Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 12811, 2024 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-38834738

RESUMO

Macrophages provide a crucial environment for Salmonella enterica serovar Typhi (S. Typhi) to multiply during typhoid fever, yet our understanding of how human macrophages and S. Typhi interact remains limited. In this study, we delve into the dynamics of S. Typhi replication within human macrophages and the resulting heterogeneous transcriptomic responses of macrophages during infection. Our study reveals key factors that influence macrophage diversity, uncovering distinct immune and metabolic pathways associated with different stages of S. Typhi intracellular replication in macrophages. Of note, we found that macrophages harboring replicating S. Typhi are skewed towards an M1 pro-inflammatory state, whereas macrophages containing non-replicating S. Typhi exhibit neither a distinct M1 pro-inflammatory nor M2 anti-inflammatory state. Additionally, macrophages with replicating S. Typhi were characterized by the increased expression of genes associated with STAT3 phosphorylation and the activation of the STAT3 transcription factor. Our results shed light on transcriptomic pathways involved in the susceptibility of human macrophages to intracellular S. Typhi replication, thereby providing crucial insight into host phenotypes that restrict and support S. Typhi infection.


Assuntos
Macrófagos , Fator de Transcrição STAT3 , Salmonella typhi , Febre Tifoide , Humanos , Macrófagos/metabolismo , Macrófagos/microbiologia , Salmonella typhi/genética , Febre Tifoide/microbiologia , Febre Tifoide/imunologia , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT3/genética , Perfilação da Expressão Gênica , Fenótipo , Transcriptoma , Fosforilação
2.
Sci Adv ; 9(1): eadd4333, 2023 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-36608122

RESUMO

Macrophages mediate key antimicrobial responses against intracellular bacterial pathogens, such as Salmonella enterica. Yet, they can also act as a permissive niche for these pathogens to persist in infected tissues within granulomas, which are immunological structures composed of macrophages and other immune cells. We apply single-cell transcriptomics to investigate macrophage functional diversity during persistent S. enterica serovar Typhimurium (STm) infection in mice. We identify determinants of macrophage heterogeneity in infected spleens and describe populations of distinct phenotypes, functional programming, and spatial localization. Using an STm mutant with impaired ability to polarize macrophage phenotypes, we find that angiotensin-converting enzyme (ACE) defines a granuloma macrophage population that is nonpermissive for intracellular bacteria, and their abundance anticorrelates with tissue bacterial burden. Disruption of pathogen control by neutralizing TNF is linked to preferential depletion of ACE+ macrophages in infected tissues. Thus, ACE+ macrophages have limited capacity to serve as cellular niche for intracellular bacteria to establish persistent infection.


Assuntos
Infecções por Salmonella , Salmonella typhimurium , Animais , Camundongos , Salmonella typhimurium/genética , Infecção Persistente , Infecções por Salmonella/genética , Macrófagos/microbiologia , Granuloma
3.
Cell Host Microbe ; 27(1): 54-67.e5, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31883922

RESUMO

Many intracellular bacteria can establish chronic infection and persist in tissues within granulomas composed of macrophages. Granuloma macrophages exhibit heterogeneous polarization states, or phenotypes, that may be functionally distinct. Here, we elucidate a host-pathogen interaction that controls granuloma macrophage polarization and long-term pathogen persistence during Salmonella Typhimurium (STm) infection. We show that STm persists within splenic granulomas that are densely populated by CD11b+CD11c+Ly6C+ macrophages. STm preferentially persists in granuloma macrophages reprogrammed to an M2 state, in part through the activity of the effector SteE, which contributes to the establishment of persistent infection. We demonstrate that tumor necrosis factor (TNF) signaling limits M2 granuloma macrophage polarization, thereby restricting STm persistence. TNF neutralization shifts granuloma macrophages toward an M2 state and increases bacterial persistence, and these effects are partially dependent on SteE activity. Thus, manipulating granuloma macrophage polarization represents a strategy for intracellular bacteria to overcome host restriction during persistent infection.


Assuntos
Granuloma/imunologia , Interações Hospedeiro-Patógeno/imunologia , Ativação de Macrófagos/imunologia , Infecções por Salmonella/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Granuloma/microbiologia , Humanos , Interleucina-4/metabolismo , Macrófagos/microbiologia , Camundongos , Salmonella typhimurium/imunologia , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidade , Baço/citologia , Baço/microbiologia , Baço/patologia , Transativadores/metabolismo , Fatores de Virulência/metabolismo
4.
Cell Host Microbe ; 27(1): 41-53.e6, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31862381

RESUMO

Many Gram-negative bacterial pathogens antagonize anti-bacterial immunity through translocated effector proteins that inhibit pro-inflammatory signaling. In addition, the intracellular pathogen Salmonella enterica serovar Typhimurium initiates an anti-inflammatory transcriptional response in macrophages through its effector protein SteE. However, the target(s) and molecular mechanism of SteE remain unknown. Here, we demonstrate that SteE converts both the amino acid and substrate specificity of the host pleiotropic serine/threonine kinase GSK3. SteE itself is a substrate of GSK3, and phosphorylation of SteE is required for its activity. Remarkably, phosphorylated SteE then forces GSK3 to phosphorylate the non-canonical substrate signal transducer and activator of transcription 3 (STAT3) on tyrosine-705. This results in STAT3 activation, which along with GSK3 is required for SteE-mediated upregulation of the anti-inflammatory M2 macrophage marker interleukin-4Rα (IL-4Rα). Overall, the conversion of GSK3 to a tyrosine-directed kinase represents a tightly regulated event that enables a bacterial virulence protein to reprogram innate immune signaling and establish an anti-inflammatory environment.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Macrófagos/microbiologia , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Transcrição STAT3/metabolismo , Salmonella typhimurium , Animais , Proteínas de Bactérias/metabolismo , Células HEK293 , Células HeLa , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Interleucina-4/metabolismo , Ativação de Macrófagos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Tirosina Quinases/metabolismo , Salmonella typhimurium/imunologia , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidade , Virulência/imunologia
5.
Pediatrics ; 141(3)2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29437933

RESUMO

A term, appropriate-for-gestational-age, male infant born via normal spontaneous vaginal delivery presented at birth with a full-body erythematous, vesiculobullous rash. He was well-appearing with normal vital signs and hypoglycemia that quickly resolved. His father had a history of herpes labialis. His mother had an episode of herpes zoster during pregnancy and a prolonged rupture of membranes that was adequately treated. The patient underwent a sepsis workup, including 2 attempted but unsuccessful lumbar punctures, and was started on broad-spectrum antibiotics and acyclovir, given concerns about bacterial or viral infection. The rash evolved over the course of several days. Subsequent workup, with particular attention to his history and presentation, led to his diagnosis.


Assuntos
Exantema/etiologia , Mastocitose Cutânea/diagnóstico , Dermatopatias Vesiculobolhosas/etiologia , Diagnóstico Diferencial , Quimioterapia Combinada , Humanos , Recém-Nascido , Masculino , Mastocitose Cutânea/tratamento farmacológico , Dermatopatias Infecciosas/diagnóstico
6.
Eur J Immunol ; 41(12): 3423-35, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21969276

RESUMO

Deficiency in the guanine nucleotide exchange factor dedicator of cytokinesis 8 (DOCK8) causes a human immunodeficiency syndrome associated with recurrent sinopulmonary and viral infections. We have recently identified a DOCK8-deficient mouse strain, carrying an ethylnitrosourea-induced splice-site mutation that shows a failure to mature a humoral immune response due to the loss of germinal centre B cells. In this study, we turned to T-cell immunity to investigate further the human immunodeficiency syndrome and its association with decreased peripheral CD4(+) and CD8(+) T cells. Characterisation of the DOCK8-deficient mouse revealed T-cell lymphopenia, with increased T-cell turnover and decreased survival. Egress of mature CD4(+) thymocytes was reduced with increased migration of these cells to the chemokine CXCL12. However, despite the two-fold reduction in peripheral naïve T cells, the DOCK8-deficient mice generated a normal primary CD8(+) immune response and were able to survive acute influenza virus infection. The limiting effect of DOCK8 was in the normal survival of CD8(+) memory T cells after infection. These findings help to explain why DOCK8-deficient patients are susceptible to recurrent infections and provide new insights into how T-cell memory is sustained.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Fatores de Troca do Nucleotídeo Guanina/deficiência , Fatores de Troca do Nucleotídeo Guanina/imunologia , Memória Imunológica/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Movimento Celular/imunologia , Sobrevivência Celular/imunologia , Células Cultivadas , Quimiocina CXCL12/imunologia , Humanos , Síndromes de Imunodeficiência/imunologia , Ativação Linfocitária/imunologia , Linfoma de Células T/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA