Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Immunol Rev ; 293(1): 25-37, 2020 01.
Article in English | MEDLINE | ID: mdl-31762040

ABSTRACT

Natural killer cells employ a diverse arsenal of effector mechanisms to target intracellular pathogens. Differentiation of natural killer (NK) cell activation pathways occurs along a continuum from reliance on innate pro-inflammatory cytokines and stress-induced host ligands through to interaction with signals derived from acquired immune responses. Importantly, the degree of functional differentiation of the NK cell lineage influences the magnitude and specificity of interactions with host cells infected with viruses, bacteria, fungi, and parasites. Individual humans possess a vast diversity of distinct NK cell clones, each with the capacity to vary along this functional differentiation pathway, which - when combined - results in unique individual responses to different infections. Here we summarize these NK cell differentiation events, review evidence for direct interaction of malaria-infected host cells with NK cells and assess how innate inflammatory signals induced by malaria parasite-associated molecular patterns influence the indirect activation and function of NK cells. Finally, we discuss evidence that anti-malarial immunity develops in parallel with advancing NK differentiation, coincident with a loss of reliance on inflammatory signals, and a refined capacity of NK cells to target malaria parasites more precisely, particularly through antibody-dependent mechanisms.


Subject(s)
Adaptation, Physiological/immunology , Cell Differentiation/immunology , Host-Parasite Interactions/immunology , Killer Cells, Natural/immunology , Malaria/immunology , Malaria/parasitology , Plasmodium/immunology , Adaptive Immunity , Animals , Biomarkers , Cell Differentiation/genetics , Cytokines/metabolism , Disease Models, Animal , Gene-Environment Interaction , Host-Parasite Interactions/genetics , Humans , Immunity, Innate , Inflammation Mediators/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Malaria/genetics , Malaria/metabolism , Plasmodium falciparum/immunology
2.
J Infect Dis ; 223(7): 1171-1182, 2021 04 08.
Article in English | MEDLINE | ID: mdl-31821493

ABSTRACT

BACKGROUND: Antibody Fc-mediated functions, such as antibody-dependent cellular cytotoxicity, contribute to vaccine-induced protection against viral infections. Fc-mediated function of anti-Ebola glycoprotein (GP) antibodies suggest that Fc-dependent activation of effector cells, including natural killer (NK) cells, could play a role in vaccination against Ebola virus disease. METHODS: We analyzed the effect on primary human NK cell activation of anti-Ebola GP antibody in the serum of United Kingdom-based volunteers vaccinated with the novel 2-dose heterologous adenovirus type 26.ZEBOV, modified vaccinia Ankara-BN-Filo vaccine regimen. RESULTS: We demonstrate primary human NK cell CD107a and interferon γ expression, combined with down-regulation of CD16, in response to recombinant Ebola virus GP and post-vaccine dose 1 and dose 2 serum samples. These responses varied significantly with vaccine regimen, and NK cell activation was found to correlate with anti-GP antibody concentration. We also reveal an impact of NK cell differentiation phenotype on antibody-dependent NK cell activation, with highly differentiated CD56dimCD57+ NK cells being the most responsive. CONCLUSIONS: These findings highlight the dual importance of vaccine-induced antibody concentration and NK cell differentiation status in promoting Fc-mediated activation of NK cells after vaccination, raising a potential role for antibody-mediated NK cell activation in vaccine-induced immune responses.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Ebola Vaccines , Hemorrhagic Fever, Ebola , Killer Cells, Natural/immunology , Antibodies, Viral/blood , Ebola Vaccines/immunology , Ebolavirus/immunology , Glycoproteins/immunology , Hemorrhagic Fever, Ebola/prevention & control , Humans , Vaccination , Viral Proteins/immunology
3.
J Immunol ; 203(6): 1609-1618, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31427444

ABSTRACT

Cytokine-induced memory-like (CIML) NK cells generated in response to proinflammatory cytokines in vitro and in vivo can also be generated by vaccination, exhibiting heightened responses to cytokine stimulation months after their initial induction. Our previous study demonstrated that in vitro human NK cell responses to inactivated influenza virus were also indirectly augmented by very low doses of IL-15, which increased induction of myeloid cell-derived cytokine secretion. These findings led us to hypothesize that IL-15 stimulation could reveal a similar effect for active influenza vaccination and influence CIML NK cell effector functions. In this study, 51 healthy adults were vaccinated with seasonal influenza vaccine, and PBMC were collected before and up to 30 d after vaccination. Myeloid and lymphoid cell cytokine secretion was measured after in vitro PBMC restimulation with low-dose IL-15, alone or in combination with inactivated H3N2 virus; the associated NK cell response was assessed by flow cytometry. PBMC collected 30 d postvaccination showed heightened cytokine production in response to IL-15 compared with PBMC collected at baseline; these responses were further enhanced when IL-15 was combined with H3N2. NK cell activation in response to IL-15 alone (CD25) and H3N2 plus IL-15 (CD25 and IFN-γ) was enhanced postvaccination. We also observed proliferation of less-differentiated NK cells with downregulation of cytokine receptors as early as 3 d after vaccination, suggesting cytokine stimulation in vivo. We conclude that vaccination-induced "training" of accessory cells combines with the generation of CIML NK cells to enhance the overall NK cell response postvaccination.


Subject(s)
Cytokines/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Killer Cells, Natural/immunology , Myeloid Cells/immunology , Adult , Aged , Female , Humans , Influenza A Virus, H3N2 Subtype/immunology , Interferon-gamma/immunology , Interleukin-15/immunology , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/immunology , Male , Middle Aged , Vaccination/methods , Young Adult
4.
J Immunol ; 200(8): 2738-2747, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29491009

ABSTRACT

IL-15 is a key regulator of NK cell maintenance and proliferation and synergizes with other myeloid cell-derived cytokines to enhance NK cell effector function. At low concentrations, trans-presentation of IL-15 by dendritic cells can activate NK cells, whereas at higher concentrations it can act directly on NK cells, independently of accessory cells. In this study, we investigate the potential for IL-15 to boost responses to influenza virus by promoting accessory cell function. We find that coculture of human PBMCs with inactivated whole influenza virus (A/Victoria/361/2011) in the presence of very low concentrations of IL-15 results in increased production of myeloid cell-derived cytokines, including IL-12, IFN-α2, GM-CSF, and IL-1ß, and an increased frequency of polyfunctional NK cells (defined by the expression of two or more of CD107a, IFN-γ, and CD25). Neutralization experiments demonstrate that IL-15-mediated enhancement of NK cell responses is primarily dependent on IL-12 and partially dependent on IFN-αßR1 signaling. Critically, IL-15 boosted the production of IL-12 in influenza-stimulated blood myeloid dendritic cells. IL-15 costimulation also restored the ability of less-differentiated NK cells from human CMV-seropositive individuals to respond to influenza virus. These data suggest that very low concentrations of IL-15 play an important role in boosting accessory cell function to support NK cell effector functions.


Subject(s)
Influenza A virus/immunology , Interleukin-12/biosynthesis , Interleukin-15/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Humans , Influenza, Human/immunology , Interleukin-12/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism
5.
Eur J Immunol ; 48(1): 50-65, 2018 01.
Article in English | MEDLINE | ID: mdl-28960320

ABSTRACT

Cytomegaloviruses (CMVs) are highly prevalent, persistent human pathogens that not only evade but also shape our immune responses. Natural killer (NK) cells play an important role in the control of CMV and CMVs have in turn developed a plethora of immunoevasion mechanisms targeting NK cells. This complex interplay can leave a long-lasting imprint on the immune system in general and affect responses toward other pathogens and vaccines. This review aims to provide an overview of NK cell biology and development, the manipulation of NK cells by CMVs and the potential impact of these evasion strategies on responses to vaccination.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Killer Cells, Natural/immunology , T-Lymphocytes/immunology , Viral Vaccines/immunology , Animals , Antigens, Viral/immunology , Cytomegalovirus Infections/virology , Humans , Immune Evasion/immunology , Mice , Receptors, Virus/immunology , Vaccination
6.
Eur J Immunol ; 47(6): 1040-1050, 2017 06.
Article in English | MEDLINE | ID: mdl-28383105

ABSTRACT

Human cytomegalovirus (HCMV) infection drives the phenotypic and functional differentiation of NK cells, thereby influencing the responses of these cells after vaccination. NK cell functional differentiation is particularly advanced in African populations with universal exposure to HCMV. To investigate the impact of advanced differentiation on vaccine-induced responses, we studied NK-cell function before and after vaccination with Trivalent Influenza Vaccine (TIV) or diphtheria, tetanus, pertussis, inactivated poliovirus vaccine (DTPiP) in Africans with universal, lifelong HCMV exposure. In contrast to populations with lower prevalence of HCMV infection, no significant enhancement of NK-cell responses (IFN-γ, CD107a, CD25) occurred after in vitro re-stimulation of post-vaccination NK cells with TIV or DTPiP antigens compared to pre-vaccination baseline cells. However, both vaccinations resulted in higher frequencies of NK cells producing IFN-γ in response to exogenous IL-12 with IL-18, which persisted for up to 6 months. Enhanced cytokine responsiveness was restricted to less differentiated NK cells, with increased frequencies of IFN-γ+ cells observed within CD56bright CD57- , CD56dim CD57- NKG2C- and CD56dim CD57- NKG2C+ NK-cell subsets. These data suggest a common mechanism whereby different vaccines enhance NK cell IFN-γ function in HCMV infected donors and raise the potential for further exploitation of NK cell "pre-activation" to improve vaccine effectiveness.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Diphtheria Toxoid/immunology , Influenza Vaccines/immunology , Interferon-gamma/biosynthesis , Interleukins/immunology , Killer Cells, Natural/immunology , Poliovirus Vaccines/immunology , Tetanus Toxoid/immunology , Adolescent , Adult , Africa/epidemiology , Aged , Child , Child, Preschool , Cytomegalovirus Infections/ethnology , Cytomegalovirus Infections/virology , Diphtheria Toxoid/administration & dosage , Female , Humans , Immunization, Secondary , Influenza Vaccines/administration & dosage , Influenza Vaccines/pharmacology , Interferon-gamma/immunology , Interleukin-12/immunology , Interleukin-12/pharmacology , Interleukin-18/immunology , Interleukin-18/pharmacology , Interleukin-2 Receptor alpha Subunit/immunology , Killer Cells, Natural/drug effects , Lysosomal-Associated Membrane Protein 1/immunology , Male , Middle Aged , Poliovirus Vaccines/administration & dosage , Tetanus Toxoid/administration & dosage , Vaccination , Vaccine Potency , Vaccines, Combined/administration & dosage , Vaccines, Combined/immunology , Young Adult
7.
J Immunol ; 197(1): 313-25, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27233958

ABSTRACT

Human NK cells are activated by cytokines, immune complexes, and signals transduced via activating ligands on other host cells. After vaccination, or during secondary infection, adaptive immune responses can enhance both cytokine-driven and Ab-dependent NK cell responses. However, induction of NK cells for enhanced function after in vitro exposure to innate inflammatory cytokines has also been reported and may synergize with adaptive signals to potentiate NK cell activity during infection or vaccination. To test this hypothesis, we examined the effect of seasonal influenza vaccination on NK cell function and phenotype in 52 previously unvaccinated individuals. Enhanced, IL-2-dependent, NK cell IFN-γ responses to Influenza A/California/7/2009 virus were detected up to 4 wk postvaccination and higher in human CMV (HCMV)-seronegative (HCMV(-)) individuals than in HCMV-seropositive (HCMV(+)) individuals. By comparison, robust NK cell degranulation responses were observed both before and after vaccination, due to high titers of naturally occurring anti-influenza Abs in human plasma, and did not differ between HCMV(+) and HCMV(-) subjects. In addition to these IL-2-dependent and Ab-dependent responses, NK cell responses to innate cytokines were also enhanced after influenza vaccination; this was associated with proliferation of CD57(-) NK cells and was most evident in HCMV(+) subjects. Similar enhancement of cytokine responsiveness was observed when NK cells were cocultured in vitro with Influenza A/California/7/2009 virus, and this was at least partially dependent upon IFN-αßR2. In summary, our data indicate that attenuated or live viral vaccines promote cytokine-induced memory-like NK cells and that this process is influenced by HCMV infection.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Immunologic Memory , Influenza Vaccines/immunology , Influenza, Human/immunology , Killer Cells, Natural/immunology , Orthomyxoviridae/immunology , Adult , Cells, Cultured , Cytomegalovirus Infections/complications , Humans , Influenza, Human/complications , Interferon-gamma/metabolism , Interleukin-2/metabolism , Lymphocyte Activation , Middle Aged , Vaccination , Young Adult
8.
J Immunol ; 194(10): 4657-67, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25855356

ABSTRACT

NK cells contribute to postvaccination immune responses after activation by IL-2 from Ag-specific memory T cells or by cross-linking of the low-affinity IgG receptor, CD16, by Ag-Ab immune complexes. Sensitivity of NK cells to these signals from the adaptive immune system is heterogeneous and influenced by their stage of differentiation. CD56(dim)CD57(+) NK cells are less responsive to IL-2 and produce less IFN-γ in response to T cell-mediated activation than do CD56(bright) or CD56(dim)CD57(-) NK cells. Conversely, NK cell cytotoxicity, as measured by degranulation, is maintained across the CD56(dim) subsets. Human CMV (HCMV), a highly prevalent herpes virus causing lifelong, usually latent, infections, drives the expansion of the CD56(dim)CD57(+)NKG2C(+) NK cell population, skewing the NK cell repertoire in favor of cytotoxic responses at the expense of cytokine-driven responses. We hypothesized, therefore, that HCMV seropositivity would be associated with altered NK cell responses to vaccine Ags. In a cross-sectional study of 152 U.K. adults, with HCMV seroprevalence rate of 36%, we find that HCMV seropositivity is associated with lower NK cell IFN-γ production and degranulation after in vitro restimulation with pertussis or H1N1 influenza vaccine Ags. Higher expression of CD57/NKG2C and lower expression of IL-18Rα on NK cells from HCMV seropositive subjects do not fully explain these impaired responses, which are likely the result of multiple receptor-ligand interactions. This study demonstrates for the first time, to our knowledge, that HCMV serostatus influences NK cell contributions to adaptive immunity and raises important questions regarding the impact of HCMV infection on vaccine efficacy.


Subject(s)
Cytomegalovirus Infections/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Killer Cells, Natural/immunology , Pertussis Vaccine/immunology , Adult , Aged , Cross-Sectional Studies , Cytomegalovirus/immunology , Female , Flow Cytometry , Humans , Male , Middle Aged , Seroepidemiologic Studies , Young Adult
9.
Hum Genet ; 135(8): 939-51, 2016 08.
Article in English | MEDLINE | ID: mdl-27312142

ABSTRACT

NKG2C is an activating receptor that is preferentially expressed on natural killer (NK) cells. The gene encoding NKG2C (killer cell lectin-like receptor C2, KLRC2) is present at different copy numbers in the genomes of different individuals. Deletion at the NKG2C locus was investigated in a case-control study of 1522 individuals indigenous to East- and West-Africa and the association with the ocular Chlamydia trachomatis infection and its sequelae was explored. The frequency of homozygous KLRC2 deletion was 13.7 % in Gambians and 4.7 % in Tanzanians. A significantly higher frequency of the deletion allele was found in West-Africans from the Gambia and Guinea-Bissau (36.2 % p = 2.105 × 10(-8), 26.8 % p = 0.050; respectively) in comparison to East-African Tanzanians where the frequency of the deletion is comparable to other human populations (20.9 %). We found no evidence for an association between the numbers of KLRC2 gene copies and the clinical manifestations of trachoma (follicular trachoma or conjunctival scarring). A new method for imputation of KLRC2 genotypes from single nucleotide polymorphism (SNP) data in 2621 individuals from the Gambia further confirmed these results. Our data suggest that NKG2C does not play a major role in trachomatous disease. We found that the deletion allele is present at different frequencies in different populations but the reason behind these differences is currently not understood. The new method offers the potential to use SNP arrays from genome wide association studies to study the frequency of KLRC2 deletion in other populations and its association with other diseases.


Subject(s)
Genetic Predisposition to Disease , Genome-Wide Association Study , NK Cell Lectin-Like Receptor Subfamily C/genetics , Trachoma/genetics , Adolescent , Adult , Africa, Western , Aged , Aged, 80 and over , Alleles , Child , Child, Preschool , Female , Genotype , Homozygote , Humans , Infant , Infant, Newborn , Linkage Disequilibrium , Male , Middle Aged , Polymorphism, Single Nucleotide , Sequence Deletion/genetics , Trachoma/epidemiology , Trachoma/pathology
10.
Blood ; 124(14): 2213-22, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-25150297

ABSTRACT

Natural killer (NK) cells differentiate and mature during the human life course; human cytomegalovirus (HCMV) infection is a known driver of this process. We have explored human NK cell phenotypic and functional maturation in a rural African (Gambian) population with a high prevalence of HCMV. The effect of age on the frequency, absolute number, phenotype, and functional capacity of NK cells was monitored in 191 individuals aged from 1 to 49 years. Increasing frequencies of NK cells with age were associated with increased proportions of CD56dim cells expressing the differentiation marker CD57 and expansion of the NKG2C+ subset. Frequencies of NK cells responding to exogenous cytokines declined with age in line with a decreased proportion of CD57- cells. These changes coincided with a highly significant drop in anti-HCMV IgG titers by the age of 10 years, suggesting that HCMV infection is brought under control as NK cells differentiate (or vice versa). Deletion at the NKG2C locus was associated with a gene dose-dependent reduction in proportions of CD94+ and CD57+ NK cells. Importantly, anti-HCMV IgG titers were significantly elevated in NKG2C-/- children, suggesting that lack of expression of NKG2C may be associated with altered control of HCMV in childhood.


Subject(s)
Cytomegalovirus Infections/metabolism , Cytomegalovirus/metabolism , Gene Deletion , Killer Cells, Natural/cytology , NK Cell Lectin-Like Receptor Subfamily C/genetics , Adolescent , Adult , CD57 Antigens/metabolism , Cell Differentiation , Cells, Cultured , Child , Child, Preschool , Female , Gene Expression Profiling , Genotype , Humans , Infant , Leukocytes, Mononuclear/cytology , Male , Middle Aged , NK Cell Lectin-Like Receptor Subfamily D/metabolism , Phenotype , Young Adult
11.
Immunology ; 142(1): 140-50, 2014 May.
Article in English | MEDLINE | ID: mdl-24843874

ABSTRACT

Natural killer (NK) cells contribute to the effector phase of vaccine-induced adaptive immune responses, secreting cytokines and releasing cytotoxic granules. The proportion of responding NK cells varies between individuals and by vaccine, suggesting that functionally discrete subsets of NK cells with different activation requirements may be involved. Here, we have used responses to individual components of the DTP vaccine [tetanus toxoid (TT), diphtheria toxoid (DT), whole cell inactivated pertussis] to characterize the NK cell subsets involved in interleukin-2-dependent recall responses. Culture with TT, DT or pertussis induced NK cell CD25 expression and interferon-γ production in previously vaccinated individuals. Responses were the most robust against whole cell pertussis, with responses to TT being particularly low. Functional analysis of discrete NK cell subsets revealed that transition from CD56bright to CD56dim correlated with increased responsiveness to CD16 cross-linking, whereas increasing CD57 expression correlated with a loss of responsiveness to cytokines. A higher frequency of CD56dim CD57− NK cells expressed CD25 and interferon-γ following stimulation with vaccine antigen compared with CD56dim CD57+ NK cells and made the largest overall contribution to this response. CD56dim CD57int NK cells represent an intermediate functional phenotype in response to vaccine-induced and receptor-mediated stimuli. These findings have implications for the ability of NK cells to contribute to the effector response after vaccination and for vaccine-induced immunity in older individuals.


Subject(s)
Adaptive Immunity , Antigens, Bacterial/immunology , CD57 Antigens/metabolism , Diphtheria-Tetanus-Pertussis Vaccine/immunology , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , Adult , Aged , Biomarkers/metabolism , CD56 Antigen/metabolism , Cells, Cultured , Diphtheria Toxoid/immunology , GPI-Linked Proteins/metabolism , Humans , Immunologic Memory , Interferon-gamma/metabolism , Interleukin-18 Receptor alpha Subunit/metabolism , Killer Cells, Natural/classification , Killer Cells, Natural/metabolism , Lymphocyte Subsets/classification , Lymphocyte Subsets/metabolism , Lysosomal-Associated Membrane Protein 1/metabolism , Middle Aged , Pertussis Vaccine/immunology , Phenotype , Receptors, IgG/metabolism , Receptors, Interleukin-12/metabolism , Tetanus Toxoid/immunology , Young Adult
12.
Eur J Immunol ; 41(7): 1958-68, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21509779

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) establishes life-long infection by evading clearance by the host immune system. In de novo infection and lytic replication, KSHV escapes cytotoxic T cells and NK cells through downregulation of MHC class-I and ICAM-1 molecules and associated antigens involved in forming and sustaining the immunological synapse. However, the efficacy of such mechanisms in the context of the predominantly latent KSHV infection reported in Kaposi's sarcoma (KS) lesions is unclear. Using primary dermal fibroblasts in a novel in vitro model of chronic latent KSHV infection, we generated target cells with viral loads similar to those in spindle cells extracted from KS lesions. We show that latently KSHV-infected fibroblasts had normal levels of MHC-class I, ICAM-1, HLA-E and NKG2D ligand expression, were resistant to NK-cell natural cytotoxicity and were highly susceptible to killing by cytokine-activated immunocompetent NK cells. KSHV-infected fibroblasts expressed normal levels of IFN-γR1 and responded to exogenous IFN-γ by upregulating MHC class I, ICAM-1 and HLA-E and resisting activated NK-cell killing. These data demonstrate that physiologically relevant levels of latent KSHV infection in primary cells cause limited activation of resting NK cells and confer little specific resistance to control by activated NK cells.


Subject(s)
Fibroblasts/immunology , Fibroblasts/virology , Herpesvirus 8, Human/physiology , Killer Cells, Natural/immunology , Lymphocyte Activation , Sarcoma, Kaposi/virology , Cells, Cultured , Cytokines/metabolism , Cytotoxicity, Immunologic , Flow Cytometry , Gene Expression , HLA Antigens/metabolism , Herpesvirus 8, Human/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Interferon-gamma/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Phenotype , Polymerase Chain Reaction , Sarcoma, Kaposi/immunology , Virus Latency , HLA-E Antigens
13.
Vaccines (Basel) ; 10(6)2022 May 31.
Article in English | MEDLINE | ID: mdl-35746491

ABSTRACT

Natural killer cells play an important role in the control of viral infections both by regulating acquired immune responses and as potent innate or antibody-mediated cytotoxic effector cells. NK cells have been implicated in control of Ebola virus infections and our previous studies in European trial participants have demonstrated durable activation, proliferation and antibody-dependent NK cell activation after heterologous two-dose Ebola vaccination with adenovirus type 26.ZEBOV followed by modified vaccinia Ankara-BN-Filo. Regional variation in immunity and environmental exposure to pathogens, in particular human cytomegalovirus, have profound impacts on NK cell functional capacity. We therefore assessed the NK cell phenotype and function in African trial participants with universal exposure to HCMV. We demonstrate a significant redistribution of NK cell subsets after vaccine dose two, involving the enrichment of less differentiated CD56dimCD57- and CD56dimFcεR1γ+ (canonical) cells and the increased proliferation of these subsets. Sera taken after vaccine dose two support robust antibody-dependent NK cell activation in a standard NK cell readout; these responses correlate strongly with the concentration of anti-Ebola glycoprotein specific antibodies. These sera also promote comparable IFN-γ production in autologous NK cells taken at baseline and post-vaccine dose two. However, degranulation responses of post-vaccination NK cells were reduced compared to baseline NK cells and these effects could not be directly attributed to alterations in NK cell phenotype after vaccination. These studies demonstrate consistent changes in NK cell phenotypic composition and robust antibody-dependent NK cell function and reveal novel characteristics of these responses after heterologous two dose Ebola vaccination in African individuals.

14.
Clin Transl Immunology ; 10(1): e1244, 2021.
Article in English | MEDLINE | ID: mdl-33505682

ABSTRACT

Natural killer cells constitute a phenotypically diverse population of innate lymphoid cells with a broad functional spectrum. Classically defined as cytotoxic lymphocytes with the capacity to eliminate cells lacking self-MHC or expressing markers of stress or neoplastic transformation, critical roles for NK cells in immunity to infection in the regulation of immune responses and as vaccine-induced effector cells have also emerged. A crucial feature of NK cell biology is their capacity to integrate signals from pathogen-, tumor- or stress-induced innate pathways and from antigen-specific immune responses. The extent to which innate and acquired immune mediators influence NK cell effector function is influenced by the maturation and differentiation state of the NK cell compartment; moreover, NK cell differentiation is driven in part by exposure to infection. Pathogens can thus mould the NK cell response to maximise their own success and/or minimise the damage they cause. Here, we review recent evidence that pathogen- and vaccine-derived signals influence the differentiation, adaptation and subsequent effector function of human NK cells.

15.
NPJ Vaccines ; 6(1): 19, 2021 Jan 29.
Article in English | MEDLINE | ID: mdl-33514756

ABSTRACT

Natural killer (NK) cells are implicated among immune effectors after vaccination against viral pathogens, including Ebola virus. The two-dose heterologous Ebola virus vaccine regimen, adenovirus type 26.ZEBOV followed by modified vaccinia Ankara-BN-Filo (EBOVAC2 consortium, EU Innovative Medicines Initiative), induces NK cell activation and anti-Ebola glycoprotein (GP) antibody-dependent NK cell activation post-dose 1, which is further elevated post-dose 2. Here, in a multicentre, phase 2 clinical trial (EBL2001), we demonstrate durable ex vivo NK cell activation 180 days after dose 2, with responses enriched in CD56bright NK cells. In vitro antibody-dependent responses to immobilised Ebola GP increased after dose 1, and remained elevated compared to pre-vaccination levels in serum collected 180 days later. Peak NK cell responses were observed post-dose 2 and NK cell IFN-γ responses remained significantly elevated at 180 days post-dose 2. Individual variation in NK cell responses were influenced by both anti-Ebola GP antibody concentrations and intrinsic interindividual differences in NK cell functional capacity. In summary, this study demonstrates durable NK cell responses after Ad26.ZEBOV, MVA-BN-Filo Ebola virus vaccination and could inform the immunological evaluation of future iterations of the vaccine regimen and vaccination schedules.

16.
Front Immunol ; 11: 533, 2020.
Article in English | MEDLINE | ID: mdl-32296438

ABSTRACT

Human adaptive natural killer (NK) cells have diminished reliance on accessory cytokines for their activation whilst being efficiently activated by infected host cells in conjunction with pathogen specific antibodies. Here, we show that potent antibody-dependent NK cell responses are induced by Plasmodium falciparum infected erythrocytes (iRBC) in peripheral blood mononuclear cells (PBMC) from malaria-exposed Gambian individuals in the presence of autologous sera, which are absent in those from malaria-naïve UK individuals. However, malaria hyper-immune serum promotes rapid NK cell responses to iRBC in cells from both Gambian and UK individuals. Among Gambians, highly differentiated, adaptive (CD56dimFcεR1γ-CD57+) NK cells dominate both antibody-dependent NK cell IFN-γ responses and degranulation responses, whereas among UK individuals these responses are predominantly found within canonical, highly differentiated CD56dimFcεR1γ+CD57+ NK cells. Indeed, overall frequencies of adaptive, FcεR1γ-CD57+ NK cells are significantly higher among Gambian donors compared to HCMV-infected and HCMV-uninfected UK adults. Among UK individuals, antibody-dependent NK cell IFN-γ responses to iRBC were dependent on IL-18 whereas among Gambians, the predominant adaptive FcεR1γ- NK cell response was IL-18 (and accessory cell) independent (although the lower frequency response of canonical FcεR1γ NK cells did rely on this cytokine).


Subject(s)
Interleukin-18/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Adaptive Immunity/immunology , Adult , Antibody-Dependent Cell Cytotoxicity/immunology , Female , Humans , Immunity, Innate/immunology , Male
17.
J Clin Invest ; 130(7): 3936-3946, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32315287

ABSTRACT

BACKGROUNDNK cells are activated by innate cytokines and viral ligands to kill virus-infected cells. These functions are enhanced during secondary immune responses and after vaccination by synergy with effector T cells and virus-specific antibodies. In human Ebola virus infection, clinical outcome is strongly associated with the initial innate cytokine response, but the role of NK cells has not been thoroughly examined.METHODSThe novel 2-dose heterologous Adenovirus type 26.ZEBOV (Ad26.ZEBOV) and modified vaccinia Ankara-BN-Filo (MVA-BN-Filo) vaccine regimen is safe and provides specific immunity against Ebola glycoprotein, and is currently in phase 2 and 3 studies. Here, we analyzed NK cell phenotype and function in response to Ad26.ZEBOV, MVA-BN-Filo vaccination regimen and in response to in vitro Ebola glycoprotein stimulation of PBMCs isolated before and after vaccination.RESULTSWe show enhanced NK cell proliferation and activation after vaccination compared with baseline. Ebola glycoprotein-induced activation of NK cells was dependent on accessory cells and TLR-4-dependent innate cytokine secretion (predominantly from CD14+ monocytes) and enriched within less differentiated NK cell subsets. Optimal NK cell responses were dependent on IL-18 and IL-12, whereas IFN-γ secretion was restricted by high concentrations of IL-10.CONCLUSIONThis study demonstrates the induction of NK cell effector functions early after Ad26.ZEBOV, MVA-BN-Filo vaccination and provides a mechanism for the activation and regulation of NK cells by Ebola glycoprotein.TRIAL REGISTRATIONClinicalTrials.gov NCT02313077.FUNDINGUnited Kingdom Medical Research Council Studentship in Vaccine Research, Innovative Medicines Initiative 2 Joint Undertaking, EBOVAC (grant 115861) and Crucell Holland (now Janssen Vaccines and Prevention B.V.), European Union's Horizon 2020 research and innovation programme and European Federation of Pharmaceutical Industries and Associations (EFPIA).


Subject(s)
Ebola Vaccines/immunology , Ebolavirus/immunology , Interleukin-18/immunology , Killer Cells, Natural/immunology , Viral Envelope Proteins/immunology , Adolescent , Adult , Antibodies, Viral/immunology , Ebola Vaccines/administration & dosage , Ebola Vaccines/genetics , Ebolavirus/genetics , Female , Humans , Male , Middle Aged , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/genetics
18.
Front Immunol ; 11: 594107, 2020.
Article in English | MEDLINE | ID: mdl-33343571

ABSTRACT

Innate lymphoid cell (ILC) lineages mirror those of CD4+ T helper cell subsets, producing type 1, 2 and 3 cytokines respectively. Studies in adult human populations have shown contributions of non-cytotoxic ILC to immune regulation or pathogenesis in a wide range of diseases and have prompted investigations of potential functional redundancy between ILC and T helper cell compartments in neonates and children. To investigate the potential for ILC to contribute to immune responses across the human lifespan, we examined the numbers and frequencies of peripheral blood ILC subsets in a cohort of Gambians aged between 5 and 73 years of age. ILC2 were the most abundant peripheral blood ILC subset in this Gambian cohort, while ILC1 were the rarest at all ages. Moreover, the frequency of ILC1s (as a proportion of all lymphocytes) was remarkably stable over the life course whereas ILC3 cell frequencies and absolute numbers declined steadily across the life course and ILC2 frequencies and absolute numbers declined from childhood until the age of approx. 30 years of age. Age-related reductions in ILC2 cell numbers appeared to be partially offset by increasing numbers of total and GATA3+ central memory (CD45RA-CCR7+) CD4+ T cells, although there was also a gradual decline in numbers of total and GATA3+ effector memory (CD45RA-CCR7-) CD4+ T cells. Despite reduced overall abundance of ILC2 cells, we observed a coincident increase in the proportion of CD117+ ILC2, indicating potential for age-related adaptation of these cells in childhood and early adulthood. While both CD117+ and CD117- ILC2 cells produced IL-13, these responses occurred predominantly within CD117- cells. Furthermore, comparison of ILC frequencies between aged-matched Gambian and UK young adults (25-29 years) revealed an overall higher proportion of ILC1 and ILC2, but not ILC3 in Gambians. Thus, these data indicate ongoing age-related changes in ILC2 cells throughout life, which retain the capacity to differentiate into potent type 2 cytokine producing cells, consistent with an ongoing role in immune modulation.


Subject(s)
Immunity, Innate , Lymphocyte Count , Lymphocytes/immunology , Adolescent , Adult , Age Factors , Aged , Aging/blood , Aging/immunology , Biomarkers , Black People , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Child , Child, Preschool , Female , Humans , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunologic Memory , Immunophenotyping , Interleukin-13/metabolism , Lymphocytes/metabolism , Male , Middle Aged , Proto-Oncogene Proteins c-kit/metabolism , Young Adult
19.
Front Immunol ; 11: 135, 2020.
Article in English | MEDLINE | ID: mdl-32117282

ABSTRACT

Despite evidence of augmented Natural Killer (NK) cell responses after influenza vaccination, the role of these cells in vaccine-induced immunity remains unclear. Here, we hypothesized that NK cells might increase viral clearance but possibly at the expense of increased severity of pathology. On the contrary, we found that NK cells serve a homeostatic role during influenza virus infection of vaccinated mice, allowing viral clearance with minimal pathology. Using a diphtheria toxin receptor transgenic mouse model, we were able to specifically deplete NKp46+ NK cells through the administration of diphtheria toxin. Using this model, we assessed the effect of NK cell depletion prior to influenza challenge in vaccinated and unvaccinated mice. NK-depleted, vaccinated animals lost significantly more weight after viral challenge than vaccinated NK intact animals, indicating that NK cells ameliorate disease in vaccinated animals. However, there was also a significant reduction in viral load in NK-depleted, unvaccinated animals indicating that NK cells also constrain viral clearance. Depletion of NK cells after vaccination, but 21 days before infection, did not affect viral clearance or weight loss-indicating that it is the presence of NK cells during the infection itself that promotes homeostasis. Further work is needed to identify the mechanism(s) by which NK cells regulate adaptive immunity in influenza-vaccinated animals to allow efficient and effective virus control whilst simultaneously minimizing inflammation and pathology.


Subject(s)
Influenza Vaccines/immunology , Killer Cells, Natural/immunology , Orthomyxoviridae Infections/immunology , Animals , Mice , Mice, Inbred C57BL
20.
Clin Transl Immunology ; 7(1): e1010, 2018.
Article in English | MEDLINE | ID: mdl-29484187

ABSTRACT

Vaccination has proved to be highly effective in reducing global mortality and eliminating infectious diseases. Building on this success will depend on the development of new and improved vaccines, new methods to determine efficacy and optimum dosing and new or refined adjuvant systems. NK cells are innate lymphoid cells that respond rapidly during primary infection but also have adaptive characteristics enabling them to integrate innate and acquired immune responses. NK cells are activated after vaccination against pathogens including influenza, yellow fever and tuberculosis, and their subsequent maturation, proliferation and effector function is dependent on myeloid accessory cell-derived cytokines such as IL-12, IL-18 and type I interferons. Activation of antigen-presenting cells by live attenuated or whole inactivated vaccines, or by the use of adjuvants, leads to enhanced and sustained NK cell activity, which in turn contributes to T cell recruitment and memory cell formation. This review explores the role of cytokine-activated NK cells as vaccine-induced effector cells and in recall responses and their potential contribution to vaccine and adjuvant development.

SELECTION OF CITATIONS
SEARCH DETAIL