Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 May 14.
Article in English | MEDLINE | ID: mdl-38798324

ABSTRACT

Plasmodium falciparum infection can trigger high levels of inflammation that lead to fever and sometimes severe disease. People living in malaria-endemic areas gradually develop resistance to symptomatic malaria and control both parasite numbers and the inflammatory response. We previously found that adaptive natural killer (NK) cells correlate with reduced parasite load and protection from symptoms. We also previously found that murine NK cell production of IL-10 can protect mice from experimental cerebral malaria. Human NK cells can also secrete IL-10, but it was unknown what NK cell subsets produce IL-10 and if this is affected by malaria experience. We hypothesize that NK cell immunoregulation may lower inflammation and reduce fever induction. Here, we show that NK cells from subjects with malaria experience make significantly more IL-10 than subjects with no malaria experience. We then determined the proportions of NK cells that are cytotoxic and produce interferon gamma and/or IL-10 and identified a signature of adaptive and checkpoint molecules on IL-10-producing NK cells. Lastly, we find that co-culture with primary monocytes, Plasmodium -infected RBCs, and antibody induces IL-10 production by NK cells. These data suggest that NK cells may contribute to protection from malaria symptoms via IL-10 production.

2.
bioRxiv ; 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38659969

ABSTRACT

Multisystem Inflammatory Syndrome in Children (MIS-C) is a severe complication of SARS-CoV-2 infection characterized by multi-organ involvement and inflammation. Testing of cellular function ex vivo to understand the aberrant immune response in MIS-C is limited. Despite strong antibody production in MIS-C, SARS-CoV-2 nucleic acid testing can remain positive for 4-6 weeks after infection. Therefore, we hypothesized that dysfunctional cell-mediated antibody responses downstream of antibody production may be responsible for delayed clearance of viral products in MIS-C. In MIS-C, monocytes were hyperfunctional for phagocytosis and cytokine production, while natural killer (NK) cells were hypofunctional for both killing and cytokine production. The decreased NK cell cytotoxicity correlated with an NK exhaustion marker signature and systemic IL-6 levels. Potentially providing a therapeutic option, cellular engagers of CD16 and SARS-CoV-2 proteins were found to rescue NK cell function in vitro. Together, our results reveal dysregulation in antibody-mediated cellular responses unique to MIS-C that likely contribute to the immune pathology of this disease.

3.
J Immunother Cancer ; 11(12)2023 12 06.
Article in English | MEDLINE | ID: mdl-38056893

ABSTRACT

BACKGROUND: Antibody therapies can direct natural killer (NK) cells to tumor cells, tumor-associated cells, and suppressive immune cells to mediate antibody-dependent cell-mediated cytotoxicity (ADCC). This antigen-specific effector function of human NK cells is mediated by the IgG Fc receptor CD16A (FcγRIIIA). Preclinical and clinical studies indicate that increasing the binding affinity and avidity of CD16A for antibodies improves the therapeutic potential of ADCC. CD64 (FcγRI), expressed by myeloid cells but not NK cells, is the only high affinity IgG Fc receptor and is uniquely capable of stably binding to free monomeric IgG as a physiological function. We have reported on the generation of the FcγR fusion CD64/16A, consisting of the extracellular region of CD64 and the transmembrane and cytoplasmic regions from CD16A, retaining its signaling and cellular activity. Here, we generated induced pluripotent stem cell (iPSC)-derived NK (iNK) cells expressing CD64/16A as a potential adoptive NK cell therapy for increased ADCC potency. METHODS: iPSCs were engineered to express CD64/16A as well as an interleukin (IL)-15/IL-15Rα fusion (IL-15RF) protein and differentiated into iNK cells. iNK cells and peripheral blood NK cells were expanded using irradiated K562-mbIL21-41BBL feeder cells and examined. NK cells, ovarian tumor cell lines, and therapeutic monoclonal antibodies were used to assess ADCC in vitro, performed by a DELFIA EuTDA assay or in real-time by IncuCyte assays, and in vivo. For the latter, we developed a xenograft mouse model with high circulating levels of human IgG for more physiological relevance. RESULTS: We demonstrate that (1) iNK-CD64/16A cells after expansion or thaw from cryopreservation can be coupled to therapeutic antibodies, creating armed iNK cells; (2) antibody-armed iNK-CD64/16A cells can be redirected by added antibodies to target new tumor antigens, highlighting additional potential of these cells; (3) cytokine-autonomous activity by iNK-CD64/16A cells engineered to express IL-15RF; and that (4) antibody-armed iNK-CD64/16A cells thawed from cryopreservation are capable of sustained and robust ADCC in vitro and in vivo, as determined by using a modified tumor xenograft model with high levels of competing human IgG. CONCLUSIONS: iNK cells expressing CD64/16A provide an off-the-shelf multiantigen targeting platform to address tumor heterogeneity and mitigate antigen escape.


Subject(s)
Induced Pluripotent Stem Cells , Receptors, IgG , Humans , Animals , Mice , Receptors, IgG/metabolism , Induced Pluripotent Stem Cells/metabolism , Killer Cells, Natural , Cell Line, Tumor , Immunoglobulin G
4.
Front Immunol ; 14: 1267774, 2023.
Article in English | MEDLINE | ID: mdl-37928543

ABSTRACT

In the setting of viral challenge, natural killer (NK) cells play an important role as an early immune responder against infection. During this response, significant changes in the NK cell population occur, particularly in terms of their frequency, location, and subtype prevalence. In this review, changes in the NK cell repertoire associated with several pathogenic viral infections are summarized, with a particular focus placed on changes that contribute to NK cell dysregulation in these settings. This dysregulation, in turn, can contribute to host pathology either by causing NK cells to be hyperresponsive or hyporesponsive. Hyperresponsive NK cells mediate significant host cell death and contribute to generating a hyperinflammatory environment. Hyporesponsive NK cell populations shift toward exhaustion and often fail to limit viral pathogenesis, possibly enabling viral persistence. Several emerging therapeutic approaches aimed at addressing NK cell dysregulation have arisen in the last three decades in the setting of cancer and may prove to hold promise in treating viral diseases. However, the application of such therapeutics to treat viral infections remains critically underexplored. This review briefly explores several therapeutic approaches, including the administration of TGF-ß inhibitors, immune checkpoint inhibitors, adoptive NK cell therapies, CAR NK cells, and NK cell engagers among other therapeutics.


Subject(s)
Killer Cells, Natural , Virus Diseases , Humans , Virus Diseases/therapy , Immunotherapy, Adoptive
5.
J Immunol ; 210(8): 1108-1122, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36881874

ABSTRACT

CMV infection alters NK cell phenotype and function toward a more memory-like immune state. These cells, termed adaptive NK cells, typically express CD57 and NKG2C but lack expression of the FcRγ-chain (gene: FCER1G, FcRγ), PLZF, and SYK. Functionally, adaptive NK cells display enhanced Ab-dependent cellular cytotoxicity (ADCC) and cytokine production. However, the mechanism behind this enhanced function is unknown. To understand what drives enhanced ADCC and cytokine production in adaptive NK cells, we optimized a CRISPR/Cas9 system to ablate genes from primary human NK cells. We ablated genes that encode molecules in the ADCC pathway, such as FcRγ, CD3ζ, SYK, SHP-1, ZAP70, and the transcription factor PLZF, and tested subsequent ADCC and cytokine production. We found that ablating the FcRγ-chain caused a modest increase in TNF-α production. Ablation of PLZF did not enhance ADCC or cytokine production. Importantly, SYK kinase ablation significantly enhanced cytotoxicity, cytokine production, and target cell conjugation, whereas ZAP70 kinase ablation diminished function. Ablating the phosphatase SHP-1 enhanced cytotoxicity but reduced cytokine production. These results indicate that the enhanced cytotoxicity and cytokine production of CMV-induced adaptive NK cells is more likely due to the loss of SYK than the lack of FcRγ or PLZF. We found the lack of SYK expression could improve target cell conjugation through enhanced CD2 expression or limit SHP-1-mediated inhibition of CD16A signaling, leading to enhanced cytotoxicity and cytokine production.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Humans , Syk Kinase/genetics , CRISPR-Cas Systems , Killer Cells, Natural , Cytokines , Antibody-Dependent Cell Cytotoxicity
6.
Methods Mol Biol ; 2470: 641-657, 2022.
Article in English | MEDLINE | ID: mdl-35881380

ABSTRACT

Antibody-dependent cellular cytotoxicity (ADCC) is a mechanism of cell defense that bridges the innate and adaptive immune systems. ADCC has been found to be a major route of immune protection against both viral infections and cancer. Recently, natural killer (NK) cell-mediated ADCC has been shown as a mechanism of parasite clearance and protection against malaria (Hart et al. J Exp Med 216(6):1280-1290, 2019). NK cells bind to antibodies on the surface of Plasmodium falciparum infected red blood cells (iRBCs) via their Fc receptor, FcγRIIIa (CD16). This interaction induces the release of lytic granules (degranulation) by NK cells, which contain perforin, granzyme B, and granulysin, as well as the secretion of IFNγ. ADCC subsequently limits the growth of Plasmodium falciparum in vitro (Arora et al. Elife:7, e36806, 2018). Because NK cell-mediated ADCC has been shown to be important in malaria parasite clearance and protection, further studies understanding ADCC in malaria are warranted (Hart et al. J Exp Med 216(6):1280-1290, 2019). Therefore, this protocol describes methods to perform an in vitro ADCC functional assay with iRBCs. Specifically, it includes protocols on how to grow and culture iRBCs, how to culture peripheral blood mononuclear cells (PMBCs), and how to do in vitro PBMC assays to measure NK degranulation and IFNγ production as measures of NK ADCC function.


Subject(s)
Leukocytes, Mononuclear , Plasmodium falciparum , Antibody-Dependent Cell Cytotoxicity , Erythrocytes , Killer Cells, Natural
7.
Nat Med ; 28(2): 392-400, 2022 02.
Article in English | MEDLINE | ID: mdl-35102335

ABSTRACT

There is no cure for HIV infection, and lifelong antiretroviral therapy (ART) is required. N-803 is an IL-15 superagonist comprised of an N72D mutant IL-15 molecule attached to its alpha receptor and a human IgG1 fragment designed to increase IL-15 activity. Preclinical studies with both HIV and SIV suggest that the drug has potential to reduce virus reservoirs by activating virus from latency and enhancing effector function. We conducted a phase 1 study of N-803 ( NCT02191098 ) in people living with HIV, the primary objective of which was to assess the safety and tolerability of the drug, with an exploratory objective of assessing the impact on peripheral virus reservoirs. ART-suppressed individuals were enrolled into a dose-escalation study of N-803 in four different cohorts (0.3, 1.0, 3.0 and 6.0 mcg kg-1). Each cohort received three doses total, separated by at least 1 week. We enrolled 16 individuals, of whom 11 completed all three doses. The maximum tolerated dose was 6.0 mcg kg-1. The primary clinical adverse events (AEs) reported were injection site rash and adenopathy, and four participants experienced a grade 1 or grade 2 QTc prolongation. No significant laboratory AEs attributable to N-803 were observed. In exploratory analyses, N-803 was associated with proliferation and/or activation of CD4+ and CD8+ T cells and natural killer cells that peaked at 4 d after dosing. IFN-γ, IP-10, MCP-1 and IL-15 increased during treatment. HIV transcription in memory CD4 T cells and intact proviral DNA initially increased after N-803 treatment; however, there was a small but significant decrease in the frequency of peripheral blood mononuclear cells with an inducible HIV provirus that persisted for up to 6 months after therapy. These data suggest that N-803 administration in ART-suppressed people living with HIV is safe and that larger clinical trials are needed to further investigate the effects of N-803 on HIV reservoirs.


Subject(s)
HIV Infections , HIV-1 , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , HIV Infections/drug therapy , Humans , Interleukin-15/genetics , Leukocytes, Mononuclear , Recombinant Fusion Proteins , Viral Load
8.
Cancer Immunol Res ; 9(11): 1270-1282, 2021 11.
Article in English | MEDLINE | ID: mdl-34452926

ABSTRACT

Metastatic castration-resistant prostate cancer (mCRPC) has been largely resistant to immunotherapy. Natural killer (NK) cells are cytotoxic lymphocytes that detect and kill transformed cells without prior sensitization, and their infiltration into prostate tumors corresponds with an increased overall survival among patients with mCRPC. We sought to harness this knowledge to develop an approach to NK-cell based immunotherapy for mCRPC. We engineered an NK cell line (NK-92MI) to express CD64, the sole human high-affinity IgG Fcγ receptor (FcγR1), and bound these cells with antibodies to provide interchangeable tumor-targeting elements. NK-92MICD64 cells were evaluated for cell-activation mechanisms and antibody-dependent cell-mediated cytotoxicity (ADCC). A combination of mAbs was used to target the prostate tumor antigen tumor-associated calcium signal transducer 2 (TROP2) and the cancer-associated fibroblast marker fibroblast activation protein alpha (FAP). We found that CD64, which is normally expressed by myeloid cells and associates with the adaptor molecule FcRγ, can be expressed by NK-92MI cells and mediate ADCC through an association with CD3ζ. Cytotoxicity from the combination approach was two-fold higher compared to treatment with NK-92MICD64 cells and either mAb alone, and seven-fold higher than NK-92MICD64 cells alone at an effector-target cell ratio of 20:1. The cytotoxic effect was lost when using isotype control antibodies, indicating a selective targeting mechanism. The combination approach demonstrated efficacy in vivo as well and significantly reduced tumor growth compared with the saline control. This combination therapy presents a potential approach for treating mCRPC and could improve immunotherapy response.


Subject(s)
Antibodies, Monoclonal/metabolism , Killer Cells, Natural/metabolism , Prostatic Neoplasms, Castration-Resistant/genetics , Receptors, IgG/metabolism , Tissue Engineering/methods , Animals , Cell Line, Tumor , Humans , Male , Mice , Neoplasm Metastasis , Prostatic Neoplasms, Castration-Resistant/mortality , Survival Analysis
9.
J Immunol ; 207(3): 860-867, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34281999

ABSTRACT

Vaccines and mAbs offer promising strategies to treat substance use disorders (SUDs) and prevent overdose. Despite vaccines and mAbs against SUDs demonstrating proof of efficacy, selectivity, and safety in animal models, it is unknown whether the mechanism of action of these immunotherapeutics relies exclusively on the formation of Ab/drug complexes, or also involves Ab-mediated effector functions. Hence, this study tested whether the efficacy of active and passive immunization against drugs of abuse requires phagocytosis, the intact Fc portion of the anti-drug Ab, FcγRs, or the neonatal FcR (FcRn). The efficacy of a lead vaccine against oxycodone was not diminished in mice after depletion of macrophages or granulocytes. Anti-oxycodone F(ab')2 fragments resulted in lower serum levels of F(ab')2 compared with intact mAbs, and F(ab')2s were not as effective as the parent mAbs in reducing distribution of oxycodone to the brain. The efficacy of vaccines and mAbs against oxycodone was preserved in either FcγIII or FcγI-IV ablated mice, suggesting that FcγRs are not required for Ab efficacy. Finally, both active and passive immunization against oxycodone in FcRn-/- mice yielded reduced efficacy compared with wild-type control mice. These data identified a role for FcRn, but not for phagocytosis or Fc-dependent effector functions, in mediating the efficacy of vaccines and mAbs against SUD. This study supports rational design of vaccines and mAbs engineered for maximal neutralization activity and optimal FcRn binding.


Subject(s)
Opioid-Related Disorders , Vaccines , Animals , Antibodies, Monoclonal , Mice , Oxycodone
10.
J Immunol ; 206(5): 931-935, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33441437

ABSTRACT

The magnitude of SARS-CoV-2-specific T cell responses correlates inversely with human disease severity, suggesting T cell involvement in primary control. Whereas many COVID-19 vaccines focus on establishing humoral immunity to viral spike protein, vaccine-elicited T cell immunity may bolster durable protection or cross-reactivity with viral variants. To better enable mechanistic and vaccination studies in mice, we identified a dominant CD8 T cell SARS-CoV-2 nucleoprotein epitope. Infection of human ACE2 transgenic mice with SARS-CoV-2 elicited robust responses to H2-Db/N219-227, and 40% of HLA-A*02+ COVID-19 PBMC samples isolated from hospitalized patients responded to this peptide in culture. In mice, i.m. prime-boost nucleoprotein vaccination with heterologous vectors favored systemic CD8 T cell responses, whereas intranasal boosting favored respiratory immunity. In contrast, a single i.v. immunization with recombinant adenovirus established robust CD8 T cell memory both systemically and in the respiratory mucosa.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , COVID-19 Vaccines/immunology , COVID-19/immunology , COVID-19/prevention & control , Epitopes, T-Lymphocyte/immunology , SARS-CoV-2/immunology , Vaccination/methods , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/virology , Cells, Cultured , Coronavirus Nucleocapsid Proteins/immunology , Disease Models, Animal , Female , Genetic Vectors/immunology , HLA-A2 Antigen/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic
11.
J Infect Dis ; 222(7): 1170-1179, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32386415

ABSTRACT

BACKGROUND: Chronic active Epstein-Barr virus (CAEBV) presents with high levels of viral genomes in blood and tissue infiltration with Epstein-Barr virus (EBV)-positive lymphocytes. The pathogenesis of CAEBV is poorly understood. METHODS: We evaluated 2 patients with natural killer (NK) cell CAEBV and studied their NK cell phenotype and signaling pathways in cells. RESULTS: Both patients had increased numbers of NK cells, EBV predominantly in NK cells, and immature NK cells in the blood. Both patients had increased phosphorylation of Akt, S6, and STAT1 in NK cells, and increased total STAT1. Treatment of 1 patient with sirolimus reduced phosphorylation of S6 in T and B cells, but not in NK cells and did not reduce levels of NK cells or EBV DNA in the blood. Treatment of both patients' cells with JAK inhibitors in vitro reduced phosphorylated STAT1 to normal. Patients with T- or B-cell CAEBV had increased phosphorylation of Akt and S6 in NK cells, but no increase in total STAT1. CONCLUSIONS: The increase in phosphorylated Akt, S6, and STAT1, as well as immature NK cells describe a new phenotype for NK cell CAEBV. The reduction of STAT1 phosphorylation in their NK cells with JAK inhibitors suggests a novel approach to therapy.


Subject(s)
Epstein-Barr Virus Infections/diagnosis , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphoproliferative Disorders/diagnosis , Signal Transduction , Adolescent , Adult , B-Lymphocytes/immunology , B-Lymphocytes/virology , Chronic Disease , Epstein-Barr Virus Infections/virology , Female , Humans , Killer Cells, Natural/virology , Lymphoproliferative Disorders/virology , Male , Phosphorylation , Prospective Studies , Proto-Oncogene Proteins c-akt/metabolism , STAT1 Transcription Factor/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/virology
12.
Mol Ther Methods Clin Dev ; 16: 1-10, 2020 Mar 13.
Article in English | MEDLINE | ID: mdl-31673565

ABSTRACT

Chimeric antigen receptor (CAR)-T cells show great promise in treating cancers and viral infections. However, most protocols developed to expand T cells require relatively long periods of time in culture, potentially leading to progression toward populations of terminally differentiated effector memory cells. Here, we describe in detail a 9-day protocol for CAR gene transduction and expansion of primary rhesus macaque peripheral blood mononuclear cells (PBMCs). Cells produced and expanded with this method show high levels of viability, high levels of co-expression of two transduced genes, retention of the central memory phenotype, and sufficient quantity for immunotherapeutic infusion of 1-2 × 108 cells/kg in a 10 kg rhesus macaque. This 9-day protocol may be broadly used for CAR-T cell and other T cell immunotherapy approaches to decrease culture time and increase maintenance of central memory populations.

13.
Malar J ; 18(1): 321, 2019 Sep 18.
Article in English | MEDLINE | ID: mdl-31533835

ABSTRACT

Natural killer (NK) cells are important innate effector cells that are well described in their ability to kill virally-infected cells and tumors. However, there is increasing appreciation for the role of NK cells in the control of other pathogens, including intracellular parasites such as Plasmodium, the cause of malaria. NK cells may be beneficial during the early phase of Plasmodium infection-prior to the activation and expansion of antigen-specific T cells-through cooperation with myeloid cells to produce inflammatory cytokines like IFNγ. Recent work has defined how Plasmodium can activate NK cells to respond with natural cytotoxicity, and inhibit the growth of parasites via antibody-dependent cellular cytotoxicity mechanisms (ADCC). A specialized subset of adaptive NK cells that are negative for the Fc receptor γ chain have enhanced ADCC function and correlate with protection from malaria. Additionally, production of the regulatory cytokine IL-10 by NK cells prevents overt pathology and death during experimental cerebral malaria. Now that conditional NK cell mouse models have been developed, previous studies need to be reevaluated in the context of what is now known about other immune populations with similarity to NK cells (i.e., NKT cells and type I innate lymphoid cells). This brief review summarizes recent findings which support the potentially beneficial roles of NK cells during Plasmodium infection in mice and humans. Also highlighted are how the actions of NK cells can be explored using new experimental strategies, and the potential to harness NK cell function in vaccination regimens.


Subject(s)
Immunity, Innate , Killer Cells, Natural/immunology , Plasmodium/physiology , Animals , Humans , Killer Cells, Natural/parasitology , Mice
14.
Immunity ; 51(4): 750-765.e10, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31492649

ABSTRACT

Immunity that controls parasitemia and inflammation during Plasmodium falciparum (Pf) malaria can be acquired with repeated infections. A limited understanding of this complex immune response impedes the development of vaccines and adjunctive therapies. We conducted a prospective systems biology study of children who differed in their ability to control parasitemia and fever following Pf infection. By integrating whole-blood transcriptomics, flow-cytometric analysis, and plasma cytokine and antibody profiles, we demonstrate that a pre-infection signature of B cell enrichment, upregulation of T helper type 1 (Th1) and Th2 cell-associated pathways, including interferon responses, and p53 activation associated with control of malarial fever and coordinated with Pf-specific immunoglobulin G (IgG) and Fc receptor activation to control parasitemia. Our hypothesis-generating approach identified host molecules that may contribute to differential clinical outcomes during Pf infection. As a proof of concept, we have shown that enhanced p53 expression in monocytes attenuated Plasmodium-induced inflammation and predicted protection from fever.


Subject(s)
B-Lymphocytes/immunology , Blood Proteins/metabolism , Inflammation/metabolism , Malaria, Falciparum/metabolism , Plasmodium falciparum/physiology , Th1 Cells/immunology , Th2 Cells/immunology , Tumor Suppressor Protein p53/metabolism , Adolescent , Adult , Animals , Antibodies, Protozoan/metabolism , Child , Child, Preschool , Disease Resistance , Female , Gene Expression Profiling , Humans , Infant , Interferons/metabolism , Male , Mice , Mice, Inbred C57BL , Prospective Studies , Receptors, Fc/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics , Young Adult
15.
J Exp Med ; 216(6): 1280-1290, 2019 06 03.
Article in English | MEDLINE | ID: mdl-30979790

ABSTRACT

How antibodies naturally acquired during Plasmodium falciparum infection provide clinical immunity to blood-stage malaria is unclear. We studied the function of natural killer (NK) cells in people living in a malaria-endemic region of Mali. Multi-parameter flow cytometry revealed a high proportion of adaptive NK cells, which are defined by the loss of transcription factor PLZF and Fc receptor γ-chain. Adaptive NK cells dominated antibody-dependent cellular cytotoxicity responses, and their frequency within total NK cells correlated with lower parasitemia and resistance to malaria. P. falciparum-infected RBCs induced NK cell degranulation after addition of plasma from malaria-resistant individuals. Malaria-susceptible subjects with the largest increase in PLZF-negative NK cells during the transmission season had improved odds of resistance during the subsequent season. Thus, antibody-dependent lysis of P. falciparum-infected RBCs by NK cells may be a mechanism of acquired immunity to malaria. Consideration of antibody-dependent NK cell responses to P. falciparum antigens is therefore warranted in the design of malaria vaccines.


Subject(s)
Killer Cells, Natural/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/physiology , Adolescent , Antibodies, Protozoan/immunology , CD56 Antigen/metabolism , Child , Child, Preschool , Erythrocytes/parasitology , Humans , Promyelocytic Leukemia Zinc Finger Protein/metabolism , Receptors, IgG/metabolism , Young Adult
16.
Elife ; 72018 06 26.
Article in English | MEDLINE | ID: mdl-29943728

ABSTRACT

Antibodies acquired naturally through repeated exposure to Plasmodium falciparum are essential in the control of blood-stage malaria. Antibody-dependent functions may include neutralization of parasite-host interactions, complement activation, and activation of Fc receptor functions. A role of antibody-dependent cellular cytotoxicity (ADCC) by natural killer (NK) cells in protection from malaria has not been established. Here we show that IgG isolated from adults living in a malaria-endemic region activated ADCC by primary human NK cells, which lysed infected red blood cells (RBCs) and inhibited parasite growth in an in vitro assay for ADCC-dependent growth inhibition. RBC lysis by NK cells was highly selective for infected RBCs in a mixed culture with uninfected RBCs. Human antibodies to P. falciparum antigens PfEMP1 and RIFIN were sufficient to promote NK-dependent growth inhibition. As these results implicate acquired immunity through NK-mediated ADCC, antibody-based vaccines that target bloodstream parasites should consider this new mechanism of action.


Subject(s)
Antibodies, Protozoan/pharmacology , Antibody-Dependent Cell Cytotoxicity , Immunoglobulin G/pharmacology , Killer Cells, Natural/drug effects , Malaria, Falciparum/immunology , Plasmodium falciparum/drug effects , Antibodies, Protozoan/isolation & purification , Antigens, Protozoan/chemistry , Antigens, Protozoan/immunology , Coculture Techniques , Erythrocytes/parasitology , Hemolysis , Humans , Immune Sera/chemistry , Immunity, Cellular/drug effects , Immunoglobulin G/isolation & purification , Killer Cells, Natural/immunology , Killer Cells, Natural/parasitology , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Parasitic Sensitivity Tests , Plasmodium falciparum/growth & development , Plasmodium falciparum/immunology , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Time-Lapse Imaging
17.
Immunity ; 48(4): 760-772.e4, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29625893

ABSTRACT

Cerebral malaria is a deadly complication of Plasmodium infection and involves blood brain barrier (BBB) disruption following infiltration of white blood cells. During experimental cerebral malaria (ECM), mice inoculated with Plasmodium berghei ANKA-infected red blood cells develop a fatal CM-like disease caused by CD8+ T cell-mediated pathology. We found that treatment with interleukin-15 complex (IL-15C) prevented ECM, whereas IL-2C treatment had no effect. IL-15C-expanded natural killer (NK) cells were necessary and sufficient for protection against ECM. IL-15C treatment also decreased CD8+ T cell activation in the brain and prevented BBB breakdown without influencing parasite load. IL-15C induced NK cells to express IL-10, which was required for IL-15C-mediated protection against ECM. Finally, we show that ALT-803, a modified human IL-15C, mediates similar induction of IL-10 in NK cells and protection against ECM. These data identify a regulatory role for cytokine-stimulated NK cells in the prevention of a pathogenic immune response.


Subject(s)
Interleukin-10/immunology , Interleukin-15/immunology , Killer Cells, Natural/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Proteins/pharmacology , Animals , Blood-Brain Barrier/pathology , Brain/immunology , Brain/pathology , CD8-Positive T-Lymphocytes/immunology , Interleukin-10/biosynthesis , Lymphocyte Activation/immunology , Malaria, Cerebral/microbiology , Malaria, Cerebral/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Recombinant Fusion Proteins
18.
PLoS Pathog ; 12(12): e1006022, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27907215

ABSTRACT

Cerebral malaria (CM) is a severe complication of Plasmodium falciparum infection that results in thousands of deaths each year, mostly in African children. The in vivo mechanisms underlying this fatal condition are not entirely understood. Using the animal model of experimental cerebral malaria (ECM), we sought mechanistic insights into the pathogenesis of CM. Fatal disease was associated with alterations in tight junction proteins, vascular breakdown in the meninges / parenchyma, edema, and ultimately neuronal cell death in the brainstem, which is consistent with cerebral herniation as a cause of death. At the peak of ECM, we revealed using intravital two-photon microscopy that myelomonocytic cells and parasite-specific CD8+ T cells associated primarily with the luminal surface of CNS blood vessels. Myelomonocytic cells participated in the removal of parasitized red blood cells (pRBCs) from cerebral blood vessels, but were not required for the disease. Interestingly, the majority of disease-inducing parasite-specific CD8+ T cells interacted with the lumen of brain vascular endothelial cells (ECs), where they were observed surveying, dividing, and arresting in a cognate peptide-MHC I dependent manner. These activities were critically dependent on IFN-γ, which was responsible for activating cerebrovascular ECs to upregulate adhesion and antigen-presenting molecules. Importantly, parasite-specific CD8+ T cell interactions with cerebral vessels were impaired in chimeric mice rendered unable to present EC antigens on MHC I, and these mice were in turn resistant to fatal brainstem pathology. Moreover, anti-adhesion molecule (LFA-1 / VLA-4) therapy prevented fatal disease by rapidly displacing luminal CD8+ T cells from cerebrovascular ECs without affecting extravascular T cells. These in vivo data demonstrate that parasite-specific CD8+ T cell-induced fatal vascular breakdown and subsequent neuronal death during ECM is associated with luminal, antigen-dependent interactions with cerebrovasculature.


Subject(s)
Blood Vessels/pathology , Blood-Brain Barrier/pathology , Brain Stem/pathology , CD8-Positive T-Lymphocytes/pathology , Malaria, Cerebral/pathology , Animals , Disease Models, Animal , Flow Cytometry , Immunohistochemistry , Malaria, Cerebral/immunology , Mice , Mice, Transgenic , Plasmodium berghei
19.
J Immunol ; 197(10): 3841-3849, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27798155

ABSTRACT

Plasmodium falciparum malaria is a deadly infectious disease in which Abs play a critical role in naturally acquired immunity. However, the specificity and nature of Abs elicited in response to malaria are only partially understood. Autoreactivity and polyreactivity are common features of Ab responses in several infections and were suggested to contribute to effective pathogen-specific Ab responses. In this article, we report on the regulation of B cells expressing the inherently autoreactive VH4-34 H chain (identified by the 9G4 mAb) and 9G4+ plasma IgG in adults and children living in a P. falciparum malaria-endemic area in West Africa. The frequency of 9G4+ peripheral blood CD19+ B cells was similar in United States adults and African adults and children; however, more 9G4+ B cells appeared in classical and atypical memory B cell compartments in African children and adults compared with United States adults. The levels of 9G4+ IgG increased following acute febrile malaria but did not increase with age as humoral immunity is acquired or correlate with protection from acute disease. This was the case, even though a portion of 9G4+ B cells acquired phenotypes of atypical and classical memory B cells and 9G4+ IgG contained equivalent numbers of somatic hypermutations compared with all other VHs, a characteristic of secondary Ab repertoire diversification in response to Ag stimulation. Determining the origin and function of 9G4+ B cells and 9G4+ IgG in malaria may contribute to a better understanding of the varied roles of autoreactivity in infectious diseases.


Subject(s)
Antibodies, Protozoan/blood , Autoimmunity , B-Lymphocytes/immunology , Immunoglobulin G/blood , Immunoglobulin Heavy Chains/immunology , Malaria, Falciparum/immunology , Adult , Africa, Western/epidemiology , Antibodies, Protozoan/immunology , B-Lymphocytes/chemistry , Child , Endemic Diseases , Gene Expression Regulation , Humans , Immunity, Humoral , Immunoglobulin G/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin M/blood , Immunoglobulin M/immunology , Malaria/epidemiology , Malaria/immunology , Malaria, Falciparum/epidemiology , Phenotype , Plasmodium falciparum/immunology , United States/epidemiology
20.
Proc Natl Acad Sci U S A ; 112(42): 13075-80, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26438846

ABSTRACT

The most deadly complication of Plasmodium falciparum infection is cerebral malaria (CM) with a case fatality rate of 15-25% in African children despite effective antimalarial chemotherapy. There are no adjunctive treatments for CM, so there is an urgent need to identify new targets for therapy. Here we show that the glutamine analog 6-diazo-5-oxo-L-norleucine (DON) rescues mice from CM when administered late in the infection a time at which mice already are suffering blood-brain barrier dysfunction, brain swelling, and hemorrhaging accompanied by accumulation of parasite-specific CD8(+) effector T cells and infected red blood cells in the brain. Remarkably, within hours of DON treatment mice showed blood-brain barrier integrity, reduced brain swelling, decreased function of activated effector CD8(+) T cells in the brain, and levels of brain metabolites that resembled those in uninfected mice. These results suggest DON as a strong candidate for an effective adjunctive therapy for CM in African children.


Subject(s)
Antimalarials/therapeutic use , Diazooxonorleucine/therapeutic use , Glutamine/metabolism , Malaria, Cerebral/drug therapy , Malaria, Falciparum/drug therapy , Animals , Antimalarials/pharmacology , Blood-Brain Barrier/drug effects , Diazooxonorleucine/pharmacology , Malaria, Cerebral/metabolism , Malaria, Falciparum/metabolism , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...