Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Trends Parasitol ; 40(4): 275-277, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38485579

RESUMO

In a recent publication, Ramalho et al. investigated monocyte-derived dendritic cell (MODC) mobilization in response to Plasmodium infection. The authors showed that elevated levels of itaconate in MODCs results in reduced CD8 T cell activation and that the absence of itaconate is associated with enhanced parasite control.


Assuntos
Antimaláricos , Succinatos , Antimaláricos/farmacologia , Antimaláricos/uso terapêutico , Monócitos/metabolismo , Linfócitos T CD8-Positivos , Células Dendríticas
2.
J Immunol ; 212(9): 1467-1478, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38477614

RESUMO

Development of Plasmodium-specific humoral immunity is critically dependent on CD4 Th cell responses and germinal center (GC) reactions during blood-stage Plasmodium infection. IL-21, a cytokine primarily produced by CD4 T cells, is an essential regulator of affinity maturation, isotype class-switching, B cell differentiation, and maintenance of GC reactions in response to many infection and immunization models. In models of experimental malaria, mice deficient in IL-21 or its receptor IL-21R fail to develop memory B cell populations and are not protected against secondary infection. However, whether sustained IL-21 signaling in ongoing GCs is required for maintaining GC magnitude, organization, and output is unclear. In this study, we report that CD4+ Th cells maintain IL-21 expression after resolution of primary Plasmodium yoelii infection. We generated an inducible knockout mouse model that enabled cell type-specific and timed deletion of IL-21 in peripheral, mature CD4 T cells. We found that persistence of IL-21 signaling in active GCs had no impact on the magnitude of GC reactions or their capacity to produce memory B cell populations. However, the memory B cells generated in the absence of IL-21 exhibited reduced recall function upon challenge. Our data support that IL-21 prevents premature cellular dissolution within the GC and promotes stringency of selective pressures during B cell fate determination required to produce high-quality Plasmodium-specific memory B cells. These data are additionally consistent with a temporal requirement for IL-21 in fine-tuning humoral immune memory responses during experimental malaria.


Assuntos
Linfócitos T CD4-Positivos , Interleucinas , Malária , Plasmodium , Animais , Camundongos , Linfócitos B , Linfócitos T CD4-Positivos/metabolismo , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Malária/imunologia , Células B de Memória/imunologia , Camundongos Endogâmicos C57BL , Plasmodium/imunologia
3.
Viruses ; 15(6)2023 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-37376652

RESUMO

Macrophages are critical in the pathogenesis of a diverse group of viral pathogens, both as targets of infection and for eliciting primary defense mechanisms. Our prior in vitro work identified that CD40 signaling in murine peritoneal macrophages protects against several RNA viruses by eliciting IL-12, which stimulates the production of interferon gamma (IFN-γ). Here, we examine the role of CD40 signaling in vivo. We show that CD40 signaling is a critical, but currently poorly appreciated, component of the innate immune response using two distinct infectious agents: mouse-adapted influenza A virus (IAV, PR8) and recombinant VSV encoding the Ebola virus glycoprotein (rVSV-EBOV GP). We find that stimulation of CD40 signaling decreases early IAV titers, whereas loss of CD40 elevated early titers and compromised lung function by day 3 of infection. Protection conferred by CD40 signaling against IAV is dependent on IFN-γ production, consistent with our in vitro studies. Using rVSV-EBOV GP that serves as a low-biocontainment model of filovirus infection, we demonstrate that macrophages are a CD40-expressing population critical for protection within the peritoneum and T-cells are the key source of CD40L (CD154). These experiments reveal the in vivo mechanisms by which CD40 signaling in macrophages regulates the early host responses to RNA virus infection and highlight how CD40 agonists currently under investigation for clinical use may function as a novel class of broad antiviral treatments.


Assuntos
Antígenos CD40 , Infecções por Vírus de RNA , Vírus de RNA , Animais , Camundongos , Antígenos CD40/metabolismo , Interferon gama , Macrófagos , Infecções por Vírus de RNA/imunologia
4.
Front Cell Infect Microbiol ; 13: 1132647, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37009496

RESUMO

Plasmodium ookinetes use an invasive apparatus to invade mosquito midguts, and tubulins are the major structural proteins of this apical complex. We examined the role of tubulins in malaria transmission to mosquitoes. Our results demonstrate that the rabbit polyclonal antibodies (pAb) against human α-tubulin significantly reduced the number of P. falciparum oocysts in Anopheles gambiae midguts, while rabbit pAb against human ß-tubulin did not. Further studies showed that pAb, specifically against P. falciparum α-tubulin-1, also significantly limited P. falciparum transmission to mosquitoes. We also generated mouse monoclonal antibodies (mAb) using recombinant P. falciparum α-tubulin-1. Out of 16 mAb, two mAb, A3 and A16, blocked P. falciparum transmission with EC50 of 12 µg/ml and 2.8 µg/ml. The epitopes of A3 and A16 were determined to be a conformational and linear sequence of EAREDLAALEKDYEE, respectively. To understand the mechanism of the antibody-blocking activity, we studied the accessibility of live ookinete α-tubulin-1 to antibodies and its interaction with mosquito midgut proteins. Immunofluorescent assays showed that pAb could bind to the apical complex of live ookinetes. Moreover, both ELISA and pull-down assays demonstrated that insect cell-expressed mosquito midgut protein, fibrinogen-related protein 1 (FREP1), interacts with P. falciparum α-tubulin-1. Since ookinete invasion is directional, we conclude that the interaction between Anopheles FREP1 protein and Plasmodium α-tubulin-1 anchors and orients the ookinete invasive apparatus towards the midgut PM and promotes the efficient parasite infection in the mosquito.


Assuntos
Anopheles , Malária Falciparum , Malária , Plasmodium , Animais , Camundongos , Coelhos , Humanos , Tubulina (Proteína)/metabolismo , Plasmodium falciparum , Mosquitos Vetores , Malária Falciparum/parasitologia , Anopheles/parasitologia
5.
J Exp Med ; 220(2)2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36515679

RESUMO

The longevity of plasma cells is dependent on their ability to access and reside in so-called niches that are predominantly located in the bone marrow. Here, by employing a traceable method to label recently generated plasma cells, we showed that homeostatic plasma cells in the bone marrow and spleen were continuously replenished by newly generated B220hiMHC-IIhi populations that progressively differentiated into B220loMHC-IIlo long-lived plasma cell (LLPC) populations. We also found that, in the bone marrow, germinal center (GC)-independent and GC-dependent plasma cells decayed similarly upon NP-CGG engagement, and both entered the B220loMHC-IIlo LLPC pool. Compared with NP+B220hiMHC-IIhi plasma cells, NP+B220loMHC-IIlo cells were more immobilized in the bone marrow niches and showed better survival potential. Thus, our results suggest that the adhesion status of bone marrow plasma cells is dynamically altered during their differentiation and is associated with provision of survival signals.


Assuntos
Medula Óssea , Plasmócitos , Plasmócitos/metabolismo , Diferenciação Celular , Células da Medula Óssea , Centro Germinativo , Sobrevivência Celular
6.
J Exp Med ; 220(2)2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36512034

RESUMO

In contrast to a second dose of the SARS-CoV-2 mRNA vaccine, a third dose elicits potent neutralizing activity against the Omicron variant. To address the underlying mechanism for this differential antibody response, we examined spike receptor-binding domain (RBD)-specific memory B cells in vaccinated individuals. Frequency of Omicron-reactive memory B cells increased ∼9 mo after the second vaccine dose. These memory B cells show an altered distribution of epitopes from pre-second memory B cells, presumably due to an antibody feedback mechanism. This hypothesis was tested using mouse models, showing that an addition or a depletion of RBD-induced serum antibodies results in a concomitant increase or decrease, respectively, of Omicron-reactive germinal center (GC) and memory B cells. Our data suggest that pre-generated antibodies modulate the selection of GC and subsequent memory B cells after the second vaccine dose, accumulating more Omicron-reactive memory B cells over time, which contributes to the generation of Omicron-neutralizing antibodies elicited by the third vaccine dose.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Animais , Camundongos , Humanos , Retroalimentação , Células B de Memória , SARS-CoV-2 , COVID-19/prevenção & controle , RNA Mensageiro , Anticorpos Neutralizantes , Anticorpos Antivirais
7.
Pathogens ; 11(5)2022 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-35631044

RESUMO

Protective immunity against blood-stage Plasmodium infection and the disease malaria depends on antibodies secreted from high-affinity B cells selected during the germinal center (GC) response. The induction and stability of the GC response require the activation and direct cell-cell communication between parasite-specific CD4 helper T cells and B cells. However, cytokines secreted by helper T cells, B cells, and multiple other innate and adaptive immune cells also contribute to regulating the magnitude and protective functions of GC-dependent humoral immune responses. Here, we briefly review emerging data supporting the finding that specific cytokines can exhibit temporally distinct and context-dependent influences on the induction and maintenance of antimalarial humoral immunity.

8.
Cell Rep ; 37(5): 109956, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34731605

RESUMO

Circulating memory CD8 T cell trafficking and protective capacity during liver-stage malaria infection remains undefined. We find that effector memory CD8 T cells (Tem) infiltrate the liver within 6 hours after malarial or bacterial infections and mediate pathogen clearance. Tem recruitment coincides with rapid transcriptional upregulation of inflammatory genes in Plasmodium-infected livers. Recruitment requires CD8 T cell-intrinsic LFA-1 expression and the presence of liver phagocytes. Rapid Tem liver infiltration is distinct from recruitment to other non-lymphoid tissues in that it occurs both in the absence of liver tissue resident memory "sensing-and-alarm" function and ∼42 hours earlier than in lung infection by influenza virus. These data demonstrate relevance for Tem in protection against malaria and provide generalizable mechanistic insights germane to control of liver infections.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Memória Imunológica , Fígado/imunologia , Malária/imunologia , Plasmodium berghei/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/microbiologia , Linfócitos T CD8-Positivos/parasitologia , Modelos Animais de Doenças , Feminino , Interações Hospedeiro-Parasita , Listeria monocytogenes/imunologia , Listeria monocytogenes/patogenicidade , Listeriose/sangue , Listeriose/imunologia , Listeriose/microbiologia , Fígado/metabolismo , Fígado/microbiologia , Fígado/parasitologia , Antígeno-1 Associado à Função Linfocitária/metabolismo , Malária/sangue , Malária/parasitologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Carga Parasitária , Fagócitos/imunologia , Fagócitos/metabolismo , Fagócitos/microbiologia , Fagócitos/parasitologia , Plasmodium berghei/patogenicidade , Fatores de Tempo
9.
Cell Rep ; 36(8): 109586, 2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34433049

RESUMO

During acute malaria, most individuals mount robust inflammatory responses that limit parasite burden. However, long-lived sterilizing anti-malarial memory responses are not efficiently induced, even following repeated Plasmodium exposures. Using multiple Plasmodium species, genetically modified parasites, and combinations of host genetic and pharmacologic approaches, we find that the deposition of the malarial pigment hemozoin directly limits the abundance and capacity of conventional type 1 dendritic cells to prime helper T cell responses. Hemozoin-induced dendritic cell dysfunction results in aberrant Plasmodium-specific CD4 T follicular helper cell differentiation, which constrains memory B cell and long-lived plasma cell formation. Mechanistically, we identify that dendritic cell-intrinsic NLRP3 inflammasome activation reduces conventional type 1 dendritic cell abundance, phagocytosis, and T cell priming functions in vivo. These data identify biological consequences of hemozoin deposition during malaria and highlight the capacity of the malarial pigment to program immune evasion during the earliest events following an initial Plasmodium exposure.


Assuntos
Hemeproteínas/farmacologia , Inflamassomos/efeitos dos fármacos , Ativação Linfocitária/imunologia , Malária/tratamento farmacológico , Animais , Antimaláricos/farmacologia , Células Dendríticas/imunologia , Inflamassomos/metabolismo , Malária/imunologia , Células B de Memória/efeitos dos fármacos , Células B de Memória/imunologia , Camundongos Endogâmicos C57BL , Fagocitose/fisiologia , Plasmodium/imunologia , Linfócitos T Auxiliares-Indutores/imunologia
10.
Elife ; 102021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-34143731

RESUMO

Protective lung tissue-resident memory CD8+T cells (Trm) form after influenza A virus (IAV) infection. We show that IAV infection of mice generates CD69+CD103+and other memory CD8+T cell populations in lung-draining mediastinal lymph nodes (mLNs) from circulating naive or memory CD8+T cells. Repeated antigen exposure, mimicking seasonal IAV infections, generates quaternary memory (4M) CD8+T cells that protect mLN from viral infection better than 1M CD8+T cells. Better protection by 4M CD8+T cells associates with enhanced granzyme A/B expression and stable maintenance of mLN CD69+CD103+4M CD8+T cells, vs the steady decline of CD69+CD103+1M CD8+T cells, paralleling the durability of protective CD69+CD103+4M vs 1M in the lung after IAV infection. Coordinated upregulation in canonical Trm-associated genes occurs in circulating 4M vs 1M populations without the enrichment of canonical downregulated Trm genes. Thus, repeated antigen exposure arms circulating memory CD8+T cells with enhanced capacity to form long-lived populations of Trm that enhance control of viral infections of the mLN.


Assuntos
Linfócitos T CD8-Positivos , Linfonodos , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Antígenos CD/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Células Cultivadas , Feminino , Vírus da Influenza A/imunologia , Pulmão/citologia , Pulmão/imunologia , Linfonodos/citologia , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/imunologia , Transcriptoma/genética
11.
J Exp Med ; 218(6)2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-33830176

RESUMO

Antimalarial antibody responses are essential for mediating the clearance of Plasmodium parasite-infected RBCs from infected hosts. However, the rapid appearance of large numbers of plasmablasts in Plasmodium-infected hosts can suppress the development and function of durable humoral immunity. Here, we identify that the formation of plasmablast populations in Plasmodium-infected mice is mechanistically linked to both hemolysis-induced exposure of phosphatidylserine on damaged RBCs and inflammatory cues. We also show that virus and Trypanosoma infections known to trigger hemolytic anemia and high-grade inflammation also induce exuberant plasmablast responses. The induction of hemolysis or administration of RBC membrane ghosts increases plasmablast differentiation. The phosphatidylserine receptor Axl is critical for optimal plasmablast formation, and blocking phosphatidylserine limits plasmablast expansions and reduces Plasmodium parasite burden in vivo. Our findings support that strategies aimed at modulating polyclonal B cell activation and phosphatidylserine exposure may improve immune responses against Plasmodium parasites and potentially other infectious diseases that are associated with anemia.


Assuntos
Diferenciação Celular/imunologia , Hemólise/imunologia , Fosfatidilserinas/imunologia , Plasmócitos/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Antimaláricos/imunologia , Linfócitos B/imunologia , Linfócitos B/parasitologia , Células Cultivadas , Eritrócitos/imunologia , Eritrócitos/parasitologia , Humanos , Imunidade Humoral/imunologia , Malária/imunologia , Malária/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmócitos/parasitologia , Plasmodium yoelii/imunologia
12.
Microbes Infect ; 23(4-5): 104807, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33684519

RESUMO

Humoral immunity is critical for limiting Plasmodium parasite infections and the severity of malaria. Naturally acquired immunity against malaria occurs inefficiently and protection is relatively short-lived. Here we review recent advances and explore emerging hypotheses regarding the molecular and cellular pathways that regulate Plasmodium parasite-specific B cell responses and durable anti-malarial humoral immunity.


Assuntos
Imunidade Humoral , Malária/imunologia , Humanos , Fatores de Tempo
13.
PLoS Pathog ; 17(2): e1009288, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33529242

RESUMO

Immunity against malaria depends on germinal center (GC)-derived antibody responses that are orchestrated by T follicular helper (TFH) cells. Emerging data show that the regulatory cytokine IL-10 plays an essential role in promoting GC B cell responses during both experimental malaria and virus infections. Here we investigated the cellular source and temporal role of IL-10, and whether IL-10 additionally signals to CD4 T-cells to support anti-Plasmodium humoral immunity. Distinct from reports of virus infection, we found that IL-10 was expressed by conventional, Foxp3-negative effector CD4 T cells and functioned in a B cell-intrinsic manner only during the first 96 hours of Plasmodium infection to support humoral immunity. The critical functions of IL-10 manifested only before the orchestration of GC responses and were primarily localized outside of B cell follicles. Mechanistically, our studies showed that the rapid and transient provision of IL-10 promoted B cell expression of anti-apoptotic factors, MHC class II, CD83, and cell-cell adhesion proteins that are essential for B cell survival and interaction with CD4 T cells. Together, our data reveal temporal features and mechanisms by which IL-10 critically supports humoral immunity during blood-stage Plasmodium infection, information that may be useful for developing new strategies designed to lessen the burden of malaria.


Assuntos
Formação de Anticorpos/imunologia , Antimaláricos/imunologia , Linfócitos T CD4-Positivos/imunologia , Interleucina-10/metabolismo , Ativação Linfocitária/imunologia , Malária/imunologia , Plasmodium yoelii/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Citocinas/metabolismo , Malária/metabolismo , Malária/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo
14.
J Immunol ; 206(6): 1171-1180, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33514512

RESUMO

The dysregulated sepsis-induced cytokine storm evoked during systemic infection consists of biphasic and interconnected pro- and anti-inflammatory responses. The contrasting inflammatory cytokine responses determine the severity of the septic event, lymphopenia, host survival, and the ensuing long-lasting immunoparalysis state. NK cells, because of their capacity to elaborate pro- (i.e., IFN-γ) and anti-inflammatory (i.e., IL-10) responses, exist at the inflection of sepsis-induced inflammatory responses. Thus, NK cell activity could be beneficial or detrimental during sepsis. In this study, we demonstrate that murine NK cells promote host survival during sepsis by limiting the scope and duration of the cytokine storm. Specifically, NK cell-derived IL-10, produced in response to IL-15, is relevant to clinical manifestations in septic patients and critical for survival during sepsis. This role of NK cells demonstrates that regulatory mechanisms of classical inflammatory cells are beneficial and critical for controlling systemic inflammation, a notion relevant for therapeutic interventions during dysregulated infection-induced inflammatory responses.


Assuntos
Síndrome da Liberação de Citocina/imunologia , Interleucina-10/metabolismo , Células Matadoras Naturais/imunologia , Sepse/complicações , Animais , Síndrome da Liberação de Citocina/sangue , Síndrome da Liberação de Citocina/diagnóstico , Humanos , Interferon gama/metabolismo , Interleucina-10/genética , Interleucina-15/metabolismo , Células Matadoras Naturais/metabolismo , Camundongos , Camundongos Transgênicos , Sepse/sangue , Sepse/diagnóstico , Sepse/imunologia , Índice de Gravidade de Doença , Transdução de Sinais/imunologia
15.
J Leukoc Biol ; 109(2): 309-325, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32441445

RESUMO

Many acute viral infections target tissue Mϕs, yet the mechanisms of Mϕ-mediated control of viruses are poorly understood. Here, we report that CD40 expressed by peritoneal Mϕs restricts early infection of a broad range of RNA viruses. Loss of CD40 expression enhanced virus replication as early as 12-24 h of infection and, conversely, stimulation of CD40 signaling with an agonistic Ab blocked infection. With peritoneal cell populations infected with the filovirus, wild-type (WT) Ebola virus (EBOV), or a BSL2 model virus, recombinant vesicular stomatitis virus encoding Ebola virus glycoprotein (rVSV/EBOV GP), we examined the mechanism conferring protection. Here, we demonstrate that restricted virus replication in Mϕs required CD154/CD40 interactions that stimulated IL-12 production through TRAF6-dependent signaling. In turn, IL-12 production resulted in IFN-γ production, which induced proinflammatory polarization of Mϕs, protecting the cells from infection. These CD40-dependent events protected mice against virus challenge. CD40-/- mice were exquisitely sensitive to intraperitoneal challenge with a dose of rVSV/EBOV GP that was sublethal to CD40+/+ mice, exhibiting viremia within 12 h of infection and rapidly succumbing to infection. This study identifies a previously unappreciated role for Mϕ-intrinsic CD40 signaling in controlling acute virus infection.


Assuntos
Antígenos CD40/metabolismo , Imunidade Inata , Macrófagos/imunologia , Macrófagos/virologia , Vírus de RNA/fisiologia , Transdução de Sinais , Viroses/imunologia , Replicação Viral/fisiologia , Doença Aguda , Animais , Ligante de CD40/metabolismo , Ebolavirus/fisiologia , Glicoproteínas/imunologia , Humanos , Interferon gama/metabolismo , Interleucina-12/biossíntese , Camundongos Endogâmicos C57BL , Modelos Biológicos , Peritônio/patologia , Peritônio/virologia , Fator 6 Associado a Receptor de TNF/metabolismo , Viroses/virologia
16.
Nat Immunol ; 21(7): 790-801, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32424361

RESUMO

Plasmodium parasite-specific antibodies are critical for protection against malaria, yet the development of long-lived and effective humoral immunity against Plasmodium takes many years and multiple rounds of infection and cure. Here, we report that the rapid development of short-lived plasmablasts during experimental malaria unexpectedly hindered parasite control by impeding germinal center responses. Metabolic hyperactivity of plasmablasts resulted in nutrient deprivation of the germinal center reaction, limiting the generation of memory B cell and long-lived plasma cell responses. Therapeutic administration of a single amino acid to experimentally infected mice was sufficient to overcome the metabolic constraints imposed by plasmablasts and enhanced parasite clearance and the formation of protective humoral immune memory responses. Thus, our studies not only challenge the current model describing the role and function of blood-stage Plasmodium-induced plasmablasts but they also reveal new targets and strategies to improve anti-Plasmodium humoral immunity.


Assuntos
Imunidade Humoral , Malária/imunologia , Plasmócitos/metabolismo , Plasmodium falciparum/imunologia , Adolescente , Adulto , Aminoácidos/administração & dosagem , Aminoácidos/metabolismo , Animais , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/imunologia , Anticorpos Antiprotozoários/metabolismo , Antimaláricos/administração & dosagem , DNA de Protozoário/isolamento & purificação , Modelos Animais de Doenças , Centro Germinativo/citologia , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Interações Hospedeiro-Parasita/imunologia , Humanos , Malária/sangue , Malária/tratamento farmacológico , Malária/parasitologia , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Nutrientes/metabolismo , Plasmócitos/imunologia , Plasmócitos/parasitologia , Plasmodium falciparum/genética , Plasmodium falciparum/isolamento & purificação , Estudo de Prova de Conceito , Adulto Jovem
17.
Cell Rep ; 30(12): 4041-4051.e4, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32209467

RESUMO

During the 2013-2016 Ebola virus (EBOV) epidemic, a significant number of patients admitted to Ebola treatment units were co-infected with Plasmodium falciparum, a predominant agent of malaria. However, there is no consensus on how malaria impacts EBOV infection. The effect of acute Plasmodium infection on EBOV challenge was investigated using mouse-adapted EBOV and a biosafety level 2 (BSL-2) model virus. We demonstrate that acute Plasmodium infection protects from lethal viral challenge, dependent upon interferon gamma (IFN-γ) elicited as a result of parasite infection. Plasmodium-infected mice lacking the IFN-γ receptor are not protected. Ex vivo incubation of naive human or mouse macrophages with sera from acutely parasitemic rodents or macaques programs a proinflammatory phenotype dependent on IFN-γ and renders cells resistant to EBOV infection. We conclude that acute Plasmodium infection can safeguard against EBOV by the production of protective IFN-γ. These findings have implications for anti-malaria therapies administered during episodic EBOV outbreaks in Africa.


Assuntos
Resistência à Doença/imunologia , Ebolavirus/fisiologia , Doença pelo Vírus Ebola/complicações , Doença pelo Vírus Ebola/imunologia , Interferon gama/metabolismo , Malária/complicações , Plasmodium falciparum/fisiologia , Animais , Feminino , Glicoproteínas/metabolismo , Doença pelo Vírus Ebola/prevenção & controle , Macrófagos Peritoneais/patologia , Malária/parasitologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptor de Interferon alfa e beta/metabolismo , Receptores de Interferon/deficiência , Receptores de Interferon/metabolismo , Vesiculovirus/fisiologia , Receptor de Interferon gama
18.
PLoS Negl Trop Dis ; 13(6): e0006983, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31242184

RESUMO

BACKGROUND: T cell immunoglobulin mucin domain-1 (TIM-1) is a phosphatidylserine (PS) receptor, mediating filovirus entry into cells through interactions with PS on virions. TIM-1 expression has been implicated in Ebola virus (EBOV) pathogenesis; however, it remains unclear whether this is due to TIM-1 serving as a filovirus receptor in vivo or, as others have suggested, TIM-1 induces a cytokine storm elicited by T cell/virion interactions. Here, we use a BSL2 model virus that expresses EBOV glycoprotein to demonstrate the importance of TIM-1 as a virus receptor late during in vivo infection. METHODOLOGY/PRINCIPAL FINDINGS: Infectious, GFP-expressing recombinant vesicular stomatitis virus encoding either full length EBOV glycoprotein (EBOV GP/rVSV) or mucin domain deleted EBOV glycoprotein (EBOV GPΔO/rVSV) was used to assess the role of TIM-1 during in vivo infection. GFP-expressing rVSV encoding its native glycoprotein G (G/rVSV) served as a control. TIM-1-sufficient or TIM-1-deficient BALB/c interferon α/ß receptor-/- mice were challenged with these viruses. While G/rVSV caused profound morbidity and mortality in both mouse strains, TIM-1-deficient mice had significantly better survival than TIM-1-expressing mice following EBOV GP/rVSV or EBOV GPΔO/rVSV challenge. EBOV GP/rVSV or EBOV GPΔO/rVSV in spleen of infected animals was high and unaffected by expression of TIM-1. However, infectious virus in serum, liver, kidney and adrenal gland was reduced late in infection in the TIM-1-deficient mice, suggesting that virus entry via this receptor contributes to virus load. Consistent with higher virus loads, proinflammatory chemokines trended higher in organs from infected TIM-1-sufficient mice compared to the TIM-1-deficient mice, but proinflammatory cytokines were more modestly affected. To assess the role of T cells in EBOV GP/rVSV pathogenesis, T cells were depleted in TIM-1-sufficient and -deficient mice and the mice were challenged with virus. Depletion of T cells did not alter the pathogenic consequences of virus infection. CONCLUSIONS: Our studies provide evidence that at late times during EBOV GP/rVSV infection, TIM-1 increased virus load and associated mortality, consistent with an important role of this receptor in virus entry. This work suggests that inhibitors which block TIM-1/virus interaction may serve as effective antivirals, reducing virus load at late times during EBOV infection.


Assuntos
Ebolavirus/fisiologia , Doença pelo Vírus Ebola/virologia , Receptor Celular 1 do Vírus da Hepatite A/metabolismo , Receptores Virais/metabolismo , Internalização do Vírus , Animais , Ebolavirus/genética , Feminino , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Receptor Celular 1 do Vírus da Hepatite A/deficiência , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptores Virais/deficiência , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Genética Reversa , Vesiculovirus/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
19.
Nat Rev Immunol ; 19(7): 457-471, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30940932

RESUMO

Immunity to malaria has been linked to the availability and function of helper CD4+ T cells, cytotoxic CD8+ T cells and γδ T cells that can respond to both the asymptomatic liver stage and the symptomatic blood stage of Plasmodium sp. infection. These T cell responses are also thought to be modulated by regulatory T cells. However, the precise mechanisms governing the development and function of Plasmodium-specific T cells and their capacity to form tissue-resident and long-lived memory populations are less well understood. The field has arrived at a point where the push for vaccines that exploit T cell-mediated immunity to malaria has made it imperative to define and reconcile the mechanisms that regulate the development and functions of Plasmodium-specific T cells. Here, we review our current understanding of the mechanisms by which T cell subsets orchestrate host resistance to Plasmodium infection on the basis of observational and mechanistic studies in humans, non-human primates and rodent models. We also examine the potential of new experimental strategies and human infection systems to inform a new generation of approaches to harness T cell responses against malaria.


Assuntos
Malária/imunologia , Linfócitos T/imunologia , Animais , Citocinas/fisiologia , Humanos , Vacinas Antimaláricas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...