Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Nat Immunol ; 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956379

ABSTRACT

The functional diversity of natural killer (NK) cell repertoires stems from differentiation, homeostatic, receptor-ligand interactions and adaptive-like responses to viral infections. In the present study, we generated a single-cell transcriptional reference map of healthy human blood- and tissue-derived NK cells, with temporal resolution and fate-specific expression of gene-regulatory networks defining NK cell differentiation. Transfer learning facilitated incorporation of tumor-infiltrating NK cell transcriptomes (39 datasets, 7 solid tumors, 427 patients) into the reference map to analyze tumor microenvironment (TME)-induced perturbations. Of the six functionally distinct NK cell states identified, a dysfunctional stressed CD56bright state susceptible to TME-induced immunosuppression and a cytotoxic TME-resistant effector CD56dim state were commonly enriched across tumor types, the ratio of which was predictive of patient outcome in malignant melanoma and osteosarcoma. This resource may inform the design of new NK cell therapies and can be extended through transfer learning to interrogate new datasets from experimental perturbations or disease conditions.

2.
Proc Natl Acad Sci U S A ; 120(12): e2220320120, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36917669

ABSTRACT

Pre-existing SARS-CoV-2-reactive T cells have been identified in SARS-CoV-2-unexposed individuals, potentially modulating COVID-19 and vaccination outcomes. Here, we provide evidence that functional cross-reactive memory CD4+ T cell immunity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is established in early childhood, mirroring early seroconversion with seasonal human coronavirus OC43. Humoral and cellular immune responses against OC43 and SARS-CoV-2 were assessed in SARS-CoV-2-unexposed children (paired samples at age two and six) and adults (age 26 to 83). Pre-existing SARS-CoV-2-reactive CD4+ T cell responses targeting spike, nucleocapsid, and membrane were closely linked to the frequency of OC43-specific memory CD4+ T cells in childhood. The functional quality of the cross-reactive memory CD4+ T cell responses targeting SARS-CoV-2 spike, but not nucleocapsid, paralleled OC43-specific T cell responses. OC43-specific antibodies were prevalent already at age two. However, they did not increase further with age, contrasting with the antibody magnitudes against HKU1 (ß-coronavirus), 229E and NL63 (α-coronaviruses), rhinovirus, Epstein-Barr virus (EBV), and influenza virus, which increased after age two. The quality of the memory CD4+ T cell responses peaked at age six and subsequently declined with age, with diminished expression of interferon (IFN)-γ, interleukin (IL)-2, tumor necrosis factor (TNF), and CD38 in late adulthood. Age-dependent qualitative differences in the pre-existing SARS-CoV-2-reactive T cell responses may reflect the ability of the host to control coronavirus infections and respond to vaccination.


Subject(s)
COVID-19 , Epstein-Barr Virus Infections , Child, Preschool , Adult , Child , Humans , Middle Aged , Aged , Aged, 80 and over , SARS-CoV-2 , T-Lymphocytes , Herpesvirus 4, Human , CD4-Positive T-Lymphocytes , Spike Glycoprotein, Coronavirus , Antibodies, Viral , Cross Reactions
3.
Blood ; 139(5): 704-716, 2022 02 03.
Article in English | MEDLINE | ID: mdl-34699594

ABSTRACT

Neutrophils have been thought to play a critical role in terminal differentiation of NK cells. Whether this effect is direct or a consequence of global immune changes with effects on NK-cell homeostasis remains unknown. In this study, we used high-resolution flow and mass cytometry to examine NK-cell repertoires in 64 patients with neutropenia and 27 healthy age- and sex-matched donors. A subgroup of patients with chronic neutropenia showed severely disrupted NK-cell homeostasis manifesting as increased frequencies of CD56bright NK cells and a lack of mature CD56dim NK cells. These immature NK-cell repertoires were characterized by expression of the proliferation/exhaustion markers Ki-67, Tim-3, and TIGIT and displayed blunted tumor target cell responses. Systems-level immune mapping revealed that the changes in immunophenotypes were confined to NK cells, leaving T-cell differentiation intact. RNA sequencing of NK cells from these patients showed upregulation of a network of genes, including TNFSF9, CENPF, MKI67, and TOP2A, associated with apoptosis and the cell cycle, but different from the conventional CD56bright signatures. Profiling of 249 plasma proteins showed a coordinated enrichment of pathways related to apoptosis and cell turnover, which correlated with immature NK-cell repertoires. Notably, most of these patients exhibited severe-grade neutropenia, suggesting that the profoundly altered NK-cell homeostasis was connected to the severity of their underlying etiology. Hence, although our data suggest that neutrophils are dispensable for NK-cell development and differentiation, some patients displayed a specific gap in the NK repertoire, associated with poor cytotoxic function and more severe disease manifestations.


Subject(s)
Killer Cells, Natural/pathology , Neutropenia/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Hepatitis A Virus Cellular Receptor 2/analysis , Homeostasis , Humans , Infant , Ki-67 Antigen/analysis , Male , Middle Aged , Receptors, Immunologic/analysis , Severity of Illness Index , Young Adult
4.
Semin Immunol ; 31: 20-29, 2017 06.
Article in English | MEDLINE | ID: mdl-28888619

ABSTRACT

The contribution of natural killer (NK) cells to immunosurveillance of human cancer remains debatable. Here, we discuss advances in several areas of human NK cell research, many of which support the ability of NK cells to prevent cancer development and avoid relapse following adoptive immunotherapy. We describe the molecular basis for NK cell recognition of human tumor cells and provide evidence for NK cell-mediated killing of human primary tumor cells ex vivo. Subsequently, we highlight studies demonstrating the ability of NK cells to migrate to, and reside in, the human tumor microenvironment where selection of tumor escape variants from NK cells can occur. Indirect evidence for NK cell immunosurveillance against human malignancies is provided by the reduced incidence of cancer in individuals with high levels of NK cell cytotoxicity, and the significant clinical responses observed following infusion of human NK cells into cancer patients. Finally, we describe studies showing enhanced tumor progression, or increased cancer incidence, in patients with inherited and acquired defects in cellular cytotoxicity. All these observations have in common that they, either indirectly or directly, suggest a role for NK cells in mediating immunosurveillance against human cancer. This opens up for exciting possibilities with respect to further exploring NK cells in settings of adoptive immunotherapy in human cancer.


Subject(s)
Cytotoxicity, Immunologic , Immunologic Surveillance , Immunotherapy, Adoptive/methods , Killer Cells, Natural/immunology , Neoplasms/immunology , Animals , Carcinogenesis/immunology , Humans , Tumor Escape , Tumor Microenvironment
5.
Immunogenetics ; 69(8-9): 547-556, 2017 08.
Article in English | MEDLINE | ID: mdl-28699110

ABSTRACT

The ability of NK cells to specifically recognize cells lacking expression of self-MHC class I molecules was discovered over 30 years ago. It provided the foundation for the "missing self" hypothesis. Research in the two past decades has contributed to a detailed understanding of the molecular mechanisms that determine the specificity and strength of NK cell-mediated "missing self" responses to tumor cells. However, in light of the recent remarkable breakthroughs in clinical cancer immunotherapy, the cytolytic potential of NK cells still remains largely untapped in clinical settings. There is abundant evidence demonstrating partial or complete loss of HLA class I expression in a wide spectrum of human tumor types. Such loss may result from immune selection of escape variants by tumor-specific CD8 T cells and has more recently also been linked to acquired resistance to checkpoint inhibition therapy. In the present review, we discuss the early predictions of the "missing self" hypothesis, its molecular basis and outline the potential for NK cell-based adoptive immunotherapy to convert checkpoint inhibitor therapy-resistant patients into clinical responders.


Subject(s)
CTLA-4 Antigen/antagonists & inhibitors , Killer Cells, Natural/immunology , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Histocompatibility Antigens Class I/immunology , Humans , Neoplasms/immunology , Receptors, KIR/genetics , T-Lymphocytes, Cytotoxic/immunology
6.
J Immunol ; 195(7): 3374-81, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26320254

ABSTRACT

NK cells are functionally educated by self-MHC specific receptors, including the inhibitory killer cell Ig-like receptors (KIRs) and the lectin-like CD94/NKG2A heterodimer. Little is known about how NK cell education influences qualitative aspects of cytotoxicity such as migration behavior and efficacy of activation and killing at the single-cell level. In this study, we have compared the behavior of FACS-sorted CD56(dim)CD57(-)KIR(-)NKG2A(+) (NKG2A(+)) and CD56(dim)CD57(-)KIR(-)NKG2A(-) (lacking inhibitory receptors; IR(-)) human NK cells by quantifying migration, cytotoxicity, and contact dynamics using microchip-based live cell imaging. NKG2A(+) NK cells displayed a more dynamic migration behavior and made more contacts with target cells than IR(-) NK cells. NKG2A(+) NK cells also more frequently killed the target cells once a conjugate had been formed. NK cells with serial killing capacity were primarily found among NKG2A(+) NK cells. Conjugates involving IR(-) NK cells were generally more short-lived and IR(-) NK cells did not become activated to the same extent as NKG2A(+) NK cells when in contact with target cells, as evident by their reduced spreading response. In contrast, NKG2A(+) and IR(-) NK cells showed similar dynamics in terms of duration of conjugation periods and NK cell spreading response in conjugates that led to killing. Taken together, these observations suggest that the high killing capacity of NKG2A(+) NK cells is linked to processes regulating events in the recognition phase of NK-target cell contact rather than events after cytotoxicity has been triggered.


Subject(s)
Cell Movement/immunology , Cytotoxicity, Immunologic/immunology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily C/immunology , CD56 Antigen/metabolism , CD57 Antigens/metabolism , Cell Line , Flow Cytometry , HEK293 Cells , Humans , Image Processing, Computer-Assisted , Microchip Analytical Procedures , NK Cell Lectin-Like Receptor Subfamily C/biosynthesis , Receptors, KIR/metabolism
7.
Blood ; 121(14): 2678-88, 2013 Apr 04.
Article in English | MEDLINE | ID: mdl-23325834

ABSTRACT

Human natural killer (NK) cells are functionally regulated by killer cell immunoglobulin-like receptors (KIRs) and their interactions with HLA class I molecules. As KIR expression in a given NK cell is genetically hard-wired, we hypothesized that KIR repertoire perturbations reflect expansions of unique NK-cell subsets and may be used to trace adaptation of the NK-cell compartment to virus infections. By determining the human "KIR-ome" at a single-cell level in more than 200 donors, we were able to analyze the magnitude of NK cell adaptation to virus infections in healthy individuals. Strikingly, infection with human cytomegalovirus (CMV), but not with other common herpesviruses, induced expansion and differentiation of KIR-expressing NK cells, visible as stable imprints in the repertoire. Education by inhibitory KIRs promoted the clonal-like expansion of NK cells, causing a bias for self-specific inhibitory KIRs. Furthermore, our data revealed a unique contribution of activating KIRs (KIR2DS4, KIR2DS2, or KIR3DS1), in addition to NKG2C, in the expansion of human NK cells. These results provide new insight into the diversity of KIR repertoire and its adaptation to virus infection, suggesting a role for both activating and inhibitory KIRs in immunity to CMV infection.


Subject(s)
Cytomegalovirus Infections/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Receptors, KIR3DS1/immunology , Receptors, KIR/immunology , Cell Division/immunology , Flow Cytometry , Herpesviridae Infections/immunology , Histocompatibility Testing , Humans , Immunophenotyping , Killer Cells, Natural/cytology , Lymphocyte Activation/immunology , NK Cell Lectin-Like Receptor Subfamily C/immunology , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Receptors, KIR/metabolism , Receptors, KIR3DS1/metabolism
8.
J Immunol ; 191(11): 5669-76, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24140645

ABSTRACT

EBV, a human herpesvirus, is commonly acquired during childhood and persists latently in B cells. EBV seropositivity has been connected to immunomodulatory effects such as altered T and NK cell functional responses as well as protection against early IgE sensitization; however, owing to the asymptomatic presentation during childhood little is known regarding the infection process in children of different ages. In this study, we used mononuclear cells from cord blood and from 2- and 5-y-old EBV-naive children for in vitro EBV infection. We show that the degree of EBV-induced B cell activation and expansion differs between age groups and in particular in relationship to IFN-γ production capacity. EBV infection induced redistribution between B cell subsets with enrichment of IgD(+)CD27(+) cells (commonly referred to as non-switched memory) in infected cord blood cell cultures, and of IgD(-)CD27(+) cells (switched memory) in cell cultures from older children. We also related results to serostatus to CMV, a persistent herpesvirus that can affect differentiation status of T and NK cells. As compared with CMV(-) children, the EBV-induced enrichment of IgD(-)CD27(+) B cells was significantly reduced in infected cell cultures from CMV(+) children. This effect was associated with high levels of IFN-γ and frequencies of highly mature CD8(+)CD57(+) T cells in CMV(+) children. Our results demonstrate that both a child's age and serostatus to CMV will have an impact on EBV-induced B cell activation and expansion, and they point to the ability of viruses with immunomodulatory functions, such as CMV, to affect immune responses within the host system.


Subject(s)
B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Coinfection/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/immunology , Antibodies, Viral/blood , B-Lymphocytes/virology , CD57 Antigens/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Proliferation , Cells, Cultured , Child, Preschool , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/pathogenicity , Host-Pathogen Interactions , Humans , Immunoglobulin D/metabolism , Immunologic Memory , Interferon-gamma/metabolism , Lymphocyte Activation , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
9.
J Virol ; 87(24): 13446-55, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24089567

ABSTRACT

During childhood, infections with cytomegalovirus (CMV) and Epstein-Barr virus (EBV) can occur in close temporal proximity. Active, as well as latent, CMV infection is associated with enlarged subsets of differentiated natural killer (NK) and cytotoxic T cells. How EBV infection may influence CMV-driven immune differentiation is not known. We found that EBV coinfection selectively influenced the NK cell compartment of CMV-seropositive (CMV(+)) children. Coinfected children had significantly higher proportions of peripheral-blood NKG2C(+) NK cells than CMV(+) EBV(-) children. Ex vivo NK cell degranulation after target cell stimulation and plasma IL-15 levels were significantly higher in CMV(+) children. EBV coinfection was related to the highest levels of plasma interleukin-15 (IL-15) and IL-12p70. Remarkably, in vitro EBV infection of peripheral blood mononuclear cells (PBMC) from EBV(-) CMV(+) children increased NKG2C(+) NK cell proportions. A similar tendency was seen in cocultures of PBMC with EBV(+) lymphoblastoid B-cell lines (LCL) and IL-15. After K562 challenge, NKG2C(+) NK cells excelled in regard to degranulation and production of gamma interferon, regardless of whether there was previous coculture with LCL. Taken together, our data suggest that dual latency with these herpesviruses during childhood could contribute to an in vivo environment supporting differentiation and maintenance of distinct NK cell populations. This viral imprint may affect subsequent immune responses through altered distributions of effector cells.


Subject(s)
Cell Differentiation , Coinfection/immunology , Cytomegalovirus Infections/physiopathology , Cytomegalovirus/physiology , Epstein-Barr Virus Infections/physiopathology , Herpesvirus 4, Human/physiology , Killer Cells, Natural/cytology , Child, Preschool , Cohort Studies , Coinfection/virology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Female , Humans , Interleukin-12/immunology , Interleukin-15/immunology , Killer Cells, Natural/immunology , Leukocyte Count , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Male
10.
Blood Adv ; 8(3): 571-580, 2024 02 13.
Article in English | MEDLINE | ID: mdl-38039514

ABSTRACT

ABSTRACT: ACKR1/DARC-associated neutropenia (NP; ADAN; Online Mendelian Inheritance in Man 611862), caused by a variation in the ACKR1/DARC gene (rs2814778), is common in persons of African or Middle Eastern descent. In a cohort of 66 genetically confirmed subjects with ADAN, we show that absolute neutrophil counts (ANCs) may occasionally be lower than previously recognized (0.1 × 109-0.49 × 109/L for 9% of the subjects), which is similar to ANCs in severe congenital NP (SCNP). ANCs often normalized during inflammation, even mild. Individuals with ADAN (of 327 observed person-years) showed no cases of myelodysplastic syndrome (MDS), which is frequently encountered in SCNP. Unexpectedly, 22% presented with autoantibodies to neutrophils, compared with <1% in controls. Compared with healthy donors, subjects with ADAN demonstrated significantly lower human cationic antimicrobial protein-18/pro-leucin leucin-37 plasma levels; higher levels of nonclassical, proinflammatory, 6-sulfo LacNac-expressing monocytes; and differentially expressed plasma levels of 28 of the 239 analyzed cytokines related to immunity/inflammation, cell signaling, neutrophil activation, and angiogenesis. Collectively, more severe neutropenia in ADAN than previously assumed may complicate differential diagnoses compared with other SCNPs, and various (auto)immune/inflammatory reactions with a distinct profile may be a cause or consequence of this hereditary neutropenia.


Subject(s)
Duffy Blood-Group System , Neutropenia , Receptors, Cell Surface , Humans , Inflammation , Leukocyte Count , Neutropenia/genetics , Neutrophils , Receptors, Cell Surface/genetics , Duffy Blood-Group System/genetics
11.
Transplant Cell Ther ; 29(6): 376.e1-376.e11, 2023 06.
Article in English | MEDLINE | ID: mdl-36966873

ABSTRACT

Sirolimus is an inhibitor of the mammalian target of rapamycin (mTOR) and is emerging as a promising component of graft-versus-host disease (GVHD) prophylaxis regimens in the context of allogeneic hematopoietic stem cell transplantation (HSCT). Multiple studies have explored the clinical benefits of adding sirolimus to GVHD prophylaxis; however, detailed immunologic studies have not yet been carried out in this context. Mechanistically, mTOR is at the center of metabolic regulation in T cells and natural killer (NK) cells and is critical for their differentiation to mature effector cells. Therefore, close evaluation of the inhibition of mTOR in the context of immune reconstitution post-HSCT is warranted. In this work, we studied the effect of sirolimus on immune reconstitution using a biobank of longitudinal samples from patients receiving either tacrolimus/sirolimus (TAC/SIR) or cyclosporin A/methotrexate (CSA/MTX) as conventional GVHD prophylaxis. Healthy donor controls, donor graft material, and samples from 28 patients (14 with TAC/SIR, 14 with CSA/MTX) at 3 to 4 weeks and 34 to 39 weeks post- HSCT were collected. Multicolor flow cytometry was used to perform broad immune cell mapping, with a focus on NK cells. NK cell proliferation was evaluated over a 6-day in vitro homeostatic proliferation protocol. Furthermore, in vitro NK cell responses to cytokine stimulation or tumor cells were evaluated. Systems-level assessment of the immune repertoire revealed a deep and prolonged suppression (weeks 34 to 39 post-HSCT) of the naïve CD4 T cell compartment with relative sparing of regulatory T cells and enrichment of CD69+Ki-67+HLA-DR+ CD8 T cells, independent of the type of GVHD prophylaxis. Early after transplantation (weeks 3 to 4), while patients were still on TAC/SIR or CSA/MTX, we found a relative increase in less-differentiated CD56bright NK cells and NKG2A+CD57-KIR- CD56dim NK cells and a distinct loss of CD16 and DNAM-1 expression. Both regimens led to suppressed proliferative responses ex vivo and functional impairment with preferential loss of cytokine responsiveness and IFN-γ production. Patients who received TAC/SIR as GVHD prophylaxis showed delayed NK cell reconstitution with lower overall NK cell counts and fewer CD56bright and NKG2A+ CD56dim NK cells. Treatment with sirolimus-containing regimens generated similar immune cell profiles as conventional prophylaxis; however, the NK cell compartment seemed to be composed of slightly more mature NK cells. These effects were also present after the completion of GVHD prophylaxis, suggesting that mTOR inhibition with sirolimus leaves a lasting imprint on homeostatic proliferation and NK cell reconstitution following HSCT.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Humans , Sirolimus/pharmacology , Sirolimus/therapeutic use , Graft vs Host Disease/prevention & control , Killer Cells, Natural , Methotrexate , Cytokines/metabolism , TOR Serine-Threonine Kinases/metabolism
12.
Blood Adv ; 7(16): 4492-4504, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37327114

ABSTRACT

The functionality of natural killer (NK) cells is tuned during education and is associated with remodeling of the lysosomal compartment. We hypothesized that genetic variation in killer cell immunoglobulin-like receptor (KIR) and HLA, which is known to influence the functional strength of NK cells, fine-tunes the payload of effector molecules stored in secretory lysosomes. To address this possibility, we performed a high-resolution analysis of KIR and HLA class I genes in 365 blood donors and linked genotypes to granzyme B loading and functional phenotypes. We found that granzyme B levels varied across individuals but were stable over time in each individual and genetically determined by allelic variation in HLA class I genes. A broad mapping of surface receptors and lysosomal effector molecules revealed that DNAM-1 and granzyme B levels served as robust metric of the functional state in NK cells. Variation in granzyme B levels at rest was tightly linked to the lytic hit and downstream killing of major histocompatibility complex-deficient target cells. Together, these data provide insights into how variation in genetically hardwired receptor pairs tunes the releasable granzyme B pool in NK cells, resulting in predictable hierarchies in global NK cell function.


Subject(s)
Killer Cells, Natural , Receptors, KIR , Granzymes/genetics , Granzymes/metabolism , Receptors, KIR/genetics , Receptors, KIR/metabolism , Histocompatibility Antigens Class I/metabolism , Genotype
13.
Cell Rep ; 38(10): 110503, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35235832

ABSTRACT

Natural killer (NK) cells are innate immune cells that contribute to host defense against virus infections. NK cells respond to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in vitro and are activated in patients with acute coronavirus disease 2019 (COVID-19). However, by which mechanisms NK cells detect SARS-CoV-2-infected cells remains largely unknown. Here, we show that the Non-structural protein 13 of SARS-CoV-2 encodes for a peptide that is presented by human leukocyte antigen E (HLA-E). In contrast with self-peptides, the viral peptide prevents binding of HLA-E to the inhibitory receptor NKG2A, thereby rendering target cells susceptible to NK cell attack. In line with these observations, NKG2A-expressing NK cells are particularly activated in patients with COVID-19 and proficiently limit SARS-CoV-2 replication in infected lung epithelial cells in vitro. Thus, these data suggest that a viral peptide presented by HLA-E abrogates inhibition of NKG2A+ NK cells, resulting in missing self-recognition.


Subject(s)
COVID-19 , Histocompatibility Antigens Class I , Killer Cells, Natural , Methyltransferases , NK Cell Lectin-Like Receptor Subfamily C , RNA Helicases , SARS-CoV-2 , Viral Nonstructural Proteins , COVID-19/immunology , Histocompatibility Antigens Class I/immunology , Humans , Killer Cells, Natural/immunology , Methyltransferases/immunology , NK Cell Lectin-Like Receptor Subfamily C/immunology , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Peptides/metabolism , RNA Helicases/immunology , Viral Nonstructural Proteins/immunology , HLA-E Antigens
14.
J Immunother Cancer ; 10(11)2022 11.
Article in English | MEDLINE | ID: mdl-36319065

ABSTRACT

BACKGROUND: Natural killer (NK) cells hold great promise as a source for allogeneic cell therapy against hematological malignancies, including acute myeloid leukemia (AML). Current treatments are hampered by variability in NK cell subset responses, a limitation which could be circumvented by specific expansion of highly potent single killer immunoglobulin-like receptor (KIR)+NKG2C+ adaptive NK cells to maximize missing-self reactivity. METHODS: We developed a GMP-compliant protocol to expand adaptive NK cells from cryopreserved cells derived from select third-party superdonors, that is, donors harboring large adaptive NK cell subsets with desired KIR specificities at baseline. We studied the adaptive state of the cell product (ADAPT-NK) by flow cytometry and mass cytometry as well as cellular indexing of transcriptomes and epitopes by sequencing (CITE-Seq). We investigated the functional responses of ADAPT-NK cells against a wide range of tumor target cell lines and primary AML samples using flow cytometry and IncuCyte as well as in a mouse model of AML. RESULTS: ADAPT-NK cells were >90% pure with a homogeneous expression of a single self-HLA specific KIR and expanded a median of 470-fold. The ADAPT-NK cells largely retained their adaptive transcriptional signature with activation of effector programs without signs of exhaustion. ADAPT-NK cells showed high degranulation capacity and efficient killing of HLA-C/KIR mismatched tumor cell lines as well as primary leukemic blasts from AML patients. Finally, the expanded adaptive NK cells had preserved robust antibody-dependent cellular cytotoxicity potential and combination of ADAPT-NK cells with an anti-CD16/IL-15/anti-CD33 tri-specific engager led to near-complete killing of resistant CD45dim blast subtypes. CONCLUSIONS: These preclinical data demonstrate the feasibility of off-the-shelf therapy with a non-engineered, yet highly specific, NK cell population with full missing-self recognition capability.


Subject(s)
Cytotoxicity, Immunologic , Leukemia, Myeloid, Acute , Animals , Mice , Antibody-Dependent Cell Cytotoxicity , Killer Cells, Natural/metabolism , Leukemia, Myeloid, Acute/pathology , Receptors, KIR/metabolism
15.
Immunology ; 133(1): 41-50, 2011 May.
Article in English | MEDLINE | ID: mdl-21323661

ABSTRACT

Human monocytes can be divided into two major subpopulations, CD14(++) CD16(-) and CD14(+) CD16(+) cells, which are suggested to play different roles in antimicrobial responses. In neonates, characteristics and functional responses of monocyte subsets have not previously been explored, and might contribute to the qualitative difference between neonatal and adult cytokine profiles. We report that at baseline, monocyte subsets in cord blood and adult peripheral blood are present in similar frequencies, and show similar expression of CD11c, CD80/CD86, CD163 and HLA-DR. In response to the bacterial ligand peptidoglycan, cord blood monocytes had high inherent capacity for production of the early-response cytokines with levels of tumour necrosis factor and interleukin-12p70 exceeding adult levels, and also a higher phosphorylation of p38-mitogen-activated protein kinase. The CD14(+) CD16(+) cells expressed more interleukin-12p70 than CD14(++) CD16(-) cells and were present in a higher frequency in peptidoglycan-stimulated cord blood mononuclear cell cultures. Together, the behaviour of cord blood CD14(+) CD16(+) cells following peptidoglycan stimulation might indicate a qualitative difference between the neonatal antimicrobial response and that of the adult. In addition we found that serum factors in cord blood and adult sera affected cytokine production similarly, with the exception of tumour necrosis factor, regardless of the source of serum or cells. Overall, our data provide new insights into monocyte heterogeneity in cord blood and monocyte subset responses to a bacterial ligand at birth.


Subject(s)
Cytokines/biosynthesis , Fetal Blood/cytology , Infant, Newborn/immunology , Monocytes/cytology , Adult , Cell Separation , Cytokines/immunology , Fetal Blood/immunology , Flow Cytometry , Humans , Immunophenotyping , Lipopolysaccharide Receptors/biosynthesis , Lipopolysaccharide Receptors/immunology , Monocytes/immunology , Peptidoglycan/immunology , Receptors, IgG/biosynthesis , Receptors, IgG/immunology
16.
J Immunol ; 182(4): 2511-7, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19201907

ABSTRACT

EBV infection is inversely associated with IgE sensitization in children, and this association is further enhanced by CMV coinfection. In mice, herpesvirus latency causes systemic innate activation and protection from bacterial coinfection, implying the importance of herpesviruses in skewing immune responses during latent infection. Early control of viral infections depends on IFN-gamma release by NK cells, which generally requires the presence of accessory cells. We investigated IFN-gamma production by NK cells in PBMCs from children seropositive (SP) for EBV alone, for both EBV and CMV, or seronegative for both viruses. The ability of classical (CD14(++)CD16(-)) and proinflammatory (CD14(+)CD16(+)) monocytes to induce autologous NK cell IFN-gamma was studied by coculture experiments with enriched CD3(-)CD56(+) cells. Transwell experiments were used to evaluate how monocytes interact with NK cells to induce IFN-gamma synthesis. SP children had a significantly reduced proportion of IFN-gamma(+) NK cells and cognate intracellular IFN-gamma levels, which was more pronounced in CMV-coinfected subjects. Also, resting PBMCs of SP children displayed lower proportions of proinflammatory monocytes. IFN-gamma production by NK cells was dependent on interactions with monocytes, with the proinflammatory subset inducing the highest IFN-gamma. Finally, SP children had markedly lower levels of plasma IFN-gamma, concurrent with in vitro findings. Herpesvirus infections could be one contributing factor for maturation toward balanced Th1-Th2 responses. Our data indicate that early infection by herpesviruses may affect NK cell and monocyte interactions and thereby also influence the development of allergies.


Subject(s)
Herpesviridae Infections/immunology , Interferon-gamma/biosynthesis , Killer Cells, Natural/immunology , Monocytes/immunology , Child, Preschool , Coculture Techniques , Cytokines/immunology , Cytomegalovirus/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Herpesvirus 4, Human/immunology , Humans , Interferon-gamma/immunology , Killer Cells, Natural/virology , Lymphocyte Activation/immunology , Male , Monocytes/virology
17.
Front Immunol ; 11: 812, 2020.
Article in English | MEDLINE | ID: mdl-32477340

ABSTRACT

Natural killer (NK) cells have a central role within the innate immune system, eliminating virally infected, foreign and transformed cells through their natural cytotoxic capacity. Release of their cytotoxic granules is tightly controlled through the balance of a large repertoire of inhibitory and activating receptors, and it is the unique combination of these receptors expressed by individual cells that confers immense diversity both in phenotype and functionality. The diverse, yet unique, NK cell repertoire within an individual is surprisingly stable over time considering the constant renewal of these cells at steady state. Here we give an overview of NK cell differentiation and discuss metabolic requirements, intra-lineage plasticity and transcriptional reprogramming during IL-15-driven homeostatic proliferation. New insights into the regulation of NK cell differentiation and homeostasis could pave the way for the successful implementation of NK cell-based immunotherapy against cancer.


Subject(s)
Homeostasis/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Plasticity/immunology , Cell Proliferation , Cell- and Tissue-Based Therapy/methods , Humans , Interleukin-15/metabolism , Mice , Neoplasms/therapy , TOR Serine-Threonine Kinases/metabolism , Transcription, Genetic
18.
Cell Rep ; 29(8): 2284-2294.e4, 2019 Nov 19.
Article in English | MEDLINE | ID: mdl-31747601

ABSTRACT

Natural killer (NK) cell repertoires are made up of phenotypically distinct subsets with different functional properties. The molecular programs involved in maintaining NK cell repertoire diversity under homeostatic conditions remain elusive. Here, we show that subset-specific NK cell proliferation kinetics correlate with mTOR activation, and global repertoire diversity is maintained through a high degree of intra-lineage subset plasticity during interleukin (IL)-15-driven homeostatic proliferation in vitro. Slowly cycling sorted KIR+CD56dim NK cells with an induced CD57 phenotype display increased functional potential associated with increased transcription of genes involved in adhesion and immune synapse formation. Rapidly cycling cells upregulate NKG2A, display a general loss of functionality, and a transcriptional signature associated with increased apoptosis/cellular stress, actin-remodeling, and nuclear factor κB (NF-κB) activation. These results shed light on the role of intra-lineage plasticity during NK cell homeostasis and suggest that the functional fate of the cell is tightly linked to the acquired phenotype and transcriptional reprogramming.


Subject(s)
Killer Cells, Natural/metabolism , Apoptosis/genetics , Apoptosis/physiology , CD56 Antigen/metabolism , CD57 Antigens/metabolism , Humans , Interleukin-15/metabolism , Kinetics , NF-kappa B/metabolism , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Phenotype , Sequence Analysis, RNA , Synapses/metabolism
19.
Clin Cancer Res ; 24(8): 1834-1844, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29444931

ABSTRACT

Purpose: To evaluate the safety, efficacy, and immunobiological correlates of allogeneic NK-cell-based therapy in primary chemotherapy-refractory or relapsed high-risk myelodysplastic syndrome (MDS), secondary AML (MDS/AML), and de novo AML patients.Experimental Design: Sixteen patients received fludarabine/cyclophosphamide conditioning combined with total lymphoid irradiation followed by adoptive immunotherapy with IL2-activated haploidentical NK cells.Results: NK-cell infusions were well-tolerated, with only transient adverse events observed in the 16 patients. Six patients achieved objective responses with complete remission (CR), marrow CR, or partial remission (PR). Five patients proceeded to allogeneic hematopoietic stem cell transplantation (HSCT). Three patients are still free from disease >3 years after treatment. All evaluable patients with objective responses (5/5 evaluable) had detectable donor NK cells at days 7/14 following infusion and displayed reduction of tumor cell clones, some of which carried poor prognosis mutations. Residual lin-CD34+CD123+CD45RA+ blast cells in responders had increased total HLA class I and HLA-E expression. Responding patients displayed less pronounced activation of CD8+ T cells and lower levels of inflammatory cytokines following NK-cell infusion. Intriguingly, despite omission of systemic IL2, all patients displayed increased frequencies of activated Ki-67+CD127-FoxP3+CD25hiCD4+ Treg cells of recipient origin following NK-cell therapy.Conclusions: Overall, this study suggests that high-risk MDS is responsive to NK-cell therapy and supports the use of haploidentical NK-cell infusions as a bridge to HSCT in refractory patients. Objective clinical responses and reduction of high-risk clones were associated with detectable donor-derived NK cells, immunoediting of residual blast cells, and less pronounced host immune activation. Clin Cancer Res; 24(8); 1834-44. ©2018 AACR.


Subject(s)
Adoptive Transfer , Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/therapy , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/therapy , Transplantation, Haploidentical , Adoptive Transfer/adverse effects , Adoptive Transfer/methods , Adult , Aged , Biomarkers , Clonal Evolution/immunology , Combined Modality Therapy , Cytokines/biosynthesis , Female , Graft Survival , Humans , Immunophenotyping , Killer Cells, Natural/metabolism , Leukemia, Myeloid, Acute/diagnosis , Lymphocyte Activation/immunology , Male , Middle Aged , Myelodysplastic Syndromes/diagnosis , Remission Induction , Transplantation Chimera , Transplantation, Haploidentical/methods , Treatment Outcome
20.
Cell Rep ; 15(5): 1088-1099, 2016 05 03.
Article in English | MEDLINE | ID: mdl-27117418

ABSTRACT

Infection by human cytomegalovirus (HCMV) leads to NKG2C-driven expansion of adaptive natural killer (NK) cells, contributing to host defense. However, approximately 4% of all humans carry a homozygous deletion of the gene that encodes NKG2C (NKG2C(-/-)). Assessment of NK cell repertoires in 60 NKG2C(-/-) donors revealed a broad range of NK cell populations displaying characteristic footprints of adaptive NK cells, including a terminally differentiated phenotype, functional reprogramming, and epigenetic remodeling of the interferon (IFN)-γ promoter. We found that both NKG2C(-) and NKG2C(+) adaptive NK cells expressed high levels of CD2, which synergistically enhanced ERK and S6RP phosphorylation following CD16 ligation. Notably, CD2 co-stimulation was critical for the ability of adaptive NK cells to respond to antibody-coated target cells. These results reveal an unexpected redundancy in the human NK cell response to HCMV and suggest that CD2 provides "signal 2" in antibody-driven adaptive NK cell responses.


Subject(s)
Adaptive Immunity/immunology , CD2 Antigens/immunology , Cytomegalovirus/immunology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily C/genetics , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphocyte Activation/immunology , NK Cell Lectin-Like Receptor Subfamily C/deficiency , Receptors, IgG/genetics , Receptors, IgG/metabolism , Ribosomal Protein S6/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL