infected with HIV-1. Deep phenotyping of NK cells was performed using peripheral blood from patients with newly diagnosed AML (n = 48, HEMATOBIO cohort, NCT02320656) and healthy subjects (n = 18) by mass cytometry. We showed evidence of a moderate to drastic accumulation of CD56-CD16+ unconventional NK cells in 27% of patients. These NK cells displayed decreased expression of NKG2A as well as the triggering receptors NKp30 and NKp46, in line with previous observations in HIV-infected patients. High-dimensional characterization of these NK cells highlighted a decreased expression of three additional major triggering receptors required for NK cell activation, NKG2D, DNAM-1, and CD96. A high proportion of CD56-CD16+ NK cells at diagnosis was associated with an adverse clinical outcome and decreased overall survival (HR = 0.13; P = 0.0002) and event-free survival (HR = 0.33; P = 0.018) and retained statistical significance in multivariate analysis. Pseudotime analysis of the NK cell compartment highlighted a disruption of the maturation process, with a bifurcation from conventional NK cells toward CD56-CD16+ NK cells. Overall, our data suggest that the accumulation of CD56-CD16+ NK cells may be the consequence of immune escape from innate immunity during AML progression.


Subject(s)
Flow Cytometry/methods , Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Antigens, CD/immunology , Humans , Immunophenotyping , Lymphocyte Activation/immunology , Remission Induction , Treatment Outcome
2.
Eur J Immunol ; 46(7): 1758-69, 2016 07.
Article in English | MEDLINE | ID: mdl-27119199

ABSTRACT

The immunological attributes of stem cell grafts play an important role in the outcome of allogeneic stem cell transplants. Currently, ex vivo manipulation techniques such as bulk T-cell depletion or positive selection of CD34(+) cells are utilized to improve the immunological attributes of grafts and minimize the potential for graft-versus-host disease (GvHD). Here, we demonstrate a novel graft engineering technique, which utilizes the immunomodulatory drug FTY720 for in vivo depletion of naïve T (TN ) cells from donor G-CSF-mobilized grafts without ex vivo manipulation. We show that treatment of donor mice with FTY720 during mobilization depletes grafts of TN cells and prevents lethal GvHD following transplantation in a major mismatch setting. Importantly, both stem cells and NK cells are retained in the FTY720-treated grafts. FTY720 treatment does not negatively affect the engraftment potential of stem cells as demonstrated in our congenic transplants or the functionality of NK cells. In addition, potentially useful memory T cells may be retained in the graft. These findings suggest that FTY720 may be used to optimize the immunological attributes of G-CSF-mobilized grafts by removing potentially deleterious TN cells which can contribute to GvHD, and by retaining useful cells which can promote immunity in the recipient.


Subject(s)
Fingolimod Hydrochloride/pharmacology , Genetic Engineering , Graft Survival/drug effects , Graft Survival/immunology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Animals , Genetic Engineering/methods , Graft Survival/genetics , Graft vs Host Disease/etiology , Graft vs Host Disease/pathology , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cells/drug effects , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Depletion , Mice , Mice, Knockout , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplantation, Homologous
3.
Immunology ; 146(1): 173-83, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26094816

ABSTRACT

Natural killer (NK) cells are considered critical components of the innate and adaptive immune responses. Deficiencies in NK cell activity are common, such as those that occur in cancer patients, and they can be responsible for dysfunctional immune surveillance. Persistent oxidative stress is intrinsic to many malignant tumours, and numerous studies have focused on the effects of reactive oxygen species on the anti-tumour activity of NK cells. Indeed, investigations in animal models have suggested that one of the most important thiol-dependent antioxidant enzymes, peroxiredoxin 1 (PRDX1), is essential for NK cell function. In this work, our analysis of the transcriptomic expression pattern of antioxidant enzymes in human NK cells has identified PRDX1 as the most prominently induced transcript out of the 18 transcripts evaluated in activated NK cells. The change in PRDX1 expression was followed by increased expression of two other enzymes from the PRDX-related antioxidant chain: thioredoxin and thioredoxin reductase. To study the role of thiol-dependent antioxidants in more detail, we applied a novel compound, adenanthin, to induce an abrupt dysfunction of the PRDX-related antioxidant chain in NK cells. In human primary NK cells, we observed profound alterations in spontaneous and antibody-dependent NK cell cytotoxicity against cancer cells, impaired degranulation, and a decreased expression of activation markers under these conditions. Collectively, our study pinpoints the unique role for the antioxidant activity of the PRDX-related enzymatic chain in human NK cell functions. Further understanding this phenomenon will prospectively lead to fine-tuning of the novel NK-targeted therapeutic approaches to human disease.


Subject(s)
Diterpenes, Kaurane/pharmacology , Enzyme Inhibitors/pharmacology , Killer Cells, Natural/immunology , Neoplasms/immunology , Peroxiredoxins/antagonists & inhibitors , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Antioxidants , Cell Degranulation/drug effects , Cell Degranulation/immunology , Cell Line, Tumor , Glutathione/analysis , Humans , Oxidative Stress/drug effects , Peroxiredoxins/biosynthesis , Reactive Oxygen Species/metabolism , Thioredoxin Reductase 1/biosynthesis , Thioredoxins/biosynthesis
4.
Biol Blood Marrow Transplant ; 21(3): 429-39, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25579888

ABSTRACT

Recent advances in the development of reduced-intensity conditioning (RIC) have allowed a broader range of patients to access allogeneic hematopoietic stem cell transplantation (HSCT). Reconstitution of an effective immune system post-transplant, including natural killer (NK) cells, is critical for both tumor control and infectious disease control or prevention. The development and functions of NK cells in such settings remain elusive. Here we analyzed NK cell development in HLA-matched HSCT from related or unrelated donors, after RIC that included antithymocyte globulin (N = 45 patients). Our data reveal that NK cells quickly recover after RIC-HSCT, irrespective of donor type. Rapidly re-emerging NK cells, however, remain immature for more than 6 months. Effector functions resemble that of immature NK cells because they poorly produce IFN-γ and TNF-α in response to target cell stimulation, despite a rapid acquisition of degranulation ability and MIP-1ß production. Strikingly, rapid reconstitution of cytokine production correlates with a lower relapse incidence (P = .01) and a better survival rate (P < .0001) at 1 year post-transplant, whereas degranulation capacity was associated with less relapse (P = .05). Our study demonstrates rapid quantitative reconstitution of the NK cell compartment despite administration of potent immune suppressive drugs as part of the conditioning regimen and after transplantation. However, there is a prolonged persistence of functional defects, the correction of which positively correlates with clinical outcome.


Subject(s)
Cytokines/immunology , Hematologic Neoplasms , Hematopoietic Stem Cell Transplantation , Killer Cells, Natural/immunology , Recovery of Function/immunology , Transplantation Conditioning , Adult , Aged , Allografts , Disease-Free Survival , Female , Follow-Up Studies , Hematologic Neoplasms/immunology , Hematologic Neoplasms/mortality , Hematologic Neoplasms/therapy , Histocompatibility Testing , Humans , Male , Middle Aged , Survival Rate , Unrelated Donors
5.
Blood ; 122(6): 922-31, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23692853

ABSTRACT

Vγ9Vδ2 cells, the major γδ T-cell subset in human peripheral blood, represent a T-cell subset that displays reactivity against microbial agents and tumors. The biology of Vγ9Vδ2 T cells remains poorly understood. We show herein that the interaction between B- and T-lymphocyte attenuator (BTLA) and herpesvirus entry mediator (HVEM) is a major regulator of Vγ9Vδ2 T-cell proliferation control. BTLA was strongly expressed at the surface of resting Vγ9Vδ2 T cells and inversely correlated with T-cell differentiation. BTLA-HVEM blockade by monoclonal antibodies resulted in the enhancement of Vγ9Vδ2 T-cell receptor-mediated signaling, whereas BTLA-HVEM interaction led to a decrease in phosphoantigen-mediated proliferation by inducing a partial S-phase arrest. Our data also suggested that BTLA-HVEM might participate in the control of γδ T-cell differentiation. In addition, the proliferation of autologous γδ T cells after exposition to lymphoma cells was dramatically reduced through BTLA-HVEM interaction. These data suggest that HVEM interaction with BTLA may play a role in lymphomagenesis by interfering with Vγ9Vδ2 T-cell proliferation. Moreover, BTLA stimulation of Vγ9Vδ2 T cells appears as a new possible mechanism of immune escape by lymphoma cells.


Subject(s)
Gene Expression Regulation, Neoplastic , Hodgkin Disease/immunology , Lymphoma, Non-Hodgkin/immunology , Receptors, Immunologic/physiology , T-Lymphocyte Subsets/immunology , Antibodies, Monoclonal/immunology , Cell Cycle , Cell Differentiation , Cell Proliferation , Humans , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Immunologic/genetics , Receptors, Tumor Necrosis Factor, Member 14/immunology , S Phase , Signal Transduction , Tumor Escape , Tumor Necrosis Factor-alpha/metabolism
6.
Proc Natl Acad Sci U S A ; 108(8): 3324-9, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-21300876

ABSTRACT

Lymphocytes mediate cytotoxicity by polarized release of the contents of cytotoxic granules toward their target cells. Here, we have studied the role of the calcium release-activated calcium channel ORAI1 in human lymphocyte cytotoxicity. Natural killer (NK) cells obtained from an ORAI1-deficient patient displayed defective store-operated Ca(2+) entry (SOCE) and severely defective cytotoxic granule exocytosis leading to impaired target cell lysis. Similar findings were obtained using NK cells from a stromal interaction molecule 1-deficient patient. The defect occurred at a late stage of the signaling process, because activation of leukocyte functional antigen (LFA)-1 and cytotoxic granule polarization were not impaired. Moreover, pharmacological inhibition of SOCE interfered with degranulation and target cell lysis by freshly isolated NK cells and CD8(+) effector T cells from healthy donors. In addition to effects on lymphocyte cytotoxicity, synthesis of the chemokine macrophage inflammatory protein-1ß and the cytokines TNF-α and IFN-γ on target cell recognition was impaired in ORAI1-deficient NK cells, as previously described for T cells. By contrast, NK cell cytokine production induced by combinations of IL-12, IL-15, and IL-18 was not impaired by ORAI1 deficiency. Taken together, these results identify a critical role for ORAI1-mediated Ca(2+) influx in granule exocytosis for lymphocyte cytotoxicity as well as for cytokine production induced by target cell recognition.


Subject(s)
Calcium Channels/immunology , Calcium/immunology , Cell Degranulation/immunology , Cytokines/biosynthesis , Cytotoxicity, Immunologic , T-Lymphocytes, Cytotoxic/immunology , Chemokine CCL4/biosynthesis , Humans , Interferon-gamma/biosynthesis , Interleukins/biosynthesis , Killer Cells, Natural/pathology , ORAI1 Protein , Tumor Necrosis Factor-alpha/biosynthesis
7.
Blood Adv ; 8(17): 4662-4678, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38954834

ABSTRACT

ABSTRACT: The leukemic stem cell (LSC) score LSC-17 based on a stemness-related gene expression signature is an indicator of poor disease outcome in acute myeloid leukemia (AML). However, it is not known whether "niche anchoring" of LSC affects disease evolution. To address this issue, we conditionally inactivated the adhesion molecule JAM-C (Junctional Adhesion Molecule-C) expressed by hematopoietic stem cells (HSCs) and LSCs in an inducible mixed-lineage leukemia (iMLL)-AF9-driven AML mouse model. Deletion of Jam3 (encoding JAM-C) before induction of the leukemia-initiating iMLL-AF9 fusion resulted in a shift from long-term to short-term HSC expansion, without affecting disease initiation and progression. In vitro experiments showed that JAM-C controlled leukemic cell nesting irrespective of the bone marrow stromal cells used. RNA sequencing performed on leukemic HSCs isolated from diseased mice revealed that genes upregulated in Jam3-deficient animals belonged to activation protein-1 (AP-1) and tumor necrosis factor α (TNF-α)/NF-κB pathways. Human orthologs of dysregulated genes allowed to identify a score that was distinct from, and complementary to, the LSC-17 score. Substratification of patients with AML using LSC-17 and AP-1/TNF-α genes signature defined 4 groups with median survival ranging from <1 year to a median of "not reached" after 8 years. Finally, coculture experiments showed that AP-1 activation in leukemic cells was dependent on the nature of stromal cells. Altogether, our results identify the AP-1/TNF-α gene signature as a proxy of LSC anchoring in bone marrow niches, which improves the prognostic value of the LSC-17 score. This trial was registered at www.ClinicalTrials.gov as #NCT02320656.


Subject(s)
Cell Adhesion Molecules , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute , Neoplastic Stem Cells , Animals , Humans , Mice , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/genetics , Disease Models, Animal , Gene Deletion , Hematopoietic Stem Cells/metabolism , Immunoglobulins , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Longitudinal Studies
8.
EMBO Mol Med ; 15(11): e17570, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37819151

ABSTRACT

The crosstalk between cancer and stromal cells plays a critical role in tumor progression. Syntenin is a small scaffold protein involved in the regulation of intercellular communication that is emerging as a target for cancer therapy. Here, we show that certain aggressive forms of acute myeloid leukemia (AML) reduce the expression of syntenin in bone marrow stromal cells (BMSC). Stromal syntenin deficiency, in turn, generates a pro-tumoral microenvironment. From serial transplantations in mice and co-culture experiments, we conclude that syntenin-deficient BMSC stimulate AML aggressiveness by promoting AML cell survival and protein synthesis. This pro-tumoral activity is supported by increased expression of endoglin, a classical marker of BMSC, which in trans stimulates AML translational activity. In short, our study reveals a vicious signaling loop potentially at the heart of AML-stroma crosstalk and unsuspected tumor-suppressive effects of syntenin that need to be considered during systemic targeting of syntenin in cancer therapy.


Subject(s)
Leukemia, Myeloid, Acute , Syntenins , Animals , Mice , Syntenins/genetics , Syntenins/metabolism , Down-Regulation , Leukemia, Myeloid, Acute/metabolism , Signal Transduction , Stromal Cells/metabolism , Tumor Microenvironment
9.
J Clin Immunol ; 32(3): 632-46, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22318393

ABSTRACT

Despite recent advances with monoclonal antibody therapy, chronic lymphocytic leukemia (CLL) remains incurable. Natural killer (NK) cells are potent antitumoral effectors, particularly against hematological malignancies. Defective recognition of B-CLL leukemic cells by NK cells has been previously described. Here, we deciphered the mechanisms that hamper NK cell-mediated clearance of B-CLL and evaluated the potential of NK cells as therapeutic tools for treatment of CLL. First of all, leukemic B cells resemble to normal B cells with a weak expression of ligands for NK receptors. Conversely, NK cells from B-CLL patients were functionally and phenotypically competent, despite a decrease of expression of the activating receptor NKp30. Consequently, resting allogeneic NK cells were unable to kill leukemic B cells in vitro. These data suggest that patients' NK cells cannot initiate a proper immune reaction due to a lack of leukemic cell recognition. We next set up a xenotransplantation mouse model to study NK-CLL cell interactions. Together with our in vitro studies, in vivo data revealed that activation of NK cells is required in order to control B-CLL and that activated NK cells synergize to enhance rituximab effect on tumor load. This study points out the requirements for immune system manipulation for treatment of B-CLL in combination with monoclonal antibody therapy.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Immunologic Factors/pharmacology , Killer Cells, Natural/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Adult , Aged , Animals , Histocompatibility Antigens Class I/immunology , Humans , Killer Cells, Natural/drug effects , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Natural Cytotoxicity Triggering Receptor 3/immunology , Neoplasm Transplantation , Rituximab , HLA-E Antigens
10.
Blood ; 115(11): 2167-76, 2010 Mar 18.
Article in English | MEDLINE | ID: mdl-19965656

ABSTRACT

Natural killer (NK)-cell recognition of infected or neoplastic cells can induce cytotoxicity and cytokine secretion. So far, it has been difficult to assess the relative contribution of multiple NK-cell activation receptors to cytokine and chemokine production upon target cell recognition. Using Drosophila cells expressing ligands for the NK-cell receptors LFA-1, NKG2D, DNAM-1, 2B4, and CD16, we studied the minimal requirements for secretion by freshly isolated, human NK cells. Target cell stimulation induced secretion of predominately proinflammatory cytokines and chemokines. Release of chemokines MIP-1alpha, MIP-1beta, and RANTES was induced within 1 hour of stimulation, whereas release of TNF-alpha and IFN-gamma occurred later. Engagement of CD16, 2B4, or NKG2D sufficed for chemokine release, whereas induction of TNF-alpha and IFN-gamma required engagement of additional receptors. Remarkably, our results revealed that, upon target cell recognition, CD56(dim) NK cells were more prominent cytokine and chemokine producers than CD56(bright) NK cells. The present data demonstrate how specific target cell ligands dictate qualitative and temporal aspects of NK-cell cytokine and chemokine responses. Conceptually, the results point to CD56(dim) NK cells as an important source of cytokines and chemokines upon recognition of aberrant cells, producing graded responses depending on the multiplicity of activating receptors engaged.


Subject(s)
Chemokines/biosynthesis , Killer Cells, Natural/immunology , Animals , CD56 Antigen/metabolism , Chemokines/metabolism , Drosophila melanogaster/cytology , Humans , Inflammation Mediators/metabolism , Interleukin-12/pharmacology , Interleukin-18/pharmacology , K562 Cells , Killer Cells, Natural/drug effects , Kinetics , Ligands , Models, Immunological , Receptors, IgG/metabolism , Receptors, Immunologic/metabolism , Signal Transduction/drug effects
11.
Blood ; 115(6): 1166-74, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-19903900

ABSTRACT

Expression of inhibitory killer cell immunoglobulin-like receptors (KIRs) specific for self-major histocompatibility complex (MHC) class I molecules provides an educational signal that generates functional natural killer (NK) cells. However, the effects of activating KIRs specific for self-MHC class I on NK-cell education remain elusive. Here, we provide evidence that the activating receptor KIR2DS1 tunes down the responsiveness of freshly isolated human NK cells to target cell stimulation in donors homozygous for human leukocyte antigen (HLA)-C2, the ligand of KIR2DS1. The tuning was apparent in KIR2DS1(+) NK cells lacking expression of inhibitory KIRs and CD94/NKG2A, as well as in KIR2DS1(+) NK cells coexpressing the inhibitory MHC class I-specific receptors CD94/NKG2A and KIR2DL3, but not KIR2DL1. However, the tuning of responsiveness was restricted to target cell recognition because KIR2DS1(+) NK cells responded well to stimulation with exogenous cytokines. Our results provide the first example of human NK-cell education by an activating KIR and suggest that the education of NK cells via activating KIRs is a mechanism to secure tolerance that complements education via inhibitory KIRs.


Subject(s)
HLA-C Antigens/metabolism , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily C/metabolism , NK Cell Lectin-Like Receptor Subfamily D/metabolism , Receptors, KIR2DL3/metabolism , Receptors, KIR/metabolism , Cytotoxicity, Immunologic/immunology , Flow Cytometry , HLA-C Antigens/genetics , Homozygote , Humans , Interleukin-12/metabolism , Interleukin-15/metabolism , NK Cell Lectin-Like Receptor Subfamily C/genetics , NK Cell Lectin-Like Receptor Subfamily D/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, KIR/genetics , Receptors, KIR2DL3/genetics , Reverse Transcriptase Polymerase Chain Reaction
12.
Blood ; 115(13): 2686-94, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20097883

ABSTRACT

Natural killer (NK)-cell alloreactivity in recipients of hematopoietic stem cell grafts from HLA-identical siblings is intriguing and has suggested breaking of NK-cell tolerance during the posttransplantation period. To examine this possibility, we analyzed clinical outcomes in a cohort of 105 patients with myeloid malignancies who received T cell-replete grafts from HLA-matched sibling donors. Presence of inhibitory killer cell immunoglobulin-like receptors (KIRs) for nonself HLA class I ligands had no effect on disease-free survival, incidence of relapse, or graft-versus-host disease. A longitudinal analysis of the NK-cell repertoire and function revealed a global hyporesponsiveness of NK cells early after transplantation. Functional responses recovered at approximately 6 months after transplantation. Importantly, NKG2A(-) NK cells expressing KIRs for nonself HLA class I ligands remained tolerant at all time points. Furthermore, a direct comparison of NK-cell reconstitution in T cell-replete and T cell-depleted HLA-matched sibling stem cell transplantation (SCT) revealed that NKG2A(+) NK cells dominated the functional repertoire early after transplantation, with intact tolerance of NKG2A(-) NK cells expressing KIRs for nonself ligands in both settings. Our results provide evidence against the emergence of alloreactive NK cells in HLA-identical allogeneic SCT.


Subject(s)
Bone Marrow Transplantation/immunology , HLA Antigens/immunology , Immune Tolerance , Killer Cells, Natural/immunology , Living Donors , Peripheral Blood Stem Cell Transplantation , Receptors, KIR/immunology , Acute Disease , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Graft vs Host Disease/prevention & control , HLA Antigens/genetics , Hematopoietic Stem Cell Mobilization , Humans , Immunosuppressive Agents/therapeutic use , K562 Cells/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/transplantation , Leukemia, Myeloid/immunology , Leukemia, Myeloid/surgery , Lymphocyte Depletion , Male , Middle Aged , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/surgery , Receptors, KIR/genetics , Retrospective Studies , Siblings , Transplantation, Homologous , Young Adult
13.
Blood ; 116(19): 3853-64, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-20696944

ABSTRACT

Natural killer (NK) cells are lymphocytes of the innate immune system that, following differentiation from CD56(bright) to CD56(dim) cells, have been thought to retain fixed functional and phenotypic properties throughout their lifespan. In contrast to this notion, we here show that CD56(dim) NK cells continue to differentiate. During this process, they lose expression of NKG2A, sequentially acquire inhibitory killer cell inhibitory immunoglobulin-like receptors and CD57, change their expression patterns of homing molecules, and display a gradual decline in proliferative capacity. All cellular intermediates of this process are represented in varying proportions at steady state and appear, over time, during the reconstitution of the immune system, as demonstrated in humanized mice and in patients undergoing hematopoietic stem cell transplantation. CD56(dim) NK-cell differentiation, and the associated functional imprint, occurs independently of NK-cell education by interactions with self-human leukocyte antigen class I ligands and is an essential part of the formation of human NK-cell repertoires.


Subject(s)
CD56 Antigen/metabolism , CD57 Antigens/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Receptors, KIR/metabolism , Animals , Cell Differentiation , Cell Proliferation , Hematopoietic Stem Cell Transplantation , Humans , In Vitro Techniques , Killer Cells, Natural/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Phenotype , Transplantation, Heterologous
15.
Blood ; 114(1): 95-104, 2009 Jul 02.
Article in English | MEDLINE | ID: mdl-19304956

ABSTRACT

Inhibitory killer cell immunoglobulin-like receptors (KIRs) preserve tolerance to self and shape the functional response of human natural killer (NK) cells. Here, we have evaluated the influence of selection processes in the formation of inhibitory KIR repertoires in a cohort of 44 donors homozygous for the group A KIR haplotype. Coexpression of multiple KIRs was more frequent than expected by the product rule that describes random association of independent events. In line with this observation, the probability of KIR acquisition increased with the cellular expression of KIRs. Three types of KIR repertoires were distinguished that differed in frequencies of KIR- and NKG2A-positive cells but showed no dependency on the number of self-HLA class I ligands. Furthermore, the distribution of self- and nonself-KIRs at the cell surface reflected a random combination of receptors rather than a selection process conferred by cognate HLA class I molecules. Finally, NKG2A was found to buffer overall functional responses in KIR repertoires characterized by low-KIR expression frequencies. The results provide new insights into the formation of inhibitory KIR repertoires on human NK cells and support a model in which variegated KIR repertoires are generated through sequential and random acquisition of KIRs in the absence of selection.


Subject(s)
HLA Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Receptors, KIR/genetics , Receptors, KIR/metabolism , Autoantigens/metabolism , Cohort Studies , Gene Expression , HLA Antigens/genetics , Haplotypes , Histocompatibility Antigens Class I/genetics , Homozygote , Humans , Killer Cells, Natural/immunology , Ligands , Models, Immunological , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Receptors, KIR/classification
16.
Front Immunol ; 12: 756231, 2021.
Article in English | MEDLINE | ID: mdl-34867994

ABSTRACT

In the bone marrow (BM) of adult mammals, haematopoietic stem cells (HSCs) are retained in micro-anatomical structures by adhesion molecules that regulate HSC quiescence, proliferation and commitment. During decades, researchers have used engraftment to study the function of adhesion molecules in HSC's homeostasis regulation. Since the 90's, progress in genetically engineered mouse models has allowed a better understanding of adhesion molecules involved in HSCs regulation by BM niches and raised questions about the role of adhesion mechanisms in conferring drug resistance to cancer cells nested in the BM. This has been especially studied in acute myeloid leukaemia (AML) which was the first disease in which the concept of cancer stem cell (CSC) or leukemic stem cells (LSCs) was demonstrated. In AML, it has been proposed that LSCs propagate the disease and are able to replenish the leukemic bulk after complete remission suggesting that LSC may be endowed with drug resistance properties. However, whether such properties are due to extrinsic or intrinsic molecular mechanisms, fully or partially supported by molecular crosstalk between LSCs and surrounding BM micro-environment is still matter of debate. In this review, we focus on adhesion molecules that have been involved in HSCs or LSCs anchoring to BM niches and discuss if inhibition of such mechanism may represent new therapeutic avenues to eradicate LSCs.


Subject(s)
Cell Adhesion Molecules/metabolism , Hematopoiesis/physiology , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/metabolism , Stem Cell Niche/physiology , Animals , Bone Marrow/metabolism , Cell Adhesion/physiology , Hematopoietic Stem Cells/metabolism , Humans , Tumor Microenvironment/physiology
17.
Cancers (Basel) ; 13(11)2021 May 28.
Article in English | MEDLINE | ID: mdl-34071607

ABSTRACT

Background: NK cell-based immunotherapy to prevent relapse after allogeneic transplantation is an appealing strategy because NK cells can provide strong antitumor effect without inducing graft-versus-host disease (GVHD). Thus, we designed a phase-I clinical trial evaluating the safety of a prophylactic donor-derived ex vivo IL-2 activated NK cell (IL-2 NK) infusion after allo-HSCT for patients with hematologic malignancies. Methods: Donor NK cells were purified and cultured ex vivo with IL-2 before infusion, at three dose levels. To identify the maximum tolerated dose was the main objective. In addition, we performed phenotypical and functional characterization of the NK cell therapy product, and longitudinal immune monitoring of NK cell phenotype in patients. Results: Compared to unstimulated NK cells, IL-2 NK cells expressed higher levels of activating receptors and exhibited increased degranulation and cytokine production in vitro. We treated 16 patients without observing any dose-limiting toxicity. At the last follow up, 11 out of 16 treated patients were alive in complete remission of hematologic malignancies without GVHD features and immunosuppressive treatment. Conclusions: Prophylactic donor-derived IL-2 NK cells after allo-HSCT is safe with low incidence of GVHD. Promising survivals and IL-2 NK cell activated phenotype may support a potential clinical efficacy of this strategy.

18.
Front Immunol ; 12: 730970, 2021.
Article in English | MEDLINE | ID: mdl-34975835

ABSTRACT

Natural Killer (NK) cells are potent anti-leukemic immune effectors. However, they display multiple defects in acute myeloid leukemia (AML) patients leading to reduced anti-tumor potential. Our limited understanding of the mechanisms underlying these defects hampers the development of strategies to restore NK cell potential. Here, we have used a mouse model of AML to gain insight into these mechanisms. We found that leukemia progression resulted in NK cell maturation defects and functional alterations. Next, we assessed NK cell cytokine signaling governing their behavior. We showed that NK cells from leukemic mice exhibit constitutive IL-15/mTOR signaling and type I IFN signaling. However, these cells failed to respond to IL-15 stimulation in vitro as illustrated by reduced activation of the mTOR pathway. Moreover, our data suggest that mTOR-mediated metabolic responses were reduced in NK cells from AML-bearing mice. Noteworthy, the reduction of mTOR-mediated activation of NK cells during AML development partially rescued NK cell metabolic and functional defects. Altogether, our data strongly suggest that NK cells from leukemic mice are metabolically and functionally exhausted as a result of a chronic cytokine activation, at least partially IL-15/mTOR signaling. NK cells from AML patients also displayed reduced IL-2/15Rß expression and showed cues of reduced metabolic response to IL-15 stimulation in vitro, suggesting that a similar mechanism might occur in AML patients. Our study pinpoints the dysregulation of cytokine stimulation pathways as a new mechanism leading to NK cell defects in AML.


Subject(s)
Interleukin-15/pharmacology , Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/immunology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Animals , Case-Control Studies , Cells, Cultured , Disease Models, Animal , Female , Humans , Interleukin-15/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction/genetics
19.
J Immunol ; 181(9): 6010-9, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941190

ABSTRACT

Stem cell transplantation across HLA barriers may trigger NK cell-mediated graft-vs-leukemia effects leading to improved survival for patients with hematological malignancies. However, the genetic algorithm based on killer cell Ig-like receptor (KIR) and HLA genes used to predict NK cell alloreactivity have yielded discrepant results. Accordingly, it has been difficult to define transplantation settings that favor NK cell alloreactivity. In this study, we have used multiparameter flow cytometry to simultaneously analyze the cell surface expression of all four major inhibitory KIR and CD94/NKG2A to determine the size of the alloreactive NK cell repertoires in 31 individuals homozygous for the group A KIR haplotype. We observed a vast variability in the frequencies of cells with an alloreactive potential, ranging from 0 to 62% of the total NK cell population depending on which, and how many, KIR ligands were missing in theoretical recipients. This analysis required a functional examination of KIR3DL2-single positive NK cells, showing that this subset was hyporesponsive in individuals harboring the cognate ligands HLA-A3/A11. The results provide new insights into the variability of the functional alloreactive NK cell repertoire and have implications for donor selection in hematopoietic stem cell transplantation and adoptive NK cell-based immunotherapy.


Subject(s)
Haplotypes , Histocompatibility Testing , Homozygote , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Receptors, KIR3DL2/genetics , Antibodies, Monoclonal/metabolism , Cytotoxicity Tests, Immunologic , Genotype , Graft vs Leukemia Effect/genetics , Graft vs Leukemia Effect/immunology , HLA-A Antigens/metabolism , HLA-A11 Antigen , HLA-A3 Antigen/metabolism , Histocompatibility Testing/methods , Humans , K562 Cells , Killer Cells, Natural/metabolism , Ligands , NK Cell Lectin-Like Receptor Subfamily C/biosynthesis , NK Cell Lectin-Like Receptor Subfamily C/genetics , NK Cell Lectin-Like Receptor Subfamily C/immunology , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Receptors, KIR3DL2/biosynthesis , Receptors, KIR3DL2/immunology , Receptors, KIR3DL2/metabolism , Stem Cell Transplantation
20.
Front Immunol ; 10: 1307, 2019.
Article in English | MEDLINE | ID: mdl-31244851

ABSTRACT

Women with low levels of vitamin D have a higher risk of developing breast cancer. Numerous studies associated the presence of a CD8+ T cell infiltration with a good prognosis. As vitamin D may play a key role in the modulation of the immune system, the objective of this work was to evaluate the impact of vitamin D on the breast cancer progression and mammary tumor microenvironment. We show that vitamin D decreases breast cancer tumor growth. Immunomonitoring of the different immune subsets in dissociated tumors revealed an increase in tumor infiltrating CD8+ T cells in the vitamin D-treated group. Interestingly, these CD8+ T cells exhibited a more active T cell (TEM/CM) phenotype. However, in high-fat diet conditions, we observed an opposite effect of vitamin D on breast cancer tumor growth, associated with a reduction of CD8+ T cell infiltration. Our data show that vitamin D is able to modulate breast cancer tumor growth and inflammation in the tumor microenvironment in vivo. Unexpectedly, this effect is reversed in high-fat diet conditions, revealing the importance of diet on tumor growth. We believe that supplementation with vitamin D can in certain conditions represent a new adjuvant in the treatment of breast cancers.


Subject(s)
Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Vitamin D/immunology , Animals , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Cell Proliferation/physiology , Disease Progression , Female , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Inbred C57BL , Prognosis , Tumor Microenvironment/immunology
SELECTION OF CITATIONS
SEARCH DETAIL