Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
Brain Behav Immun ; 122: 44-57, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39128569

RESUMO

Prenatal stress (PNS) profoundly impacts maternal and offspring health, with enduring effects including microbiome alterations, neuroinflammation, and behavioral disturbances such as reductions in social behavior. Converging lines of evidence from preclinical and clinical studies suggest that PNS disrupts tryptophan (Trp) metabolic pathways and reduces gut Bifidobacteria, a known beneficial bacterial genus that metabolizes Trp. Specifically, previous work from our lab demonstrated that human prenatal mood disorders in mothers are associated with reduced Bifidobacterium dentium in infants at 13 months. Given that Bifidobacterium has been positively associated with neurodevelopmental and other health benefits and is depleted by PNS, we hypothesized that supplementing PNS-exposed pregnant dams with B. dentium would ameliorate PNS-induced health deficits. We measured inflammatory outputs, Trp metabolite levels and enzymatic gene expression in dams and fetal offspring, and social behavior in adult offspring. We determined that B. dentium reduced maternal systemic inflammation and fetal offspring neuroinflammation, while modulating tryptophan metabolism and increasing kynurenic acid and indole-3-propionic acid intergenerationally. Additional health benefits were demonstrated by the abrogation of PNS-induced reductions in litter weight. Finally, offspring of the B. dentium cohort demonstrated increased sociability in males primarily and increased social novelty primarily in females. Together these data illustrate that B. dentium can orchestrate interrelated host immune, metabolic and behavioral outcomes during and after gestation for both dam and offspring and may be a candidate for prevention of the negative sequelae of stress.


Assuntos
Inflamação , Efeitos Tardios da Exposição Pré-Natal , Comportamento Social , Triptofano , Feminino , Gravidez , Animais , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/microbiologia , Triptofano/metabolismo , Inflamação/metabolismo , Masculino , Bifidobacterium/metabolismo , Estresse Psicológico/metabolismo , Microbioma Gastrointestinal/fisiologia , Comportamento Animal/fisiologia , Probióticos/farmacologia , Ratos
2.
Ecotoxicol Environ Saf ; 283: 116861, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-39137463

RESUMO

BACKGROUND: The gut microbiome is central to human health, but the potential impact of ozone (O3) exposure on its establishment in early life has not been thoroughly examined. Therefore, this study aimed to investigate the relationship between prenatal O3 exposure and the variations of the human gut microbiome during the first two years of life. DESIGN: A cohort study design was used. Pregnant women in the third trimester were recruited from an obstetric clinic, and long-term follow-ups were conducted after delivery. The gut microbiome was analyzed using the 16 S rRNA V3-V4 gene regions. Functional pathway analyses of gut microbial communities in neonates were performed using Tax4fun. The average concentrations of ambient O3 and other air pollutants from pregnancy to delivery were calculated using the China High Air Pollutants (CHAP) dataset, based on the permanent residential addresses of participants. Multiple linear regression and mixed linear models were utilized to investigate the associations between prenatal O3 exposure and gut microbiome features. RESULTS: Prenatal O3 exposure did not significantly affect the gut microbial alpha diversity of mothers and neonates. However, it was found to be positively associated with the gut microbial alpha diversity in 24-month-old infants. Prenatal O3 exposure explained 13.1 % of the variation in neonatal gut microbial composition. After controlling for potential covariates, prenatal O3 exposure was associated with neonatal-specific gut microbial taxa and functional pathways. Furthermore, the mixed linear models showed that prenatal O3 exposure was negatively associated with variations of Streptococcus (p-value = 0.001, q-value = 0.005), Enterococcus (p-value = 0.001, q-value = 0.005), Escherichia-Shigella (p-value = 0.010, q-value = 0.025), and Bifidobacterium (p-value = 0.003, q-value = 0.010). CONCLUSIONS: This study is the first to examine the effects of prenatal O3 exposure on gut microbial homeostasis and variations. It demonstrates that prenatal O3 exposure is associated with variations in certain aspects of the gut microbiome. These findings provide novel insights into the dynamics and establishment of the human microbiome during the first two years of life.


Assuntos
Microbioma Gastrointestinal , Exposição Materna , Ozônio , Efeitos Tardios da Exposição Pré-Natal , Humanos , Ozônio/toxicidade , Microbioma Gastrointestinal/efeitos dos fármacos , Feminino , Gravidez , China , Estudos de Coortes , Adulto , Recém-Nascido , Exposição Materna/efeitos adversos , Efeitos Tardios da Exposição Pré-Natal/microbiologia , Poluentes Atmosféricos/toxicidade , Lactente , Masculino , Pré-Escolar , RNA Ribossômico 16S/genética , População do Leste Asiático
3.
Cell Host Microbe ; 32(7): 1192-1206.e5, 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38955186

RESUMO

The impact of gestational diabetes mellitus (GDM) on maternal or infant microbiome trajectory remains poorly understood. Utilizing large-scale longitudinal fecal samples from 264 mother-baby dyads, we present the gut microbiome trajectory of the mothers throughout pregnancy and infants during the first year of life. GDM mothers had a distinct microbiome diversity and composition during the gestation period. GDM leaves fingerprints on the infant's gut microbiome, which are confounded by delivery mode. Further, Clostridium species positively correlate with a larger head circumference at month 12 in male offspring but not females. The gut microbiome of GDM mothers with male fetuses displays depleted gut-brain modules, including acetate synthesis I and degradation and glutamate synthesis II. The gut microbiome of female infants of GDM mothers has higher histamine degradation and dopamine degradation. Together, our integrative analysis indicates that GDM affects maternal and infant gut composition, which is associated with sexually dimorphic infant head growth.


Assuntos
Diabetes Gestacional , Fezes , Microbioma Gastrointestinal , Feminino , Humanos , Diabetes Gestacional/microbiologia , Gravidez , Masculino , Lactente , Fezes/microbiologia , Cabeça/microbiologia , Adulto , Recém-Nascido , Clostridium/crescimento & desenvolvimento , Efeitos Tardios da Exposição Pré-Natal/microbiologia
4.
PLoS One ; 19(7): e0306741, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38980850

RESUMO

There has been much evidence showing the repercussions of prenatal bisphenol A (BPA) exposure with a postnatal high fat-diet (HFD) on offspring's health. However, the information on how the interaction between these two variables affects the gut microbiome is rather limited. Hence, we investigated the impact of a postnatal trans fat diet (TFD) on the gut microbiome of offspring exposed to BPA during the prenatal period in an animal model. Pregnant rats were divided into 5 mg/kg/day BPA, vehicle Tween80 (P80) or control (CTL) drinking water until delivery (N = 6 per group). Then, weaned male pups were further subdivided into three normal diet (ND) groups (CTLND, P80ND, and BPAND) and three TFD groups (CTLTFD, P80TFD, and BPATFD) (n = 6 per group). 180-250 g of faecal samples were collected on days 50 and 100 to assess the composition of the offspring's intestinal flora using next-generation sequencing. The alpha diversity indices of TFD offspring with and without BPA were markedly lower than their ND counterparts (p<0.001-p<0.05). The beta diversity, hierarchical cluster and network analyses of the offspring's microbiome demonstrated that the microbiome species of the TFD group with and without BPA were distinctly different compared to the ND group. Consistently, TFD and ND offspring pairings exhibited a higher number of significantly different species (p<0.0001-p<0.05) compared to those exposed to prenatal BPA exposure and different life stages comparisons, as shown by the multivariate parametric analysis DESeq2. Predictive functional profiling of the offspring's intestinal flora demonstrated altered expressions of genes involved in metabolic pathways. In summary, the gut flora composition of the rat offspring may be influenced by postnatal diet instead of prenatal exposure to BPA. Our data indicate the possibility of perturbed metabolic functions and epigenetic modifications, in offspring that consumed TFD, which may theoretically lead to metabolic diseases in middle or late adulthood. Further investigation is necessary to fully understand these implications.


Assuntos
Compostos Benzidrílicos , Microbioma Gastrointestinal , Fenóis , Efeitos Tardios da Exposição Pré-Natal , Animais , Microbioma Gastrointestinal/efeitos dos fármacos , Compostos Benzidrílicos/toxicidade , Fenóis/toxicidade , Feminino , Gravidez , Ratos , Efeitos Tardios da Exposição Pré-Natal/microbiologia , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Masculino , Dieta Hiperlipídica/efeitos adversos , Ratos Sprague-Dawley , Fezes/microbiologia
5.
J Affect Disord ; 362: 893-902, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39013520

RESUMO

BACKGROUND: Prenatal psychological distress and maternal inflammation can increase the risk of neurodevelopmental delay in offspring; recently, the gut microbiota has been shown to may be a potential mechanism behind this association and not fully elucidated in population study. METHODS: Seventy-two maternal-infant pairs who completed the assessments of prenatal psychological distress during the third trimester and neurodevelopment of infants at age 6-8 months of age were included in this study. The gut microbiota and its short-chain fatty acids (SCFAs) of maternal-infant were determined by 16S rRNA sequencing and liquid chromatography-mass spectrometry analysis. Inflammatory cytokines in the blood of pregnant women during the third trimester were detected by luminex liquid suspension microarrays. RESULTS: This study found that infants in the prenatal psychological distress group had poorer fine motor skills (ß = -4.396, 95 % confidence interval (CI) = -8.546, -0.246, p = 0.038), problem-solving skills (ß = -5.198, 95 % CI = -10.358, -0.038, p = 0.048) and total development (ß = -22.303, 95%CI = -41.453, -3.153, p = 0.022) compared to the control group. The study also indicated that the higher level of interleukin-1ß (IL-1ß) (ß = -1.951, 95%CI = -3.321, -0.581, p = 0.005) and interferon-inducible protein-10 (IP-10) (ß = -0.019, 95%CI = -0.034, -0.004, p = 0.015) during the third trimester, the poorer fine motor skills in infants. Also, the higher level of IL-10 (ß = -0.498, 95%CI = -0.862, -0.133, p = 0.007), IL-12p70 (ß = -0.113, 95%CI = -0.178, -0.048, p = 0.001), IL-17 A (ß = -0.817, 95%CI = -1.517, -0.118, p = 0.022), interferon-γ (ß = -0.863, 95%CI = -1.304, -0.422, p < 0.001), IP-10 (ß = -0.020, 95%CI = -0.038, -0.001, p = 0.035), and regulated upon activation normal T cell expressed and secreted (ß = -0.002, 95%CI = -0.003, -0.001, p = 0.005) during the third trimester, the poorer problem-solving skills in infants. After controlling for relevant covariates, this study found that maternal gut microbiota Roseburia mediates the relationship between prenatal psychological distress and total neurodevelopment of infants (a = 0.433, 95%CI = 0.079, 0.787, p = 0.017; b = -19.835, 95%CI = -33.877, -5.792, p = 0.006; c = 22.407, 95%CI = -43.207,-1.608, p = 0.035; indirect effect = -8.584, 95%CI = -21.227, -0.587). CONCLUSIONS: This is the first study to emphasize the role of the maternal-infant gut microbiota in prenatal psychological distress and infant neurodevelopment. Further studies are needed to explore the biological mechanisms underlying the relationship between prenatal psychological distress, maternal-infant gut microbiota, and infant neurodevelopment.


Assuntos
Desenvolvimento Infantil , Microbioma Gastrointestinal , Efeitos Tardios da Exposição Pré-Natal , Humanos , Feminino , Gravidez , Microbioma Gastrointestinal/fisiologia , Lactente , Efeitos Tardios da Exposição Pré-Natal/microbiologia , Efeitos Tardios da Exposição Pré-Natal/psicologia , Adulto , Masculino , Desenvolvimento Infantil/fisiologia , Angústia Psicológica , Citocinas/sangue , Complicações na Gravidez/microbiologia , Complicações na Gravidez/psicologia , Terceiro Trimestre da Gravidez , Transtornos do Neurodesenvolvimento/microbiologia , Estresse Psicológico/microbiologia
7.
Sci Rep ; 14(1): 16121, 2024 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-38997303

RESUMO

Microbiota imbalances are linked to inflammation and disease, as well as neurodevelopmental conditions where they may contribute to behavioral, physiological, and central nervous system dysfunction. By contrast, the role of the microbiota in Fetal Alcohol Spectrum Disorder (FASD), the group of neurodevelopmental conditions that can occur following prenatal alcohol exposure (PAE), has not received similar attention. Here we utilized a rodent model of alcohol consumption during pregnancy to characterize the impact of alcohol on the microbiota of dam-offspring dyads. Overall, bacterial diversity decreased in alcohol-consuming dams and community composition differed from that of controls in alcohol-consuming dams and their offspring. Bacterial taxa and predicted biochemical pathway composition were also altered with alcohol consumption/exposure; however, there was minimal overlap between the changes in dams and offspring. These findings illuminate the potential importance of the microbiota in the pathophysiology of FASD and support investigation into novel microbiota-based interventions.


Assuntos
Consumo de Bebidas Alcoólicas , Fezes , Efeitos Tardios da Exposição Pré-Natal , Animais , Gravidez , Feminino , Fezes/microbiologia , Consumo de Bebidas Alcoólicas/efeitos adversos , Efeitos Tardios da Exposição Pré-Natal/microbiologia , Ratos , Transtornos do Espectro Alcoólico Fetal/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Etanol/efeitos adversos , Masculino , Modelos Animais de Doenças , Microbiota/efeitos dos fármacos , Bactérias/classificação , Bactérias/efeitos dos fármacos
8.
Brain Behav Immun ; 121: 317-330, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39032541

RESUMO

The developing central nervous system is highly sensitive to nutrient changes during the perinatal period, emphasising the potential impact of alterations of maternal diet on offspring brain development and behaviour. A growing body of research implicates the gut microbiota in neurodevelopment and behaviour. Maternal overweight and obesity during the perinatal period has been linked to changes in neurodevelopment, plasticity and affective disorders in the offspring, with implications for microbial signals from the maternal gut. Here we investigate the impact of maternal high-fat diet (mHFD)-induced changes in microbial signals on offspring brain development, and neuroimmune signals, and the enduring effects on behaviour into adolescence. We first demonstrate that maternal caecal microbiota composition at term pregnancy (embryonic day 18: E18) differs significantly in response to maternal diet. Moreover, mHFD resulted in the upregulation of microbial genes in the maternal intestinal tissue linked to alterations in quinolinic acid synthesis and elevated kynurenine levels in the maternal plasma, both neuronal plasticity mediators related to glutamate metabolism. Metabolomics of mHFD embryonic brains at E18 also detected molecules linked to glutamate-glutamine cycle, including glutamic acid, glutathione disulphide, and kynurenine. During adolescence, the mHFD offspring exhibited increased locomotor activity and anxiety-like behaviour in a sex-dependent manner, along with upregulation of glutamate-related genes compared to controls. Overall, our results demonstrate that maternal exposure to high-fat diet results in microbiota changes, behavioural imprinting, altered brain metabolism, and glutamate signalling during critical developmental windows during the perinatal period.


Assuntos
Encéfalo , Dieta Hiperlipídica , Microbioma Gastrointestinal , Efeitos Tardios da Exposição Pré-Natal , Animais , Dieta Hiperlipídica/efeitos adversos , Feminino , Gravidez , Encéfalo/metabolismo , Microbioma Gastrointestinal/fisiologia , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/microbiologia , Masculino , Comportamento Animal/fisiologia , Fenômenos Fisiológicos da Nutrição Materna , Comportamento do Adolescente/fisiologia , Camundongos , Ansiedade/metabolismo , Ansiedade/microbiologia
9.
Sci Rep ; 14(1): 13762, 2024 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-38877103

RESUMO

Selective Serotonin Reuptake Inhibitor (SSRI) therapy is common among perinatal populations for the treatment of mood disorders. Medications can affect diversity and composition of the gut microbiome, which plays a key role in modulating health. While previous studies have examined the effects of antidepressant exposure on the maternal gut microbiome, whether SSRI exposure affects the offspring gut microbiome is unknown. We investigated the effects of maternal fluoxetine exposure on the gut microbiome of maternal and offspring mice during pregnancy and lactation (embryonic day 10-lactation day 21; E10-L21). Stool samples collected on E17, L11, L15, and L21 were examined using 16S rRNA sequencing. Our results suggest that maternal fluoxetine exposure may result in decreased alpha diversity of the offspring gut microbiome in early life. Furthermore, we observed several genera-specific differences in the gut microbiome based on treatment, specifically of Turicibacter, Parasutterella, and Romboutsia. These findings support our understanding of gut health, as dysbiotic development of the gut microbiome has been associated with local and systemic health problems including gastrointestinal morbidities and interrupted growth patterns in infants. Future research should pursue study in human populations and those at high risk for gut microbial dysbiosis and intestinal injury.


Assuntos
Fluoxetina , Microbioma Gastrointestinal , Lactação , RNA Ribossômico 16S , Animais , Microbioma Gastrointestinal/efeitos dos fármacos , Feminino , Gravidez , Lactação/efeitos dos fármacos , Fluoxetina/farmacologia , Fluoxetina/efeitos adversos , Camundongos , RNA Ribossômico 16S/genética , Efeitos Tardios da Exposição Pré-Natal/microbiologia , Inibidores Seletivos de Recaptação de Serotonina/efeitos adversos , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Fezes/microbiologia , Exposição Materna/efeitos adversos , Bactérias/efeitos dos fármacos , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação
10.
Environ Res ; 257: 119283, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-38830395

RESUMO

BACKGROUND: Animal and human studies indicate that exposure to air pollution and natural environments might modulate the gut microbiota, but epidemiological evidence is very scarce. OBJECTIVES: To assess the potential impact of pre- and postnatal exposure to air pollution and green spaces on infant gut microbiota assembly and trajectories during the first year of life. METHODS: MAMI ("MAternal MIcrobes") birth cohort (Valencia, Spain, N = 162) was used to study the impact of environmental exposure (acute and chronic) on infant gut microbiota during the first year of life (amplicon-based 16S rRNA sequencing). At 7 days and at 1, 6 and 12 months, residential pre- and postnatal exposure to air pollutants (NO2, black carbon -BC-, PM2.5 and O3) and green spaces indicators (NDVI and area of green spaces at 300, 500 and 1000 m buffers) were obtained. For the association between exposures and alpha diversity indicators linear regression models (cross-sectional analyses) and mixed models, including individual as a random effect (longitudinal analyses), were applied. For the differential taxon analysis, the ANCOM-BC package with a log count transformation and multiple-testing corrections were used. RESULTS: Acute exposure in the first week of life and chronic postnatal exposure to NO2 were associated with a reduction in microbial alpha diversity, while the effects of green space exposure were not evident. Acute and chronic (prenatal or postnatal) exposure to NO2 resulted in increased abundance of Haemophilus, Akkermansia, Alistipes, Eggerthella, and Tyzerella populations, while increasing green space exposure associated with increased Negativicoccus, Senegalimassilia and Anaerococcus and decreased Tyzzerella and Lachnoclostridium populations. DISCUSSION: We observed a decrease in the diversity of the gut microbiota and signs of alteration in its composition among infants exposed to higher levels of NO2. Increasing green space exposure was also associated with changes in gut microbial composition. Further research is needed to confirm these findings.


Assuntos
Poluição do Ar , Microbioma Gastrointestinal , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Feminino , Lactente , Espanha , Poluição do Ar/efeitos adversos , Recém-Nascido , Poluentes Atmosféricos/análise , Poluentes Atmosféricos/toxicidade , Coorte de Nascimento , Masculino , Exposição Ambiental/efeitos adversos , Gravidez , Estudos de Coortes , Exposição Materna/efeitos adversos , Efeitos Tardios da Exposição Pré-Natal/microbiologia
11.
Environ Res ; 259: 119442, 2024 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-38901810

RESUMO

Perfluorooctane sulfonate (PFOS) exposure is associated with harmful hepatic outcomes. Growing evidence indicates that crosstalk between the gut microbiome, immune system, and liver plays a vital role in the pathogenesis of liver diseases. However, the underlying mechanism is not fully understood. In the present study, we aimed to investigate the effects of PFOS exposure during pregnancy and lactation on hepatic inflammation in rat offspring. Features of hepatic inflammation and increased levels of aspartate-amino transferase (AST) were found in pups on postnatal day 28 (PND28) in PFOS-exposed groups. Gut microbiota analysis identified Chitinophaga, Ralstonia, and Alloprevotella as the key genera in distinguishing the PFOS-exposed group from the control group. Metabolic and transcriptomic analyses found that PFOS exposure resulted in 48 differentially expressed metabolites (DEMs) in the serum, 62 DEMs in the liver, and 289 differentially expressed genes (DEGs) in the liver of PND28 pups. The immune response is significantly enriched in PFOS-exposed liver on PND28; multi-omics analysis indicated that PFOS might lead to immune response perturbation by disturbing the metabolic profiling in the liver. The changed gut microbiota was significantly related to the serum level of the liver function index. Specifically, Alloprevotella, Chitinophage, Ruminococcus, and Allobaculum were significantly associated with the metabolic abundance changes of 4-Hydroxydebrisoquine, L-Norvaline, and Eremopetasinorol, and the gene expression changes of Acat211, Msmol, Idi1, Sqle, and Gadd45b in the liver. These findings suggest that early-life PFOS exposure may be associated with adverse hepatic inflammation in young offspring via disruption of the gut-liver crosstalk, which may provide mechanistic clues for clarifying the hepatotoxicity in offspring associated with perinatal PFOS exposure.


Assuntos
Ácidos Alcanossulfônicos , Fluorocarbonos , Microbioma Gastrointestinal , Fígado , Efeitos Tardios da Exposição Pré-Natal , Animais , Fluorocarbonos/toxicidade , Ácidos Alcanossulfônicos/toxicidade , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Gravidez , Fígado/efeitos dos fármacos , Fígado/metabolismo , Ratos , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/microbiologia , Masculino , Poluentes Ambientais/toxicidade , Ratos Sprague-Dawley , Inflamação/induzido quimicamente , Exposição Materna/efeitos adversos
12.
Gut Microbes ; 16(1): 2292224, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38108125

RESUMO

There has been a rapid increase in neonates born with a history of prenatal opioid exposure. How prenatal opioid exposure affects pain sensitivity in offspring is of interest, as this may perpetuate the opioid epidemic. While few studies have reported hypersensitivity to thermal pain, potential mechanisms have not been described. This study posits that alterations in the gut microbiome may underly hypersensitivity to pain in prenatally methadone-exposed 3-week-old male offspring, which were generated using a mouse model of prenatal methadone exposure. Fecal samples collected from dams and their offspring were subjected to 16s rRNA sequencing. Thermal and mechanical pain were assessed using the tail flick and Von Frey assays. Transcriptomic changes in whole brain samples of opioid or saline-exposed offspring were investigated using RNA-sequencing, and midbrain sections from these animals were subjected to qPCR profiling of genes related to neuropathic and inflammatory pain pathways. Prenatal methadone exposure increased sensitivity to thermal and mechanical pain and elevated serum levels of IL-17a. Taxonomical analysis revealed that prenatal methadone exposure resulted in significant alterations in fecal gut microbiota composition, including depletion of Lactobacillus, Bifidobacterium, and Lachnospiracea sp and increased relative abundance of Akkermansia, Clostridium sensu stricto 1, and Lachnoclostridium. Supplementation of the probiotic VSL#3 in dams rescued hypersensitivity to thermal and mechanical pain in prenatally methadone-exposed offspring. Similarly, cross-fostering prenatally methadone-exposed offspring to control dams also attenuated hypersensitivity to thermal pain in opioid-exposed offspring. Modulation of the maternal and neonatal gut microbiome with probiotics resulted in transcriptional changes in genes related to neuropathic and immune-related signaling in whole brain and midbrain samples of prenatally methadone-exposed offspring. Together, our work provides compelling evidence of the gut-brain-axis in mediating pain sensitivity in prenatally opioid-exposed offspring.


Assuntos
Microbioma Gastrointestinal , Efeitos Tardios da Exposição Pré-Natal , Humanos , Gravidez , Animais , Feminino , Masculino , Analgésicos Opioides/efeitos adversos , Microbioma Gastrointestinal/genética , Disbiose/induzido quimicamente , Disbiose/microbiologia , RNA Ribossômico 16S/genética , Efeitos Tardios da Exposição Pré-Natal/microbiologia , Metadona , Dor
13.
Microbiome ; 11(1): 165, 2023 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-37501202

RESUMO

BACKGROUND: During development, elevated levels of maternal glucocorticoids (GCs) can have detrimental effects on offspring morphology, cognition, and behavior as well as physiology and metabolism. Depending on the timing of exposure, such effects may vary in strength or even reverse in direction, may alleviate with age, or may concern more stable and long-term programming of phenotypic traits. Maternal effects on gut bacterial diversity, composition, and function, and the persistence of such effects into adulthood of long-lived model species in the natural habitats remain underexplored. RESULTS: In a cross-sectional sample of infant, juvenile, and adult Assamese macaques, the timing of exposure to elevated maternal GCs during ontogeny was associated with the gut bacterial community of the offspring. Specifically, naturally varying maternal GC levels during early but not late gestation or lactation were associated with reduced bacterial richness. The overall effect of maternal GCs during early gestation on the gut bacterial composition and function exacerbated with offspring age and was 10 times stronger than the effect associated with exposure during late prenatal or postnatal periods. Instead, variation in maternal GCs during the late prenatal or postnatal period had less pronounced or less stable statistical effects and therefore a weaker effect on the entire bacterial community composition, particularly in adult individuals. Finally, higher early prenatal GCs were associated with an increase in the relative abundance of several potential pro-inflammatory bacteria and a decrease in the abundance of Bifidobacterium and other anti-inflammatory taxa, an effect that exacerbated with age. CONCLUSIONS: In primates, the gut microbiota can be shaped by developmental effects with strong timing effects on plasticity and potentially detrimental consequences for adult health. Together with results on other macaque species, this study suggests potential detrimental developmental effects similar to rapid inflammaging, suggesting that prenatal exposure to high maternal GC concentrations is a common cause underlying both phenomena. Our findings await confirmation by metagenomic functional and causal analyses and by longitudinal studies of long-lived, ecologically flexible primates in their natural habitat, including developmental effects that originate before birth. Video Abstract.


Assuntos
Microbioma Gastrointestinal , Efeitos Tardios da Exposição Pré-Natal , Feminino , Animais , Gravidez , Humanos , Glucocorticoides , Microbioma Gastrointestinal/fisiologia , Estudos Transversais , Primatas , Bactérias/genética , Efeitos Tardios da Exposição Pré-Natal/microbiologia
15.
Microbiome ; 10(1): 122, 2022 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-35941695

RESUMO

BACKGROUND: Hyperglycaemia in pregnancy (HIP) is a common metabolic disorder that not only poses risks to maternal health but also associates with an increased risk of diabetes among offspring. Vertical transmission of microbiota may influence the offspring microbiome and subsequent glucose metabolism. However, the mechanism by which maternal gut microbiota may influence glucose metabolism of the offspring remains unclear and whether intervening microbiota vertical transmission could be used as a strategy to prevent diabetes in the offspring of mothers with HIP has not been investigated. So we blocked vertical transmission to investigate its effect on glucose metabolism in the offspring. RESULTS: We established a murine HIP model with a high-fat diet (HFD) and investigated the importance of vertical transmission of gut microbiota on the glucose metabolism of offspring via birth and nursing by blocking these events through caesarean section (C-section) and cross-fostering. After weaning, all offspring were fed a normal diet. Based on multi-omics analysis, biochemical and transcriptional assays, we found that the glucometabolic deficits in the mothers were subsequently 'transmitted' to the offspring. Meanwhile, the partial change in mothers' gut microbial community induced by HIP could be transmitted to offspring, supported by the closed clustering of the microbial structure and composition between the offspring and their mothers. Further study showed that the microbiota vertical transmission was blocked by C-section and cross-fostering, which resulted in improved insulin sensitivity and islet function of the offspring of the mothers with HIP. These effects were correlated with changes in the relative abundances of specific bacteria and their metabolites, such as increased relative abundances of Bifidobacterium and short-chain fatty acids. In particular, gut microbial communities of offspring were closely related to those of their foster mothers but not their biological mothers, and the effect of cross-fostering on the offspring's gut microbiota was more profound than that of C-section. CONCLUSION: Our study demonstrates that the gut microbiota transmitted via birth and nursing are important contributors to the glucose metabolism phenotype in offspring. Video Abstract.


Assuntos
Diabetes Mellitus , Microbioma Gastrointestinal , Hiperglicemia , Efeitos Tardios da Exposição Pré-Natal , Animais , Cesárea , Dieta Hiperlipídica/efeitos adversos , Feminino , Glucose , Humanos , Camundongos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/microbiologia
16.
Brain Behav Immun ; 103: 186-201, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35427758

RESUMO

Group B Streptococcus (GBS) remains a major neonatal life-threatening pathogen. We initially identified glyceraldehyde-3-phosphate dehydrogenase (GAPDH) as a promising vaccine candidate against GBS. Since GAPDH is highly conserved, we investigate whether GBS GAPDH maternal vaccination interferes with the intestinal colonization of the offspring and the development of its mucosal immune system and central nervous system. An altered gut microbiome with increased Proteobacteria is observed in pups born from vaccinated dams during early life. These pups present decreased relative expression of IL-1ß, IL-17A, RegIIIγ and MUC2 in the distal colon. They also display increased CD11b, F4/80 and MHC class II expression on microglia in early life and marked reduction of Ly6C+ cells and neutrophils. Importantly, male mice born from vaccinated mothers present behavioral abnormalities during adulthood, including decreased exploratory behavior, a subtle anxious-like phenotype and global alterations in spatial learning and memory strategies, and higher sensitivity to a stressful stimulus. Our study highlights the danger of using ubiquitous antigens in maternal human vaccines against neonatal pathogens.


Assuntos
Disbiose , Microbioma Gastrointestinal , Efeitos Tardios da Exposição Pré-Natal , Vacinas Estreptocócicas , Animais , Disbiose/induzido quimicamente , Feminino , Gliceraldeído-3-Fosfato Desidrogenases/genética , Masculino , Camundongos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/microbiologia , Vacinas Estreptocócicas/efeitos adversos , Streptococcus agalactiae , Vacinação
17.
Immunity ; 55(1): 145-158.e7, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-34879222

RESUMO

Children with autism spectrum disorders often display dysregulated immune responses and related gastrointestinal symptoms. However, the underlying mechanisms leading to the development of both phenotypes have not been elucidated. Here, we show that mouse offspring exhibiting autism-like phenotypes due to prenatal exposure to maternal inflammation were more susceptible to developing intestinal inflammation following challenges later in life. In contrast to its prenatal role in neurodevelopmental phenotypes, interleukin-17A (IL-17A) generated immune-primed phenotypes in offspring through changes in the maternal gut microbiota that led to postnatal alterations in the chromatin landscape of naive CD4+ T cells. The transfer of stool samples from pregnant mice with enhanced IL-17A responses into germ-free dams produced immune-primed phenotypes in offspring. Our study provides mechanistic insights into why children exposed to heightened inflammation in the womb might have an increased risk of developing inflammatory diseases in addition to neurodevelopmental disorders.


Assuntos
Transtorno do Espectro Autista/imunologia , Linfócitos T CD4-Positivos/imunologia , Cromatina/metabolismo , Microbioma Gastrointestinal/imunologia , Inflamação/imunologia , Interleucina-17/metabolismo , Intestinos/imunologia , Transtornos do Neurodesenvolvimento/imunologia , Efeitos Tardios da Exposição Pré-Natal/imunologia , Animais , Transtorno do Espectro Autista/microbiologia , Criança , Modelos Animais de Doenças , Transplante de Microbiota Fecal , Feminino , Humanos , Imunização , Inflamação/microbiologia , Camundongos , Transtornos do Neurodesenvolvimento/microbiologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/microbiologia
18.
Nat Commun ; 12(1): 6289, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34725359

RESUMO

Newborns are colonized by maternal microbiota that is essential for offspring health and development. The composition of these pioneer communities exhibits individual differences, but the importance of this early-life heterogeneity to health outcomes is not understood. Here we validate a human microbiota-associated model in which fetal mice are cesarean delivered and gavaged with defined human vaginal microbial communities. This model replicates the inoculation that occurs during vaginal birth and reveals lasting effects on offspring metabolism, immunity, and the brain in a community-specific manner. This microbial effect is amplified by prior gestation in a maternal obesogenic or vaginal dysbiotic environment where placental and fetal ileum development are altered, and an augmented immune response increases rates of offspring mortality. Collectively, we describe a translationally relevant model to examine the defined role of specific human microbial communities on offspring health outcomes, and demonstrate that the prenatal environment dramatically shapes the postnatal response to inoculation.


Assuntos
Microbioma Gastrointestinal , Relações Materno-Fetais/fisiologia , Microbiota , Parto/fisiologia , Efeitos Tardios da Exposição Pré-Natal/microbiologia , Vagina/microbiologia , Animais , Cesárea/métodos , Feminino , Humanos , Recém-Nascido , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Efeitos Tardios da Exposição Pré-Natal/imunologia , Efeitos Tardios da Exposição Pré-Natal/patologia , Transcriptoma
19.
FASEB J ; 35(11): e21971, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34653284

RESUMO

It has been recently recognized that prenatal androgen exposure is involved in the development of polycystic ovary syndrome (PCOS) in adulthood. In addition, the gut microbiome in adult patients and rodents with PCOS differs from that of healthy individuals. Moreover, recent studies have suggested that the gut microbiome may play a causative role in the pathogenesis of PCOS. We wondered whether prenatal androgen exposure induces gut microbial dysbiosis early in life and is associated with the development of PCOS in later life. To test this hypothesis, we studied the development of PCOS-like phenotypes in prenatally androgenized (PNA) female mice and compared the gut microbiome of PNA and control offspring from 4 to 16 weeks of age. PNA offspring showed a reproductive phenotype from 6 weeks and a metabolic phenotype from 12 weeks of age. The α-diversity of the gut microbiome of the PNA group was higher at 8 weeks and lower at 12 and 16 weeks of age, and the ß-diversity differed from control at 8 weeks. However, a significant difference in the composition of gut microbiome between the PNA and control groups was already apparent at 4 weeks. Allobaculum and Roseburia were less abundant in PNA offspring, and may therefore be targets for future interventional studies. In conclusion, abnormalities in the gut microbiome appear as early as or even before PCOS-like phenotypes develop in PNA mice. Thus, the gut microbiome in early life is a potential target for the prevention of PCOS in later life.


Assuntos
Androgênios/metabolismo , Microbioma Gastrointestinal , Síndrome do Ovário Policístico , Efeitos Tardios da Exposição Pré-Natal/microbiologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Síndrome do Ovário Policístico/metabolismo , Síndrome do Ovário Policístico/microbiologia , Gravidez
20.
Sci Rep ; 11(1): 18971, 2021 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-34556804

RESUMO

Intra-amniotic infection (IAI) is a major cause of preterm birth with a poor perinatal prognosis. We aimed to determine whether analyzing vaginal microbiota can evaluate the risk of chorioamnionitis (CAM) in preterm labor cases. Vaginal discharge samples were collected from 83 pregnant women admitted for preterm labor. Based on Blanc's classification, the participants were divided into CAM (stage ≥ II; n = 46) and non-CAM (stage ≤ I; n = 37) groups. The 16S rDNA amplicons (V1-V2) from vaginal samples were sequenced and analyzed. Using a random forest algorithm, the bacterial species associated with CAM were identified, and a predictive CAM (PCAM) scoring method was developed. The α diversity was significantly higher in the CAM than in the non-CAM group (P < 0.001). The area under the curve was 0.849 (95% confidence interval 0.765-0.934) using the PCAM score. Among patients at < 35 weeks of gestation, the PCAM group (n = 22) had a significantly shorter extended gestational period than the non-PCAM group (n = 25; P = 0.022). Multivariate analysis revealed a significant difference in the frequency of developmental disorders in 3-year-old infants (PCAM, 28%, non-PCAM, 4%; P = 0.022). Analyzing vaginal microbiota can evaluate the risk of IAI. Future studies should establish appropriate interventions for IAI high-risk patients to improve perinatal prognosis.


Assuntos
Corioamnionite/epidemiologia , Deficiências do Desenvolvimento/epidemiologia , Microbiota/imunologia , Efeitos Tardios da Exposição Pré-Natal/epidemiologia , Vagina/microbiologia , Adulto , Pré-Escolar , Corioamnionite/imunologia , Corioamnionite/microbiologia , DNA Bacteriano/isolamento & purificação , Deficiências do Desenvolvimento/imunologia , Deficiências do Desenvolvimento/microbiologia , Feminino , Seguimentos , Humanos , Lactente , Recém-Nascido , Masculino , Trabalho de Parto Prematuro/imunologia , Trabalho de Parto Prematuro/microbiologia , Projetos Piloto , Gravidez , Efeitos Tardios da Exposição Pré-Natal/imunologia , Efeitos Tardios da Exposição Pré-Natal/microbiologia , RNA Ribossômico 16S/genética , Medição de Risco/métodos , Vagina/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...