Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
BMJ Case Rep ; 17(6)2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38839399

RESUMO

A male infant was born at 40 and 4/7 weeks of gestation via caesarean section for non-reassuring foetal heart tracing. The infant was non-responsive in the delivery room. with no heart rate detected until 40 min of life. The infant's physical examination and laboratory findings were consistent with severe hypoxic-ischaemic encephalopathy. Given the presumption of a very poor neurological prognosis, redirection to comfort care was recommended to the family. However, the family opted for intensive care. The infant underwent therapeutic hypothermia and management of multiorgan dysfunction. The infant survived with no findings of ischaemic injury on MRI and was discharged with no respiratory support and taking all feeds by mouth, with normal development at a year and a half of age. This case report demonstrates the imperative to understand family goals and to acknowledge the need for ongoing humility in providing prognostication for families.


Assuntos
Hipotermia Induzida , Hipóxia-Isquemia Encefálica , Humanos , Hipóxia-Isquemia Encefálica/terapia , Hipóxia-Isquemia Encefálica/complicações , Masculino , Recém-Nascido , Hipotermia Induzida/métodos , Imageamento por Ressonância Magnética , Cesárea
2.
bioRxiv ; 2024 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-38645130

RESUMO

The immunological defects causing susceptibility to severe viral respiratory infections due to early-life dysbiosis remain ill-defined. Here, we show that influenza virus susceptibility in dysbiotic infant mice is caused by CD8+ T cell hyporesponsiveness and diminished persistence as tissue-resident memory cells. We describe a previously unknown role for nuclear factor interleukin 3 (NFIL3) in repression of memory differentiation of CD8+ T cells in dysbiotic mice involving epigenetic regulation of T cell factor 1 (TCF 1) expression. Pulmonary CD8+ T cells from dysbiotic human infants share these transcriptional signatures and functional phenotypes. Mechanistically, intestinal inosine was reduced in dysbiotic human infants and newborn mice, and inosine replacement reversed epigenetic dysregulation of Tcf7 and increased memory differentiation and responsiveness of pulmonary CD8+ T cells. Our data unveils new developmental layers controlling immune cell activation and identifies microbial metabolites that may be used therapeutically in the future to protect at-risk newborns.

3.
Gut Microbes ; 14(1): 2143222, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36404471

RESUMO

Immunotherapy has led to impressive advances in the treatment of autoimmune and pro-inflammatory disorders; yet, its clinical outcomes remain limited by a variety of factors including the pro-inflammatory microenvironment (IME). Discovering effective immunomodulatory agents, and the mechanisms by which they control disease, will lead to innovative strategies for enhancing the effectiveness of current immunotherapeutic approaches. We have metabolically engineered an attenuated bacterial strain (i.e., Brucella melitensis 16M ∆vjbR, Bm∆vjbR::tnaA) to produce indole, a tryptophan metabolite that controls the fate and function of regulatory T (Treg) cells. We demonstrated that treatment with Bm∆vjbR::tnaA polarized macrophages (Mφ) which produced anti-inflammatory cytokines (e.g., IL-10) and promoted Treg function; moreover, when combined with adoptive cell transfer (ACT) of Treg cells, a single treatment with our engineered bacterial strain dramatically reduced the incidence and score of autoimmune arthritis and decreased joint damage. These findings show how a metabolically engineered bacterium can constitute a powerful vehicle for improving the efficacy of immunotherapy, defeating autoimmunity, and reducing inflammation by remodeling the IME and augmenting Treg cell function.


Assuntos
Autoimunidade , Microbioma Gastrointestinal , Humanos , Inflamação , Citocinas/metabolismo , Linfócitos T Reguladores , Bactérias/metabolismo
4.
Sci Transl Med ; 14(649): eabl3981, 2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35704600

RESUMO

Although modern clinical practices such as cesarean sections and perinatal antibiotics have improved infant survival, treatment with broad-spectrum antibiotics alters intestinal microbiota and causes dysbiosis. Infants exposed to perinatal antibiotics have an increased likelihood of life-threatening infections, including pneumonia. Here, we investigated how the gut microbiota sculpt pulmonary immune responses, promoting recovery and resolution of infection in newborn rhesus macaques. Early-life antibiotic exposure interrupted the maturation of intestinal commensal bacteria and disrupted the developmental trajectory of the pulmonary immune system, as assessed by single-cell proteomic and transcriptomic analyses. Early-life antibiotic exposure rendered newborn macaques more susceptible to bacterial pneumonia, concurrent with increases in neutrophil senescence and hyperinflammation, broad inflammatory cytokine signaling, and macrophage dysfunction. This pathogenic reprogramming of pulmonary immunity was further reflected by a hyperinflammatory signature in all pulmonary immune cell subsets coupled with a global loss of tissue-protective, homeostatic pathways in the lungs of dysbiotic newborns. Fecal microbiota transfer was associated with partial correction of the broad immune maladaptations and protection against severe pneumonia. These data demonstrate the importance of intestinal microbiota in programming pulmonary immunity and support the idea that gut microbiota promote the balance between pathways driving tissue repair and inflammatory responses associated with clinical recovery from infection in infants. Our results highlight a potential role for microbial transfer for immune support in these at-risk infants.


Assuntos
Microbioma Gastrointestinal , Pneumonia , Animais , Antibacterianos , Disbiose , Feminino , Humanos , Imunidade , Pulmão , Macaca mulatta , Gravidez , Proteômica
5.
Sci Transl Med ; 14(638): eabl8574, 2022 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-35353543

RESUMO

Perinatal inflammatory stress is associated with early life morbidity and lifelong consequences for pulmonary health. Chorioamnionitis, an inflammatory condition affecting the placenta and fluid surrounding the developing fetus, affects 25 to 40% of preterm births. Severe chorioamnionitis with preterm birth is associated with significantly increased risk of pulmonary disease and secondary infections in childhood, suggesting that fetal inflammation may markedly alter the development of the lung. Here, we used intra-amniotic lipopolysaccharide (LPS) challenge to induce experimental chorioamnionitis in a prenatal rhesus macaque (Macaca mulatta) model that mirrors structural and temporal aspects of human lung development. Inflammatory injury directly disrupted the developing gas exchange surface of the primate lung, with extensive damage to alveolar structure, particularly the close association and coordinated differentiation of alveolar type 1 pneumocytes and specialized alveolar capillary endothelium. Single-cell RNA sequencing analysis defined a multicellular alveolar signaling niche driving alveologenesis that was extensively disrupted by perinatal inflammation, leading to a loss of gas exchange surface and alveolar simplification, with notable resemblance to chronic lung disease in newborns. Blockade of the inflammatory cytokines interleukin-1ß and tumor necrosis factor-α ameliorated LPS-induced inflammatory lung injury by blunting stromal responses to inflammation and modulating innate immune activation in myeloid cells, restoring structural integrity and key signaling networks in the developing alveolus. These data provide new insight into the pathophysiology of developmental lung injury and suggest that modulating inflammation is a promising therapeutic approach to prevent fetal consequences of chorioamnionitis.


Assuntos
Corioamnionite , Nascimento Prematuro , Animais , Corioamnionite/induzido quimicamente , Corioamnionite/patologia , Feminino , Pulmão/patologia , Macaca mulatta , Gravidez , Nascimento Prematuro/prevenção & controle , Troca Gasosa Pulmonar
6.
Biotechnol Bioeng ; 114(11): 2660-2667, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28667749

RESUMO

Adoptive transfer of anti-inflammatory FOXP3+ Tregs has gained attention as a new therapeutic strategy for auto-inflammatory disorders such as Inflammatory Bowel Disease. The isolated cells are conditioned in vitro to obtain a sufficient number of anti-inflammatory FOXP3+ Tregs that can be reintroduced into the patient to potentially reduce the pathologic inflammatory response. Previous evidence suggests that microbiota metabolites can potentially condition cells during the in vitro expansion/differentiation step. However, the number of combinations of cytokines and metabolites that can be varied is large, preventing a purely experimental investigation which would determine optimal cell therapeutic outcomes. To address this problem, a combined experimental and modeling approached is investigated here: an artificial neural network model was trained to predict the steady-state T cell population phenotype after differentiation with a variety of host cytokines and the microbial metabolite indole. This artificial neural network model was able to both reliably predict the phenotype of these T cell populations and also uncover unexpected conditions for optimal Treg differentiation that were subsequently verified experimentally. Biotechnol. Bioeng. 2017;114: 2660-2667. © 2017 Wiley Periodicals, Inc.


Assuntos
Bactérias/imunologia , Citocinas/imunologia , Microbioma Gastrointestinal/imunologia , Indóis/imunologia , Ativação Linfocitária/imunologia , Modelos Imunológicos , Linfócitos T/imunologia , Células Cultivadas , Humanos , Redes Neurais de Computação , Linfócitos T/classificação
7.
Gut Microbes ; 7(3): 246-61, 2016 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-27007819

RESUMO

Non-steroidal anti-inflammatory drugs (NSAIDs) are one of the most frequently used classes of medications in the world. Unfortunately, NSAIDs induce an enteropathy associated with high morbidity and mortality. Although the pathophysiology of this condition involves the interaction of the gut epithelium, microbiota, and NSAIDs, the precise mechanisms by which microbiota influence NSAID enteropathy are unclear. One possible mechanism is that the microbiota may attenuate the severity of disease by specific metabolite-mediated regulation of host inflammation and injury. The microbiota-derived tryptophan-metabolite indole is abundant in the healthy mammalian gut and positively influences intestinal health. We thus examined the effects of indole administration on NSAID enteropathy. Mice (n = 5 per group) were treated once daily for 7 days with an NSAID (indomethacin; 5 mg/kg), indole (20 mg/kg), indomethacin plus indole, or vehicle only (control). Outcomes compared among groups included: microscopic pathology; fecal calprotectin concentration; proportion of neutrophils in the spleen and mesenteric lymph nodes; fecal microbiota composition and diversity; small intestinal mucosal transcriptome; and, fecal tryptophan metabolites. Co-administration of indole with indomethacin: significantly reduced mucosal pathology scores, fecal calprotectin concentrations, and neutrophilic infiltration of the spleen and mesenteric lymph nodes induced by indomethacin; modulated NSAID-induced perturbation of the microbiota, fecal metabolites, and inferred metagenome; and, abrogated a pro-inflammatory gene expression profile in the small intestinal mucosa induced by indomethacin. The microbiota-derived metabolite indole attenuated multiple deleterious effects of NSAID enteropathy, including modulating inflammation mediated by innate immune responses and altering indomethacin-induced shift of the microbiota.


Assuntos
Anti-Inflamatórios não Esteroides/efeitos adversos , Anti-Inflamatórios/metabolismo , Enterite/tratamento farmacológico , Fármacos Gastrointestinais/farmacologia , Indóis/metabolismo , Indóis/farmacologia , Inflamação/patologia , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Bactérias/classificação , Bactérias/isolamento & purificação , Biota , Modelos Animais de Doenças , Enterite/induzido quimicamente , Fezes/química , Fezes/microbiologia , Fármacos Gastrointestinais/administração & dosagem , Histocitoquímica , Indóis/administração & dosagem , Complexo Antígeno L1 Leucocitário/análise , Linfonodos/patologia , Camundongos , Neutrófilos/imunologia , Baço/patologia , Resultado do Tratamento
8.
Nat Commun ; 5: 5492, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25411059

RESUMO

Metabolites produced by the intestinal microbiota are potentially important physiological modulators. Here we present a metabolomics strategy that models microbiota metabolism as a reaction network and utilizes pathway analysis to facilitate identification and characterization of microbiota metabolites. Of the 2,409 reactions in the model, ~53% do not occur in the host, and thus represent functions dependent on the microbiota. The largest group of such reactions involves amino-acid metabolism. Focusing on aromatic amino acids, we predict metabolic products that can be derived from these sources, while discriminating between microbiota- and host-dependent derivatives. We confirm the presence of 26 out of 49 predicted metabolites, and quantify their levels in the caecum of control and germ-free mice using two independent mass spectrometry methods. We further investigate the bioactivity of the confirmed metabolites, and identify two microbiota-generated metabolites (5-hydroxy-L-tryptophan and salicylate) as activators of the aryl hydrocarbon receptor.


Assuntos
Ceco/metabolismo , Metaboloma , Microbiota , Animais , Ceco/microbiologia , Espectrometria de Massas , Camundongos
9.
Philos Trans R Soc Lond B Biol Sci ; 365(1548): 1931-41, 2010 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-20478888

RESUMO

We study the epidemiology of a viral disease with dose-dependent replication and transmission by nesting a differential-equation model of the within-host viral dynamics inside a between-host epidemiological model. We use two complementary approaches for nesting the models: an agent-based (AB) simulation and a mean-field approximation called the growth-matrix (GM) model. We find that although infection rates and predicted case loads are somewhat different between the AB and GM models, several epidemiological parameters, e.g. mean immunity in the population and mean dose received, behave similarly across the methods. Further, through a comparison of our dose-dependent replication model against two control models that uncouple dose-dependent replication from transmission, we find that host immunity in a population after an epidemic is qualitatively different than when transmission depends on time-varying viral abundances within hosts. These results show that within-host dynamics and viral dose should not be neglected in epidemiological models, and that the simpler GM approach to model nesting provides a reasonable tradeoff between model complexity and accuracy of results.


Assuntos
Surtos de Doenças , Relação Dose-Resposta Imunológica , Modelos Imunológicos , Viroses/imunologia , Vírus/imunologia , Simulação por Computador , Interações Hospedeiro-Patógeno/imunologia , Humanos , Processos Estocásticos , Viroses/transmissão , Viroses/virologia
10.
J Theor Biol ; 261(1): 67-73, 2009 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-19627995

RESUMO

We analyze how lethal mutagenesis operates in a compartmentalized host. We assume that different compartments receive different amounts of mutagen and that virions can migrate among compartments. We address two main questions: (1) To what extent can refugia, i.e., compartments that receive little mutagen, prevent extinction? (2) Does migration among compartments limit the effectiveness of refugia? We find that if there is little migration, extinction has to be achieved separately in all compartments. In this case, the total dose of mutagen administered to the host needs to be so high that the mutagen is effective even in the refugia. By contrast, if migration is extensive, then lethal mutagenesis is effective as long as the average growth in all compartments is reduced to below replacement levels. The effectiveness of migration is governed by the ratio of virion replication and death rates, R(0). The smaller R(0), the less migration is necessary to neutralize refugia and the less mutagen is necessary to achieve extinction at high migration rates.


Assuntos
Modelos Biológicos , Mutagênese , Mutagênicos/farmacocinética , Vírus/genética , Antivirais/farmacocinética , Antivirais/farmacologia , Humanos , Mutagênicos/farmacologia , Vírion/fisiologia , Replicação Viral , Vírus/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...