Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 212.471
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Annu Rev Immunol ; 42(1): 259-288, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38277692

RESUMO

Gastrointestinal nematode (GIN) infection has applied significant evolutionary pressure to the mammalian immune system and remains a global economic and human health burden. Upon infection, type 2 immune sentinels activate a common antihelminth response that mobilizes and remodels the intestinal tissue for effector function; however, there is growing appreciation of the impact GIN infection also has on the distal tissue immune state. Indeed, this effect is observed even in tissues through which GINs never transit. This review highlights how GIN infection modulates systemic immunity through (a) induction of host resistance and tolerance responses, (b) secretion of immunomodulatory products, and (c) interaction with the intestinal microbiome. It also discusses the direct consequences that changes to distal tissue immunity can have for concurrent and subsequent infection, chronic noncommunicable diseases, and vaccination efficacy.


Assuntos
Microbioma Gastrointestinal , Nematoides , Infecções por Nematoides , Animais , Humanos , Infecções por Nematoides/imunologia , Nematoides/imunologia , Nematoides/fisiologia , Microbioma Gastrointestinal/imunologia , Imunomodulação , Interações Hospedeiro-Parasita/imunologia , Enteropatias Parasitárias/imunologia , Tolerância Imunológica , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/parasitologia
2.
Annu Rev Immunol ; 33: 201-25, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25533702

RESUMO

Helminth parasites are a highly successful group of pathogens that challenge the immune system in a manner distinct from rapidly replicating infectious agents. Of this group, roundworms (nematodes) that dwell in the intestines of humans and other animals are prevalent worldwide. Currently, more than one billion people are infected by at least one species, often for extended periods of time. Thus, host-protective immunity is rarely complete. The reasons for this are complex, but laboratory investigation of tractable model systems in which protective immunity is effective has provided a mechanistic understanding of resistance that is characterized almost universally by a type 2/T helper 2 response. Greater understanding of the mechanisms of susceptibility has also provided the basis for defining host immunoregulation and parasite-evasion strategies, helping place in context the changing patterns of immunological disease observed worldwide.


Assuntos
Helmintíase/imunologia , Helmintíase/parasitologia , Helmintos/imunologia , Interações Hospedeiro-Patógeno/imunologia , Imunidade Adaptativa , Animais , Antígenos de Helmintos/imunologia , Resistência à Doença , Suscetibilidade a Doenças , Gastroenteropatias/imunologia , Gastroenteropatias/microbiologia , Gastroenteropatias/parasitologia , Microbioma Gastrointestinal , Humanos , Imunidade Inata , Nematoides/imunologia , Infecções por Nematoides/imunologia , Infecções por Nematoides/microbiologia , Infecções por Nematoides/parasitologia
3.
Nat Immunol ; 25(7): 1270-1282, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38877178

RESUMO

The relative and synergistic contributions of genetics and environment to interindividual immune response variation remain unclear, despite implications in evolutionary biology and medicine. Here we quantify interactive effects of genotype and environment on immune traits by investigating C57BL/6, 129S1 and PWK/PhJ inbred mice, rewilded in an outdoor enclosure and infected with the parasite Trichuris muris. Whereas cellular composition was shaped by interactions between genotype and environment, cytokine response heterogeneity including IFNγ concentrations was primarily driven by genotype with consequence on worm burden. In addition, we show that other traits, such as expression of CD44, were explained mostly by genetics on T cells, whereas expression of CD44 on B cells was explained more by environment across all strains. Notably, genetic differences under laboratory conditions were decreased following rewilding. These results indicate that nonheritable influences interact with genetic factors to shape immune variation and parasite burden.


Assuntos
Interação Gene-Ambiente , Camundongos Endogâmicos C57BL , Tricuríase , Trichuris , Animais , Trichuris/imunologia , Tricuríase/imunologia , Tricuríase/parasitologia , Camundongos , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Linfócitos B/imunologia , Genótipo , Interferon gama/metabolismo , Linfócitos T/imunologia , Feminino , Masculino
4.
Annu Rev Immunol ; 32: 157-87, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24655294

RESUMO

Malaria is a mosquito-borne disease caused by parasites of the obligate intracellular Apicomplexa phylum the most deadly of which, Plasmodium falciparum, prevails in Africa. Malaria imposes a huge health burden on the world's most vulnerable populations, claiming the lives of nearly one million children and pregnant women each year. Although there is keen interest in eradicating malaria, we do not yet have the necessary tools to meet this challenge, including an effective malaria vaccine and adequate vector control strategies. Here we review what is known about the mechanisms at play in immune resistance to malaria in both the human and mosquito hosts at each step in the parasite's complex life cycle with a view toward developing the tools that will contribute to the prevention of disease and death and, ultimately, to the goal of malaria eradication. In so doing, we hope to inspire immunologists to participate in defeating this devastating disease.


Assuntos
Culicidae/imunologia , Interações Hospedeiro-Patógeno/imunologia , Malária/imunologia , Plasmodium/imunologia , Animais , Culicidae/parasitologia , Humanos , Estágios do Ciclo de Vida , Malária/parasitologia , Malária/prevenção & controle , Plasmodium/crescimento & desenvolvimento , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/imunologia
5.
Cell ; 184(5): 1214-1231.e16, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33636133

RESUMO

Although enteric helminth infections modulate immunity to mucosal pathogens, their effects on systemic microbes remain less established. Here, we observe increased mortality in mice coinfected with the enteric helminth Heligmosomoides polygyrus bakeri (Hpb) and West Nile virus (WNV). This enhanced susceptibility is associated with altered gut morphology and transit, translocation of commensal bacteria, impaired WNV-specific T cell responses, and increased virus infection in the gastrointestinal tract and central nervous system. These outcomes were due to type 2 immune skewing, because coinfection in Stat6-/- mice rescues mortality, treatment of helminth-free WNV-infected mice with interleukin (IL)-4 mirrors coinfection, and IL-4 receptor signaling in intestinal epithelial cells mediates the susceptibility phenotypes. Moreover, tuft cell-deficient mice show improved outcomes with coinfection, whereas treatment of helminth-free mice with tuft cell-derived cytokine IL-25 or ligand succinate worsens WNV disease. Thus, helminth activation of tuft cell-IL-4-receptor circuits in the gut exacerbates infection and disease of a neurotropic flavivirus.


Assuntos
Coinfecção , Nematospiroides dubius/fisiologia , Transdução de Sinais , Infecções por Strongylida/patologia , Vírus do Nilo Ocidental/fisiologia , Animais , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Mucosa Intestinal/parasitologia , Mucosa Intestinal/virologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/parasitologia , Neurônios/virologia , Receptores de Interleucina-4/metabolismo , Fator de Transcrição STAT6/genética , Índice de Gravidade de Doença , Infecções por Strongylida/parasitologia
6.
Cell ; 184(23): 5715-5727.e12, 2021 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-34717799

RESUMO

The enteric nervous system (ENS) controls several intestinal functions including motility and nutrient handling, which can be disrupted by infection-induced neuropathies or neuronal cell death. We investigated possible tolerance mechanisms preventing neuronal loss and disruption in gut motility after pathogen exposure. We found that following enteric infections, muscularis macrophages (MMs) acquire a tissue-protective phenotype that prevents neuronal loss, dysmotility, and maintains energy balance during subsequent challenge with unrelated pathogens. Bacteria-induced neuroprotection relied on activation of gut-projecting sympathetic neurons and signaling via ß2-adrenergic receptors (ß2AR) on MMs. In contrast, helminth-mediated neuroprotection was dependent on T cells and systemic production of interleukin (IL)-4 and IL-13 by eosinophils, which induced arginase-expressing MMs that prevented neuronal loss from an unrelated infection located in a different intestinal region. Collectively, these data suggest that distinct enteric pathogens trigger a state of disease or tissue tolerance that preserves ENS number and functionality.


Assuntos
Sistema Nervoso Entérico/microbiologia , Sistema Nervoso Entérico/parasitologia , Infecções/microbiologia , Infecções/parasitologia , Neurônios/patologia , Neuroproteção , Especificidade de Órgãos , Yersinia pseudotuberculosis/fisiologia , Animais , Eosinófilos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Imunidade , Infecções/imunologia , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Macrófagos/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Strongyloides/fisiologia , Estrongiloidíase/genética , Estrongiloidíase/imunologia , Estrongiloidíase/parasitologia , Transcriptoma/genética , Infecções por Yersinia pseudotuberculosis/genética , Infecções por Yersinia pseudotuberculosis/imunologia , Infecções por Yersinia pseudotuberculosis/microbiologia
7.
Cell ; 184(20): 5201-5214.e12, 2021 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-34536345

RESUMO

Certain obligate parasites induce complex and substantial phenotypic changes in their hosts in ways that favor their transmission to other trophic levels. However, the mechanisms underlying these changes remain largely unknown. Here we demonstrate how SAP05 protein effectors from insect-vectored plant pathogenic phytoplasmas take control of several plant developmental processes. These effectors simultaneously prolong the host lifespan and induce witches' broom-like proliferations of leaf and sterile shoots, organs colonized by phytoplasmas and vectors. SAP05 acts by mediating the concurrent degradation of SPL and GATA developmental regulators via a process that relies on hijacking the plant ubiquitin receptor RPN10 independent of substrate ubiquitination. RPN10 is highly conserved among eukaryotes, but SAP05 does not bind insect vector RPN10. A two-amino-acid substitution within plant RPN10 generates a functional variant that is resistant to SAP05 activities. Therefore, one effector protein enables obligate parasitic phytoplasmas to induce a plethora of developmental phenotypes in their hosts.


Assuntos
Arabidopsis/crescimento & desenvolvimento , Arabidopsis/parasitologia , Interações Hospedeiro-Parasita/fisiologia , Parasitos/fisiologia , Proteólise , Ubiquitinas/metabolismo , Sequência de Aminoácidos , Animais , Arabidopsis/genética , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/metabolismo , Engenharia Genética , Humanos , Insetos/fisiologia , Modelos Biológicos , Fenótipo , Fotoperíodo , Filogenia , Phytoplasma/fisiologia , Desenvolvimento Vegetal , Brotos de Planta/crescimento & desenvolvimento , Plantas Geneticamente Modificadas , Complexo de Endopeptidases do Proteassoma/metabolismo , Estabilidade Proteica , Reprodução , Nicotiana , Fatores de Transcrição/metabolismo , Transcrição Gênica
8.
Cell ; 183(2): 554-554.e1, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-33064992

RESUMO

Malaria is a prominent vector-borne illness caused by Plasmodium parasites. Therapeutic intervention remains a critical component for disease eradication efforts but is complicated by the emergence of drug resistance. This SnapShot summarizes the human-relevant stages of the P. falciparum life cycle and describes how licensed antimalarials, clinical candidates, and newly emerging compounds target each stage to prevent, treat, or block transmission of malaria. To view this SnapShot, open or download the PDF.


Assuntos
Antimaláricos/farmacologia , Antimaláricos/uso terapêutico , Malária/tratamento farmacológico , Erradicação de Doenças , Resistência a Medicamentos , Humanos , Malária/parasitologia , Malária Falciparum/parasitologia , Plasmodium/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos
9.
Cell ; 183(1): 258-268.e12, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32860739

RESUMO

Plasmodium species, the causative agent of malaria, rely on glucose for energy supply during blood stage. Inhibition of glucose uptake thus represents a potential strategy for the development of antimalarial drugs. Here, we present the crystal structures of PfHT1, the sole hexose transporter in the genome of Plasmodium species, at resolutions of 2.6 Å in complex with D-glucose and 3.7 Å with a moderately selective inhibitor, C3361. Although both structures exhibit occluded conformations, binding of C3361 induces marked rearrangements that result in an additional pocket. This inhibitor-binding-induced pocket presents an opportunity for the rational design of PfHT1-specific inhibitors. Among our designed C3361 derivatives, several exhibited improved inhibition of PfHT1 and cellular potency against P. falciparum, with excellent selectivity to human GLUT1. These findings serve as a proof of concept for the development of the next-generation antimalarial chemotherapeutics by simultaneously targeting the orthosteric and allosteric sites of PfHT1.


Assuntos
Proteínas de Transporte de Monossacarídeos/ultraestrutura , Plasmodium falciparum/metabolismo , Plasmodium falciparum/ultraestrutura , Proteínas de Protozoários/ultraestrutura , Sequência de Aminoácidos , Animais , Antimaláricos , Transporte Biológico , Glucose/metabolismo , Humanos , Malária , Malária Falciparum/parasitologia , Proteínas de Transporte de Monossacarídeos/química , Proteínas de Transporte de Monossacarídeos/metabolismo , Parasitos , Plasmodium falciparum/genética , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo , Açúcares/metabolismo
11.
Cell ; 177(2): 315-325.e14, 2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30929905

RESUMO

Transmission of malaria parasites occurs when a female Anopheles mosquito feeds on an infected host to acquire nutrients for egg development. How parasites are affected by oogenetic processes, principally orchestrated by the steroid hormone 20-hydroxyecdysone (20E), remains largely unknown. Here we show that Plasmodium falciparum development is intimately but not competitively linked to processes shaping Anopheles gambiae reproduction. We unveil a 20E-mediated positive correlation between egg and oocyst numbers; impairing oogenesis by multiple 20E manipulations decreases parasite intensities. These manipulations, however, accelerate Plasmodium growth rates, allowing sporozoites to become infectious sooner. Parasites exploit mosquito lipids for faster growth, but they do so without further affecting egg development. These results suggest that P. falciparum has adopted a non-competitive evolutionary strategy of resource exploitation to optimize transmission while minimizing fitness costs to its mosquito vector. Our findings have profound implications for currently proposed control strategies aimed at suppressing mosquito populations.


Assuntos
Ecdisterona/metabolismo , Interações Hospedeiro-Parasita/fisiologia , Malária Falciparum/parasitologia , Animais , Anopheles/parasitologia , Culicidae , Ecdisterona/fisiologia , Feminino , Células HEK293 , Humanos , Insetos Vetores , Malária/parasitologia , Camundongos , Mosquitos Vetores , Células NIH 3T3 , Oogênese/fisiologia , Plasmodium/metabolismo , Plasmodium falciparum , Esporozoítos , Esteroides/metabolismo
12.
Cell ; 178(1): 216-228.e21, 2019 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-31204103

RESUMO

The Plasmodium falciparum reticulocyte-binding protein homolog 5 (PfRH5) is the leading target for next-generation vaccines against the disease-causing blood-stage of malaria. However, little is known about how human antibodies confer functional immunity against this antigen. We isolated a panel of human monoclonal antibodies (mAbs) against PfRH5 from peripheral blood B cells from vaccinees in the first clinical trial of a PfRH5-based vaccine. We identified a subset of mAbs with neutralizing activity that bind to three distinct sites and another subset of mAbs that are non-functional, or even antagonistic to neutralizing antibodies. We also identify the epitope of a novel group of non-neutralizing antibodies that significantly reduce the speed of red blood cell invasion by the merozoite, thereby potentiating the effect of all neutralizing PfRH5 antibodies as well as synergizing with antibodies targeting other malaria invasion proteins. Our results provide a roadmap for structure-guided vaccine development to maximize antibody efficacy against blood-stage malaria.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antiprotozoários/imunologia , Eritrócitos/parasitologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Plasmodium falciparum/imunologia , Adolescente , Adulto , Animais , Sítios de Ligação , Proteínas de Transporte/imunologia , Reações Cruzadas/imunologia , Epitopos/imunologia , Feminino , Células HEK293 , Voluntários Saudáveis , Humanos , Malária Falciparum/parasitologia , Masculino , Merozoítos/fisiologia , Pessoa de Meia-Idade , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/imunologia , Coelhos , Ratos , Ratos Sprague-Dawley , Adulto Jovem
13.
Cell ; 179(5): 1112-1128.e26, 2019 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-31730853

RESUMO

Plasmodium gene functions in mosquito and liver stages remain poorly characterized due to limitations in the throughput of phenotyping at these stages. To fill this gap, we followed more than 1,300 barcoded P. berghei mutants through the life cycle. We discover 461 genes required for efficient parasite transmission to mosquitoes through the liver stage and back into the bloodstream of mice. We analyze the screen in the context of genomic, transcriptomic, and metabolomic data by building a thermodynamic model of P. berghei liver-stage metabolism, which shows a major reprogramming of parasite metabolism to achieve rapid growth in the liver. We identify seven metabolic subsystems that become essential at the liver stages compared with asexual blood stages: type II fatty acid synthesis and elongation (FAE), tricarboxylic acid, amino sugar, heme, lipoate, and shikimate metabolism. Selected predictions from the model are individually validated in single mutants to provide future targets for drug development.


Assuntos
Genoma de Protozoário , Estágios do Ciclo de Vida/genética , Fígado/metabolismo , Fígado/parasitologia , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/genética , Alelos , Amino Açúcares/biossíntese , Animais , Culicidae/parasitologia , Eritrócitos/parasitologia , Ácido Graxo Sintases/metabolismo , Ácidos Graxos/metabolismo , Técnicas de Inativação de Genes , Genótipo , Modelos Biológicos , Mutação/genética , Parasitos/genética , Parasitos/crescimento & desenvolvimento , Fenótipo , Plasmodium berghei/metabolismo , Ploidias , Reprodução
14.
Cell ; 176(1-2): 306-317.e16, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30503212

RESUMO

Trypanosome parasites control their virulence and spread by using quorum sensing (QS) to generate transmissible "stumpy forms" in their host bloodstream. However, the QS signal "stumpy induction factor" (SIF) and its reception mechanism are unknown. Although trypanosomes lack G protein-coupled receptor signaling, we have identified a surface GPR89-family protein that regulates stumpy formation. TbGPR89 is expressed on bloodstream "slender form" trypanosomes, which receive the SIF signal, and when ectopically expressed, TbGPR89 drives stumpy formation in a SIF-pathway-dependent process. Structural modeling of TbGPR89 predicts unexpected similarity to oligopeptide transporters (POT), and when expressed in bacteria, TbGPR89 transports oligopeptides. Conversely, expression of an E. coli POT in trypanosomes drives parasite differentiation, and oligopeptides promote stumpy formation in vitro. Furthermore, the expression of secreted trypanosome oligopeptidases generates a paracrine signal that accelerates stumpy formation in vivo. Peptidase-generated oligopeptide QS signals being received through TbGPR89 provides a mechanism for both trypanosome SIF production and reception.


Assuntos
Proteínas de Membrana Transportadoras/fisiologia , Percepção de Quorum/fisiologia , Trypanosoma/metabolismo , Diferenciação Celular , Sequência Conservada/genética , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Membrana Transportadoras/genética , Oligopeptídeos/genética , Oligopeptídeos/fisiologia , Filogenia , Proteínas de Protozoários/metabolismo , Percepção de Quorum/genética , Transdução de Sinais , Trypanosoma/fisiologia , Trypanosoma brucei brucei/metabolismo , Tripanossomíase Africana/parasitologia , Virulência/fisiologia
15.
Nat Immunol ; 22(3): 347-357, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33432229

RESUMO

Activated Vγ9Vδ2 (γδ2) T lymphocytes that sense parasite-produced phosphoantigens are expanded in Plasmodium falciparum-infected patients. Although previous studies suggested that γδ2 T cells help control erythrocytic malaria, whether γδ2 T cells recognize infected red blood cells (iRBCs) was uncertain. Here we show that iRBCs stained for the phosphoantigen sensor butyrophilin 3A1 (BTN3A1). γδ2 T cells formed immune synapses and lysed iRBCs in a contact, phosphoantigen, BTN3A1 and degranulation-dependent manner, killing intracellular parasites. Granulysin released into the synapse lysed iRBCs and delivered death-inducing granzymes to the parasite. All intra-erythrocytic parasites were susceptible, but schizonts were most sensitive. A second protective γδ2 T cell mechanism was identified. In the presence of patient serum, γδ2 T cells phagocytosed and degraded opsonized iRBCs in a CD16-dependent manner, decreasing parasite multiplication. Thus, γδ2 T cells have two ways to control blood-stage malaria-γδ T cell antigen receptor (TCR)-mediated degranulation and phagocytosis of antibody-coated iRBCs.


Assuntos
Antígenos de Protozoários/imunologia , Citotoxicidade Imunológica , Eritrócitos/imunologia , Linfócitos Intraepiteliais/imunologia , Ativação Linfocitária , Malária Falciparum/imunologia , Fagocitose , Plasmodium falciparum/microbiologia , Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos T/metabolismo , Antígenos de Protozoários/sangue , Boston , Brasil , Butirofilinas/metabolismo , Células Cultivadas , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Feminino , Granzimas/metabolismo , Interações Hospedeiro-Parasita , Humanos , Sinapses Imunológicas/metabolismo , Sinapses Imunológicas/parasitologia , Linfócitos Intraepiteliais/metabolismo , Linfócitos Intraepiteliais/parasitologia , Malária Falciparum/sangue , Malária Falciparum/parasitologia , Masculino , Plasmodium falciparum/crescimento & desenvolvimento
16.
Nat Immunol ; 22(5): 654-665, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33888898

RESUMO

Controlled human infections provide opportunities to study the interaction between the immune system and malaria parasites, which is essential for vaccine development. Here, we compared immune signatures of malaria-naive Europeans and of Africans with lifelong malaria exposure using mass cytometry, RNA sequencing and data integration, before and 5 and 11 days after venous inoculation with Plasmodium falciparum sporozoites. We observed differences in immune cell populations, antigen-specific responses and gene expression profiles between Europeans and Africans and among Africans with differing degrees of immunity. Before inoculation, an activated/differentiated state of both innate and adaptive cells, including elevated CD161+CD4+ T cells and interferon-γ production, predicted Africans capable of controlling parasitemia. After inoculation, the rapidity of the transcriptional response and clusters of CD4+ T cells, plasmacytoid dendritic cells and innate T cells were among the features distinguishing Africans capable of controlling parasitemia from susceptible individuals. These findings can guide the development of a vaccine effective in malaria-endemic regions.


Assuntos
Imunidade Adaptativa/imunologia , Suscetibilidade a Doenças/imunologia , Malária Falciparum/imunologia , Plasmodium falciparum/imunologia , Imunidade Adaptativa/genética , Adolescente , Adulto , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , População Negra/genética , Células Dendríticas/imunologia , Suscetibilidade a Doenças/sangue , Suscetibilidade a Doenças/parasitologia , Feminino , Voluntários Saudáveis , Interações Hospedeiro-Parasita/genética , Interações Hospedeiro-Parasita/imunologia , Humanos , Imunidade Inata/genética , Imunidade Inata/imunologia , Interferon gama/metabolismo , Malária Falciparum/sangue , Malária Falciparum/parasitologia , Masculino , RNA-Seq , Análise de Sistemas , Linfócitos T/imunologia , Linfócitos T/metabolismo , População Branca/genética , Adulto Jovem
17.
Nat Immunol ; 22(2): 216-228, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33462454

RESUMO

CD4+ effector lymphocytes (Teff) are traditionally classified by the cytokines they produce. To determine the states that Teff cells actually adopt in frontline tissues in vivo, we applied single-cell transcriptome and chromatin analyses to colonic Teff cells in germ-free or conventional mice or in mice after challenge with a range of phenotypically biasing microbes. Unexpected subsets were marked by the expression of the interferon (IFN) signature or myeloid-specific transcripts, but transcriptome or chromatin structure could not resolve discrete clusters fitting classic helper T cell (TH) subsets. At baseline or at different times of infection, transcripts encoding cytokines or proteins commonly used as TH markers were distributed in a polarized continuum, which was functionally validated. Clones derived from single progenitors gave rise to both IFN-γ- and interleukin (IL)-17-producing cells. Most of the transcriptional variance was tied to the infecting agent, independent of the cytokines produced, and chromatin variance primarily reflected activities of activator protein (AP)-1 and IFN-regulatory factor (IRF) transcription factor (TF) families, not the canonical subset master regulators T-bet, GATA3 or RORγ.


Assuntos
Bactérias/patogenicidade , Infecções Bacterianas/microbiologia , Linfócitos T CD4-Positivos/microbiologia , Linfócitos T CD4-Positivos/parasitologia , Colo/microbiologia , Colo/parasitologia , Microbioma Gastrointestinal , Heligmosomatoidea/patogenicidade , Enteropatias Parasitárias/parasitologia , Animais , Bactérias/imunologia , Infecções Bacterianas/genética , Infecções Bacterianas/imunologia , Infecções Bacterianas/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Cromatina/genética , Cromatina/metabolismo , Citrobacter rodentium/imunologia , Citrobacter rodentium/patogenicidade , Colo/imunologia , Colo/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Heligmosomatoidea/imunologia , Interações Hospedeiro-Patógeno , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Enteropatias Parasitárias/genética , Enteropatias Parasitárias/imunologia , Enteropatias Parasitárias/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Nematospiroides dubius/imunologia , Nematospiroides dubius/patogenicidade , Nippostrongylus/imunologia , Nippostrongylus/patogenicidade , Fenótipo , Salmonella enterica/imunologia , Salmonella enterica/patogenicidade , Análise de Célula Única , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Transcriptoma
18.
Immunity ; 57(6): 1215-1224.e6, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38788711

RESUMO

Malaria is a life-threatening disease of global health importance, particularly in sub-Saharan Africa. The growth inhibition assay (GIA) is routinely used to evaluate, prioritize, and quantify the efficacy of malaria blood-stage vaccine candidates but does not reliably predict either naturally acquired or vaccine-induced protection. Controlled human malaria challenge studies in semi-immune volunteers provide an unparalleled opportunity to robustly identify mechanistic correlates of protection. We leveraged this platform to undertake a head-to-head comparison of seven functional antibody assays that are relevant to immunity against the erythrocytic merozoite stage of Plasmodium falciparum. Fc-mediated effector functions were strongly associated with protection from clinical symptoms of malaria and exponential parasite multiplication, while the gold standard GIA was not. The breadth of Fc-mediated effector function discriminated clinical immunity following the challenge. These findings present a shift in the understanding of the mechanisms that underpin immunity to malaria and have important implications for vaccine development.


Assuntos
Anticorpos Antiprotozoários , Vacinas Antimaláricas , Malária Falciparum , Plasmodium falciparum , Humanos , Plasmodium falciparum/imunologia , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Anticorpos Antiprotozoários/imunologia , Vacinas Antimaláricas/imunologia , Adulto , Fragmentos Fc das Imunoglobulinas/imunologia , Merozoítos/imunologia , Eritrócitos/parasitologia , Eritrócitos/imunologia , Feminino , Masculino , Adulto Jovem
19.
Immunity ; 57(5): 935-937, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38749395

RESUMO

The intestinal epithelium interacts with immune cells to support tissue homeostasis and coordinate responses against pathogens. In this issue of Immunity, Yang et al. unveil a central role for mast cell-epithelial cell interactions in orchestrating protective type 2 immune responses following intestinal helminth infection.


Assuntos
Mucosa Intestinal , Mastócitos , Mastócitos/imunologia , Animais , Mucosa Intestinal/imunologia , Mucosa Intestinal/parasitologia , Humanos , Homeostase/imunologia , Helmintíase/imunologia , Helmintíase/parasitologia , Células Epiteliais/imunologia , Camundongos
20.
Immunity ; 57(6): 1243-1259.e8, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38744291

RESUMO

Epithelial cells secrete chloride to regulate water release at mucosal barriers, supporting both homeostatic hydration and the "weep" response that is critical for type 2 immune defense against parasitic worms (helminths). Epithelial tuft cells in the small intestine sense helminths and release cytokines and lipids to activate type 2 immune cells, but whether they regulate epithelial secretion is unknown. Here, we found that tuft cell activation rapidly induced epithelial chloride secretion in the small intestine. This response required tuft cell sensory functions and tuft cell-derived acetylcholine (ACh), which acted directly on neighboring epithelial cells to stimulate chloride secretion, independent of neurons. Maximal tuft cell-induced chloride secretion coincided with immune restriction of helminths, and clearance was delayed in mice lacking tuft cell-derived ACh, despite normal type 2 inflammation. Thus, we have uncovered an epithelium-intrinsic response unit that uses ACh to couple tuft cell sensing to the secretory defenses of neighboring epithelial cells.


Assuntos
Acetilcolina , Cloretos , Células Epiteliais , Mucosa Intestinal , Animais , Acetilcolina/metabolismo , Camundongos , Cloretos/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/parasitologia , Células Epiteliais/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/parasitologia , Intestino Delgado/imunologia , Intestino Delgado/parasitologia , Intestino Delgado/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células em Tufo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA