Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
Cell Rep Med ; 3(8): 100713, 2022 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-35932762

RESUMO

Maternal asthma status, prenatal exposures, and infant gut microbiota perturbation are associated with heightened risk of atopy and asthma risk in childhood, observations hypothetically linked by intergenerational microbial transmission. Using maternal vaginal (n = 184) and paired infant stool (n = 172) samples, we identify four compositionally and functionally distinct Lactobacillus-dominated vaginal microbiota clusters (VCs) that relate to prenatal maternal health and exposures and infant serum immunoglobulin E (IgE) status at 1 year. Variance in bacteria shared between mother and infant pairs relate to VCs, maternal allergy/asthma status, and infant IgE levels. Heritable bacterial gene pathways associated with infant IgE include fatty acid synthesis and histamine and tryptophan degradation. In vitro, vertically transmitted Lactobacillus jensenii strains induce immunosuppressive phenotypes on human antigen-presenting cells. Murine supplementation with L. jensenii reduces lung eosinophils, neutrophilic expansion, and the proportion of interleukin-4 (IL-4)+ CD4+ T cells. Thus, bacterial and atopy heritability are intimately linked, suggesting a microbial component of intergenerational disease transmission.


Assuntos
Asma , Microbioma Gastrointestinal , Hipersensibilidade Imediata , Animais , Asma/genética , Bactérias/genética , Feminino , Microbioma Gastrointestinal/genética , Humanos , Tolerância Imunológica/genética , Imunoglobulina E , Lactente , Camundongos , Gravidez
2.
Cell Rep ; 34(1): 108573, 2021 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-33406429

RESUMO

Whereas the human fetal immune system is poised to generate immune tolerance and suppress inflammation in utero, an adult-like immune system emerges to orchestrate anti-pathogen immune responses in post-natal life. It has been posited that cells of the adult immune system arise as a discrete ontological "layer" of hematopoietic stem-progenitor cells (HSPCs) and their progeny; evidence supporting this model in humans has, however, been inconclusive. Here, we combine bulk and single-cell transcriptional profiling of lymphoid cells, myeloid cells, and HSPCs from fetal, perinatal, and adult developmental stages to demonstrate that the fetal-to-adult transition occurs progressively along a continuum of maturity-with a substantial degree of inter-individual variation at the time of birth-rather than via a transition between discrete waves. These findings have important implications for the design of strategies for prophylaxis against infection in the newborn and for the use of umbilical cord blood (UCB) in the setting of transplantation.


Assuntos
Feto/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Linfócitos/metabolismo , Células Mieloides/metabolismo , Análise de Célula Única , Linfócitos T/metabolismo , Transcriptoma , Medula Óssea/metabolismo , Técnicas de Cultura de Células , Feminino , Sangue Fetal , Humanos , Imunidade , Gravidez , Análise de Sequência de RNA
3.
Sci Immunol ; 4(41)2019 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-31757834

RESUMO

T cell receptor (TCR) stimulation and cytokine cues drive the differentiation of CD4+ naïve T cells into effector T cell populations with distinct proinflammatory or regulatory functions. Unlike adult naïve T cells, human fetal naïve CD4+ T cells preferentially differentiate into FOXP3+ regulatory T (Treg) cells upon TCR activation independent of exogenous cytokine signaling. This cell-intrinsic predisposition for Treg differentiation is implicated in the generation of tolerance in utero; however, the underlying mechanisms remain largely unknown. Here, we identify epigenetic and transcriptional programs shared between fetal naïve T and committed Treg cells that are inactive in adult naïve T cells and show that fetal-derived induced Treg (iTreg) cells retain this transcriptional program. We show that a subset of Treg-specific enhancers is accessible in fetal naïve T cells, including two active superenhancers at Helios Helios is expressed in fetal naïve T cells but not in adult naïve T cells, and fetal iTreg cells maintain Helios expression. CRISPR-Cas9 ablation of Helios in fetal naïve T cells impaired their differentiation into iTreg cells upon TCR stimulation, reduced expression of immunosuppressive genes in fetal iTreg cells such as IL10, and increased expression of proinflammatory genes including IFNG Consequently, Helios knockout fetal iTreg cells had reduced IL-10 and increased IFN-γ cytokine production. Together, our results reveal important roles for Helios in enhancing preferential fetal Treg differentiation and fine-tuning eventual Treg function. The Treg-biased programs identified within fetal naïve T cells could potentially be used to engineer enhanced iTreg populations for adoptive cellular therapies.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Fator de Transcrição Ikaros/imunologia , Linfócitos T Reguladores/imunologia , Adulto , Linfócitos T CD4-Positivos/citologia , Diferenciação Celular/imunologia , Células Cultivadas , Humanos , Linfócitos T Reguladores/citologia
4.
J Clin Invest ; 129(9): 3562-3577, 2019 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-31145102

RESUMO

BACKGROUND: While the human fetal immune system defaults to a program of tolerance, there is concurrent need for protective immunity to meet the antigenic challenges encountered after birth. Activation of T cells in utero is associated with the fetal inflammatory response with broad implications for the health of the fetus and of the pregnancy. However, the characteristics of the fetal effector T cells that contribute to this process are largely unknown. METHODS: We analyzed primary human fetal lymphoid and mucosal tissues and performed phenotypic, functional, and transcriptional analysis to identify T cells with pro-inflammatory potential. The frequency and function of fetal-specific effector T cells was assessed in the cord blood of infants with localized and systemic inflammatory pathologies and compared to healthy term controls. RESULTS: We identified a transcriptionally distinct population of CD4+ T cells characterized by expression of the transcription factor Promyelocytic Leukemia Zinc Finger (PLZF). PLZF+ CD4+ T cells were specifically enriched in the fetal intestine, possessed an effector memory phenotype, and rapidly produced pro-inflammatory cytokines. Engagement of the C-type lectin CD161 on these cells inhibited TCR-dependent production of IFNγ in a fetal-specific manner. IFNγ-producing PLZF+ CD4+ T cells were enriched in the cord blood of infants with gastroschisis, a natural model of chronic inflammation originating from the intestine, as well as in preterm birth, suggesting these cells contribute to fetal systemic immune activation. CONCLUSION: Our work reveals a fetal-specific program of protective immunity whose dysregulation is associated with fetal and neonatal inflammatory pathologies.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Sistema Imunitário , Intestinos/embriologia , Tecido Linfoide/embriologia , Mucosa/embriologia , Subfamília B de Receptores Semelhantes a Lectina de Células NK/metabolismo , Linfócitos T/citologia , Linfócitos T CD4-Positivos/citologia , Estudos de Casos e Controles , Feminino , Sangue Fetal/citologia , Feto/imunologia , Regulação da Expressão Gênica , Humanos , Memória Imunológica , Terapia de Imunossupressão , Recém-Nascido , Inflamação , Interferon gama/metabolismo , Intestinos/imunologia , Leucócitos Mononucleares/citologia , Ativação Linfocitária , Fenótipo , Gravidez , Proteína com Dedos de Zinco da Leucemia Promielocítica/metabolismo , Linfócitos T/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA