Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Leukoc Biol ; 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38470858

RESUMO

The nexus between eosinophils and microbes is attracting increasing attention. We previously showed that airway administration of sterile microbial products contained in dust collected from traditional dairy farms virtually abrogated broncho-alveolar lavage (BAL) eosinophilia and other cardinal asthma phenotypes in allergen-sensitized specific pathogen-free (SPF) mice. Interestingly, comparable inhibition of allergen-induced BAL eosinophilia and promotion of airway barrier integrity were found upon administration of a sterile, pharmacological grade bacterial lysate, OM-85, to the airway compartment of allergen-sensitized SPF mice. Here we asked whether intrinsic properties of airway-delivered microbial products were sufficient to inhibit allergic lung inflammation or whether these effects were mediated by reprogramming of the host microbiota. We compared germ-free (GF) mice and offspring of GF mice associated with healthy mouse gut microbiota and maintained under SPF conditions for multiple generations (Ex-GF mice). These mice were treated intra-nasally with OM-85 and evaluated in the OVA and Alternaria models of allergic asthma focusing primarily on BAL eosinophilia. Levels of allergen-induced BAL eosinophilia were comparable in GF and conventionalized Ex-GF mice. Airway administration of the OM-85 bacterial lysate was sufficient to inhibit allergen-induced lung eosinophilia in both Ex-GF and GF mice, suggesting that host microbiota are not required for the protective effects of bacterial products in these models and local airway exposure to microbial products is an effective source of protection. OM-85-dependent inhibition of BAL eosinophilia in GF mice was accompanied by suppression of lung type-2 cytokines and eosinophil-attracting chemokines, suggesting that OM-85 may work at least by decreasing eosinophil lung recruitment.

2.
Allergy ; 77(12): 3617-3628, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35841380

RESUMO

BACKGROUND: The path to childhood asthma is thought to initiate in utero and be further promoted by postnatal exposures. However, the underlying mechanisms remain underexplored. We hypothesized that prenatal maternal immune dysfunction associated with increased childhood asthma risk (revealed by low IFN-γ:IL-13 secretion during the third trimester of pregnancy) alters neonatal immune training through epigenetic mechanisms and promotes early-life airway colonization by asthmagenic microbiota. METHODS: We examined epigenetic, immunologic, and microbial features potentially related to maternal prenatal immunity (IFN-γ:IL-13 ratio) and childhood asthma in a birth cohort of mother-child dyads sampled pre-, peri-, and postnatally (N = 155). Epigenome-wide DNA methylation and cytokine production were assessed in cord blood mononuclear cells (CBMC) by array profiling and ELISA, respectively. Nasopharyngeal microbiome composition was characterized at age 2-36 months by 16S rRNA sequencing. RESULTS: Maternal prenatal immune status related to methylome profiles in neonates born to non-asthmatic mothers. A module of differentially methylated CpG sites enriched for microbe-responsive elements was associated with childhood asthma. In vitro responsiveness to microbial products was impaired in CBMCs from neonates born to mothers with the lowest IFN-γ:IL-13 ratio, suggesting defective neonatal innate immunity in those who developed asthma during childhood. These infants exhibited a distinct pattern of upper airway microbiota development characterized by early-life colonization by Haemophilus that transitioned to a Moraxella-dominated microbiota by age 36 months. CONCLUSIONS: Maternal prenatal immune status shapes asthma development in her child by altering the epigenome and trained innate immunity at birth, and is associated with pathologic upper airway microbial colonization in early life.


Assuntos
Asma , Microbiota , Humanos , Lactente , Recém-Nascido , Gravidez , Feminino , Pré-Escolar , Interleucina-13 , RNA Ribossômico 16S , Sistema Respiratório , Microbiota/genética
3.
J Allergy Clin Immunol ; 149(3): 943-956, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34560105

RESUMO

BACKGROUND: Microbial interventions against allergic asthma have robust epidemiologic underpinnings and the potential to recalibrate disease-inducing immune responses. Oral administration of OM-85, a standardized lysate of human airways bacteria, is widely used empirically to prevent respiratory infections and a clinical trial is testing its ability to prevent asthma in high-risk children. We previously showed that intranasal administration of microbial products from farm environments abrogates experimental allergic asthma. OBJECTIVES: We sought to investigate whether direct administration of OM-85 to the airway compartment protects against experimental allergic asthma; and to identify protective cellular and molecular mechanisms activated through this natural route. METHODS: Different strains of mice sensitized and challenged with ovalbumin or Alternaria received OM-85 intranasally, and cardinal cellular and molecular asthma phenotypes were measured. Airway transfer experiments assessed whether OM-85-treated dendritic cells protect allergen-sensitized, OM-85-naive mice against asthma. RESULTS: Airway OM-85 administration suppressed allergic asthma in all models acting on multiple innate and adaptive immune targets: the airway epithelium/IL-33/ILC2 axis, lung allergen-induced type 2 responses, and dendritic cells whose Myd88/Trif-dependent tolerogenic reprogramming was sufficient to transfer OM-85-induced asthma protection. CONCLUSIONS: We provide the first demonstration that administering a standardized bacterial lysate to the airway compartment protects from experimental allergic asthma by engaging multiple immune pathways. Because protection required a cumulative dose 27- to 46-fold lower than the one reportedly active through the oral route, the efficacy of intranasal OM-85 administration may reflect its direct access to the airway mucosal networks controlling the initiation and development of allergic asthma.


Assuntos
Asma , Interleucina-33 , Alérgenos , Animais , Extratos Celulares , Células Dendríticas , Modelos Animais de Doenças , Epitélio , Humanos , Imunidade Inata , Pulmão , Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina
4.
J Allergy Clin Immunol ; 149(3): 923-933.e6, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34902435

RESUMO

BACKGROUND: Treatments for coronavirus disease 2019, which is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), are urgently needed but remain limited. SARS-CoV-2 infects cells through interactions of its spike (S) protein with angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2) on host cells. Multiple cells and organs are targeted, particularly airway epithelial cells. OM-85, a standardized lysate of human airway bacteria with strong immunomodulating properties and an impeccable safety profile, is widely used to prevent recurrent respiratory infections. We found that airway OM-85 administration inhibits Ace2 and Tmprss2 transcription in the mouse lung, suggesting that OM-85 might hinder SARS-CoV-2/host cell interactions. OBJECTIVES: We sought to investigate whether and how OM-85 treatment protects nonhuman primate and human epithelial cells against SARS-CoV-2. METHODS: ACE2 and TMPRSS2 mRNA and protein expression, cell binding of SARS-CoV-2 S1 protein, cell entry of SARS-CoV-2 S protein-pseudotyped lentiviral particles, and SARS-CoV-2 cell infection were measured in kidney, lung, and intestinal epithelial cell lines, primary human bronchial epithelial cells, and ACE2-transfected HEK293T cells treated with OM-85 in vitro. RESULTS: OM-85 significantly downregulated ACE2 and TMPRSS2 transcription and surface ACE2 protein expression in epithelial cell lines and primary bronchial epithelial cells. OM-85 also strongly inhibited SARS-CoV-2 S1 protein binding to, SARS-CoV-2 S protein-pseudotyped lentivirus entry into, and SARS-CoV-2 infection of epithelial cells. These effects of OM-85 appeared to depend on SARS-CoV-2 receptor downregulation. CONCLUSIONS: OM-85 inhibits SARS-CoV-2 epithelial cell infection in vitro by downregulating SARS-CoV-2 receptor expression. Further studies are warranted to assess whether OM-85 may prevent and/or reduce the severity of coronavirus disease 2019.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , COVID-19/prevenção & controle , Extratos Celulares/administração & dosagem , Receptores Virais/antagonistas & inibidores , Receptores Virais/imunologia , SARS-CoV-2/imunologia , Enzima de Conversão de Angiotensina 2/antagonistas & inibidores , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/imunologia , Animais , COVID-19/imunologia , COVID-19/virologia , Células CACO-2 , Extratos Celulares/imunologia , Células Cultivadas , Chlorocebus aethiops , Regulação para Baixo/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/virologia , Células HEK293 , Interações entre Hospedeiro e Microrganismos/efeitos dos fármacos , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Técnicas In Vitro , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Serina Endopeptidases/efeitos dos fármacos , Serina Endopeptidases/genética , Serina Endopeptidases/imunologia , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/imunologia , Células Vero
5.
Am J Respir Cell Mol Biol ; 59(5): 592-600, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29863910

RESUMO

Little is known about whether maternal immune status during pregnancy influences asthma development in the child. We measured cytokine production in supernatants from mitogen-stimulated peripheral blood immune cells collected during and after pregnancy from the mothers of children enrolled in the Tucson Infant Immune Study, a nonselected birth cohort. Physician-diagnosed active asthma in children through age 9 and a history of asthma in their mothers were assessed through questionnaires. Maternal production of each of the cytokines IL-13, IL-4, IL-5, IFN-γ, IL-10, and IL-17 during pregnancy was unrelated to childhood asthma. However, IFN-γ/IL-13 and IFN-γ/IL-4 ratios during pregnancy were associated with a decreased risk of childhood asthma (n = 381; odds ratio [OR], 0.33; 95% confidence interval [CI], 0.17-0.66; P = 0.002; and n = 368; OR, 0.36; 95% CI, 0.18-0.71; P = 0.003, respectively). The inverse relations of these two ratios with childhood asthma were only evident in mothers without asthma (n = 309; OR, 0.18; 95% CI, 0.08-0.42; P = 0.00007; and n = 299; OR, 0.17; 95% CI, 0.07-0.39; P = 0.00003, respectively) and not in mothers with asthma (n = 72 and 69, respectively; P for interaction by maternal asthma = 0.036 and 0.002, respectively). Paternal cytokine ratios were unrelated to childhood asthma. Maternal cytokine ratios in mothers without asthma were unrelated to the children's skin-test reactivity, total IgE, physician-confirmed allergic rhinitis at age 5, or eczema in infancy. To our knowledge, this study provides the first evidence that cytokine profiles in pregnant mothers without asthma relate to the risk for childhood asthma, but not allergy, and suggests a process of asthma development that begins in utero and is independent of allergy.


Assuntos
Asma/epidemiologia , Asma/imunologia , Citocinas/sangue , Interferon gama/sangue , Interleucina-13/sangue , Interleucina-4/sangue , Mães/estatística & dados numéricos , Adulto , Asma/sangue , Criança , Pré-Escolar , Citocinas/imunologia , Feminino , Humanos , Interferon gama/imunologia , Interleucina-13/imunologia , Interleucina-4/imunologia , Masculino , Valor Preditivo dos Testes , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Prevalência , Estudos Prospectivos , Curva ROC
6.
J Allergy Clin Immunol ; 141(6): 1992-1996, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29709672

RESUMO

The study of developmental trajectories is where epigenetics truly shines. The "epi" in epigenetics captures the fact that although epigenetic processes also preside over the maintenance and termination of gene expression, the unfolding and remodeling of chromatin architecture are especially critical to prepare genes for regulated transcription. These properties imply being on a path, a trajectory to events that will occur later thanks to epigenetic programming. Thus epigenetics is about timed and timely events. In this article we discuss epigenetic and genetic evidence from several independent studies of asthma, chronic obstructive pulmonary disease, and lung function, which converge to highlight a potential role of the TGF-ß gene pathway in these processes. These results raise the possibility that at least in a subset of subjects, these conditions might be functionally connected in ways that need to be further defined but that likely reflect the uniquely pleiotropic nature of TGF-ß pathway genes, particularly their ability to control both lung development and immune responses essential for regulation and inflammation. Further characterization of this pathway in longitudinally phenotyped populations might unmask novel trajectories to lung disease that begin in utero and unfold into old age.


Assuntos
Asma/genética , Pleiotropia Genética/genética , Doença Pulmonar Obstrutiva Crônica/genética , Fator de Crescimento Transformador beta/genética , Adulto , Animais , Asma/fisiopatologia , Criança , Epigênese Genética , Humanos , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Transdução de Sinais/fisiologia
7.
Epigenomics ; 9(4): 585-593, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28322586

RESUMO

Asthma is a heterogeneous group of conditions that typically begin in early life and result in recurrent, reversible bronchial obstruction. The role played by epigenetic mechanisms in the pathogenesis of childhood asthma is understood only in part. Here we discuss asthma epigenetics within a developmental perspective based on our recent demonstration that the epigenetic trajectory to childhood asthma begins at birth. We next discuss how this trajectory may be affected by prenatal environmental exposures. Finally, we examine in vitro studies that model the impact of asthma-associated exposures on the epigenome. All of these studies specifically surveyed human DNA methylation and involved a genome-wide component. In combination, their results broaden our understanding of asthma pathogenesis and the role the methylome plays in this process.


Assuntos
Asma/genética , Metilação de DNA , Epigenômica/métodos , Animais , Criança , Metilação de DNA/efeitos dos fármacos , Modelos Animais de Doenças , Exposição Ambiental/efeitos adversos , Predisposição Genética para Doença , Humanos , Recém-Nascido , Fatores de Risco
8.
J Allergy Clin Immunol ; 140(2): 534-542, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28011059

RESUMO

BACKGROUND: The timing and mechanisms of asthma inception remain imprecisely defined. Although epigenetic mechanisms likely contribute to asthma pathogenesis, little is known about their role in asthma inception. OBJECTIVE: We sought to assess whether the trajectory to asthma begins already at birth and whether epigenetic mechanisms, specifically DNA methylation, contribute to asthma inception. METHODS: We used the Methylated CpG Island Recovery Assay chip to survey DNA methylation in cord blood mononuclear cells from 36 children (18 nonasthmatic and 18 asthmatic subjects by age 9 years) from the Infant Immune Study (IIS), an unselected birth cohort closely monitored for asthma for a decade. SMAD3 methylation in IIS (n = 60) and in 2 replication cohorts (the Manchester Asthma and Allergy Study [n = 30] and the Childhood Origins of Asthma Study [n = 28]) was analyzed by using bisulfite sequencing or Illumina 450K arrays. Cord blood mononuclear cell-derived IL-1ß levels were measured by means of ELISA. RESULTS: Neonatal immune cells harbored 589 differentially methylated regions that distinguished IIS children who did and did not have asthma by age 9 years. In all 3 cohorts methylation in SMAD3, the most connected node within the network of asthma-associated, differentially methylated regions, was selectively increased in asthmatic children of asthmatic mothers and was associated with childhood asthma risk. Moreover, SMAD3 methylation in IIS neonates with maternal asthma was strongly and positively associated with neonatal production of IL-1ß, an innate inflammatory mediator. CONCLUSIONS: The trajectory to childhood asthma begins at birth and involves epigenetic modifications in immunoregulatory and proinflammatory pathways. Maternal asthma influences epigenetic mechanisms that contribute to the inception of this trajectory.


Assuntos
Asma/genética , Proteína Smad3/genética , Criança , Pré-Escolar , Ilhas de CpG , Metilação de DNA , Epigênese Genética , Sangue Fetal/citologia , Humanos , Recém-Nascido , Interleucina-1beta/metabolismo , Leucócitos Mononucleares/metabolismo , Mães , Regiões Promotoras Genéticas
9.
Ann Am Thorac Soc ; 13 Suppl 1: S48-50, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27027952

RESUMO

Asthma and allergic diseases are among the most prevalent chronic noncommunicable diseases of childhood, but the underlying pathogenetic mechanisms are poorly understood. Because epigenetic mechanisms link gene regulation to environmental cues and developmental trajectories, their contribution to asthma and allergy pathogenesis is under active investigation. DNA methylation signatures associated with concurrent disease and with the development of asthma during childhood asthma have been identified, but their significance is not easily interpretable. On the other hand, the characterization of early epigenetic predictors of asthma points to a potential role of epigenetic mechanisms in regulating the inception of, and the susceptibility to, this disease.


Assuntos
Asma/genética , Epigênese Genética/genética , Epigenômica/métodos , Predisposição Genética para Doença , Exposição Ambiental/efeitos adversos , Humanos
10.
Curr Opin Pediatr ; 27(6): 719-23, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26418323

RESUMO

PURPOSE OF REVIEW: Allergic diseases are among the most prevalent chronic diseases of childhood, affecting more than 7 million children in the United States. Epidemiological evidence supports the idea that the inception of allergic diseases is typically before the preschool years, even when chronic symptoms do not emerge until adulthood. The role of epigenetic mechanisms (particularly DNA methylation) in allergic disease is under active investigation because these mechanisms are known to be at the interface of gene regulation, environmental stimuli, and developmental processes, all of which are essential for the pathogenesis for asthma and allergy. This article specifically reviews genome-wide DNA methylation studies in allergic disease. RECENT FINDINGS: Differential DNA methylation at specific regions appears to be associated with concurrent allergic disease. A few studies have identified methylation signatures predictive of disease. SUMMARY: DNA methylation signatures have been shown to be associated with several allergic disease phenotypes, typically concurrently with disease. The few that have been found to precede diagnosis are especially interesting because they highlight an early trajectory to disease.


Assuntos
Asma/genética , Metilação de DNA/genética , Exposição Ambiental/efeitos adversos , Epigenômica/métodos , Rinite Alérgica/genética , Adolescente , Asma/epidemiologia , Asma/imunologia , Criança , Pré-Escolar , Suscetibilidade a Doenças , Interação Gene-Ambiente , Humanos , Fenótipo , Rinite Alérgica/epidemiologia , Rinite Alérgica/imunologia , Estados Unidos/epidemiologia
11.
Curr Opin Allergy Clin Immunol ; 15(5): 435-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26258923

RESUMO

PURPOSE OF REVIEW: Asthma and allergic diseases are among the most prevalent chronic noncommunicable diseases of childhood. Although epidemiologic studies suggest that asthma begins in the preschool years, the lack of firm diagnostic criteria to distinguish children who will wheeze only transiently during early-life lower respiratory illnesses from children who will wheeze persistently and develop asthma prevents pinpointing the time at which disease truly begins. Epigenetic mechanisms link gene regulation to environmental cues and developmental trajectories. This article reviews, the search for epigenetic predictors of asthma and/or allergy that can be identified already at birth and/or in early life. RECENT FINDINGS: DNA methylation signatures associated with asthma and/or allergy at birth, and time-dependent DNA methylation signatures associated with allergic disease phenotypes in early life have been identified. SUMMARY: The identification of early epigenetic predictors of allergic diseases points to a potential role of epigenetic mechanisms in regulating the inception of and the susceptibility to these diseases. Predictive signatures to more accurately estimate a child's risk for asthma and allergy may improve childhood asthma diagnosis. Moreover, understanding the biological implications of these signatures may help elucidate novel disease pathways and endotypes.


Assuntos
Asma/diagnóstico , Epigenômica , Hipersensibilidade/diagnóstico , Animais , Asma/genética , Biomarcadores/metabolismo , Criança , Metilação de DNA , Diagnóstico Precoce , Humanos , Hipersensibilidade/genética , Prognóstico , Transcriptoma/imunologia
12.
Asian Pac J Allergy Immunol ; 31(3): 183-9, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24053700

RESUMO

OBJECTIVE: The interest in asthma epigenetics is high because epigenetic mechanisms likely contribute to the environmental origins of the disease and its phenotypic variability. This review presents the main findings of asthma epigenetics and the challenges that still delay progress. DATA SOURCES: We examined the current literature on asthma epigenetics (31 reviews and 25 original data publications). STUDY SELECTIONS: We focused on DNA methylation studies in populations. RESULTS: Both genome-wide and candidate gene studies have explored DNA methylation in allergic disease. Genome-wide studies ask whether and which regions of the genome are differentially methylated in relation to the phenotype of interest. Identification of such regions provides clues about the identity of the genes, pathways and networks underpinning a phenotype and connects these networks to the phenotype through epigenetic mechanisms. Candidate gene studies examine DNA methylation in genes chosen because of their known or hypothesized role in immunity, responses to environmental stimuli or disease pathogenesis. Most existing studies in asthma and allergy focused on candidate genes involved in the response to environmental pollutants. CONCLUSION: Asthma epigenetics is still in its infancy. The paucity of primary literature originates from methodological and analytical challenges of genome-wide studies, the difficulties in interpreting small differences in DNA methylation, and the need to develop robust bioinformatic tools for pathway, network and system analyses of epigenetic data. Once these challenges have been overcome, epigenetic studies will likely provide important insights about the inception and pathogenesis of allergic disease and will help define disease endotypes.


Assuntos
Asma/imunologia , Metilação de DNA/imunologia , Exposição Ambiental/efeitos adversos , Epigênese Genética/imunologia , Interação Gene-Ambiente , Asma/etiologia , Estudo de Associação Genômica Ampla , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...