Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Clin Microbiol Rev ; 36(4): e0001523, 2023 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-37909789

RESUMO

MicroRNAs (miRNAs) are conserved, short, non-coding RNAs that play a crucial role in the post-transcriptional regulation of gene expression. They have been implicated in the pathogenesis of cancer and neurological, cardiovascular, and autoimmune diseases. Several recent studies have suggested that miRNAs are key players in regulating the differentiation, maturation, and activation of immune cells, thereby influencing the host immune response to infection. The resultant upregulation or downregulation of miRNAs from infection influences the protein expression of genes responsible for the immune response and can determine the risk of disease progression. Recently, miRNAs have been explored as diagnostic biomarkers and therapeutic targets in various infectious diseases. This review summarizes our current understanding of the role of miRNAs during viral, fungal, bacterial, and parasitic infections from a clinical perspective, including critical functional mechanisms and implications for their potential use as biomarkers and therapeutic targets.


Assuntos
Doenças Transmissíveis , MicroRNAs , Doenças Parasitárias , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Regulação da Expressão Gênica , Biomarcadores , Doenças Parasitárias/diagnóstico , Doenças Transmissíveis/diagnóstico , Doenças Transmissíveis/genética , Doenças Transmissíveis/terapia
2.
Infect Immun ; 89(5)2021 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-33526558

RESUMO

We aimed to determine whether T cell-specific STAT3 deletion influences the immune response to Aspergillus in the immunosuppressed context in CD4 Stat3-/- mice. Immunosuppressed and nonimmunosuppressed CD4 Stat3-/- mice and littermate Stat3flox/flox (Stat3fl/fl) mice were infected with Aspergillus fumigatus in an aerosol chamber, and the weight, activity, appearance, and respiratory rate of the mice were monitored daily for 21 days to evaluate their survival. Aspergillus infection was confirmed by lung fungal culture counts, histology, and a galactomannan test. Cytokines were measured at 3 days postinfection in bronchoalveolar lavage (BAL) fluid and serum. Immunosuppressed CD4 Stat3-/- mice began succumbing to infection by day 4, and by day 7, only 30% of mice survived. Immunosuppressed Stat3fl/fl mice started to succumb to the disease on day 5, and 40% of mice remained by day 7. The nonimmunosuppressed control Stat3fl/fl and CD4 Stat3-/- mice maintained their weight over the study period, without any evidence of infection by A. fumigatus by histology. In the BAL fluid, tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), interferon gamma (IFN-γ), IL-17A, and IL-22 levels were elevated in Stat3fl/fl immunosuppressed mice compared to immunosuppressed CD4 Stat3-/- mice at 3 days postinfection. STAT3 in CD4+ T cells modulates the production of cytokines in the IL-17 pathway in immunosuppressed mice. However, it has no meaningful effect on the clearance of Aspergillus or the concomitant increase in susceptibility to Aspergillus infection.


Assuntos
Aspergilose/etiologia , Aspergilose/metabolismo , Aspergillus fumigatus/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Interações Hospedeiro-Patógeno , Fator de Transcrição STAT3/metabolismo , Animais , Aspergilose/patologia , Líquido da Lavagem Broncoalveolar , Citocinas/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Interações Hospedeiro-Patógeno/imunologia , Mediadores da Inflamação , Camundongos , Camundongos Knockout , Transdução de Sinais
3.
J Physiol ; 597(12): 3029-3051, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31081119

RESUMO

KEY POINTS: Maternal obesity has been associated with shifts in intestinal microbiota, which may contribute to impaired barrier function Impaired barrier function may expose the placenta and fetus to pro-inflammatory mediators We investigated the impacts of diet-induced obesity in mice on maternal and fetal intestinal structure and placental vascularization Diet-induced obesity decreased maternal intestinal short chain fatty acids and their receptors, impaired gut barrier integrity and was associated with fetal intestinal inflammation. Placenta from obese mothers showed blood vessel immaturity, hypoxia, increased transcript levels of inflammation, autophagy and altered levels of endoplasmic reticulum stress markers. These data suggest that maternal intestinal changes probably contribute to adverse placental adaptations and also impart an increased risk of obesity in the offspring via alterations in fetal gut development. ABSTRACT: Shifts in maternal intestinal microbiota have been implicated in metabolic adaptations to pregnancy. In the present study, we generated cohorts of female C57BL/6J mice fed a control (17% kcal fat, n = 10-14) or a high-fat diet (HFD 60% kcal from fat, n = 10-14; ad libitum) aiming to investigate the impact on the maternal gut microbiota, intestinal inflammation and gut barrier integrity, placental inflammation and fetal intestinal development at embryonic day 18.5. HFD was associated with decreased relative abundances of short-chain fatty acid (SCFA) producing genera during pregnancy. These diet-induced shifts paralleled decreased maternal intestinal mRNA levels of SCFA receptor Gpr41, modestly decreased cecal butyrate, and altered mRNA levels of inflammatory cytokines and immune cell markers in the maternal intestine. Maternal HFD resulted in impaired gut barrier integrity, with corresponding increases in circulating maternal levels of lipopolysaccharide (LPS) and tumour necrosis factor. Placentas from HFD dams demonstrated blood vessel immaturity and hypoxia; decreased free carnitine, acylcarnitine derivatives and trimethylamine-N-oxide; and altered mRNA levels of inflammation, autophagy, and ER stress markers. HFD exposed fetuses had increased activation of nuclear factor-kappa B and inhibition of the unfolded protein response in the developing intestine. Taken together, these data suggest that HFD intake prior to and during pregnancy shifts the composition of the maternal gut microbiota and impairs gut barrier integrity, resulting in increased maternal circulating LPS, which may ultimate contribute to changes in placental vascularization and fetal gut development.


Assuntos
Dieta Hiperlipídica , Microbioma Gastrointestinal , Hipóxia , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Obesidade , Placenta/irrigação sanguínea , Animais , Feminino , Desenvolvimento Fetal , Feto , Hipóxia/metabolismo , Hipóxia/microbiologia , Hipóxia/fisiopatologia , Mucosa Intestinal/microbiologia , Lipopolissacarídeos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Obesidade/microbiologia , Obesidade/fisiopatologia , Placenta/metabolismo , Gravidez
4.
Transpl Infect Dis ; 21(3): e13060, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30753747

RESUMO

We studied cytokine profiles in BAL of LTRs with Aspergillus spp colonization who did not progress to IPA in the absence of antifungal prophylaxis. This was a retrospective, single center case-control study. BAL samples were analyzed for cytokines. Patients with Aspergillus spp in BAL who did not receive prophylaxis and did not develop IPA were compared to LTRs with Aspergillus spp that received prophylaxis, LTRs with IPA and controls. Twenty-one patients with Aspergillus colonization who did not develop IPA, seven patients with suspected IPA who received prophylaxis, 4 IPA and 19 controls were included. IPA group had significantly higher levels (median [IQR]) of MIP-1 beta compared to the Suspected IPA group (5 vs 5 P: 0.03). The Suspected IPA group had significantly higher levels of IL-12 (11.38 vs 1 P: 0.0001), IL-1 RA (86.11 vs 23.98 P: 0.0118), IP-10 (22.47 vs 0.86 P: 0.0151), HGF (40.92 vs 16.82 P: 0.0055), and MIG (169.62 vs 5 P: 0.0005) than Colonization group. We have identified a unique cytokine signature in patients with Aspergillus colonization that do not develop IPA. Our study forms basis for a larger study to use these cytokines profile to identify patients at a lower risk of developing IPA.


Assuntos
Citocinas/imunologia , Aspergilose Pulmonar Invasiva/imunologia , Transplante de Pulmão/efeitos adversos , Transplantados , Antifúngicos/uso terapêutico , Aspergillus , Lavagem Broncoalveolar , Citocinas/genética , Feminino , Humanos , Aspergilose Pulmonar Invasiva/tratamento farmacológico , Pulmão/imunologia , Pulmão/microbiologia , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Fatores de Risco , Transcriptoma
5.
Biol Reprod ; 98(5): 664-682, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29351580

RESUMO

Reproductive abnormalities are included as health complications in offspring exposed to poor prenatal nutrition. We have previously shown in a rodent model that offspring born to nutrient restriction during pregnancy are born small, enter puberty early, and display characteristics of early ovarian aging as adults. The present study investigated whether key proteins involved in follicle recruitment and growth mediate ovarian follicle loss. Pregnant rats were randomized to a standard diet throughout pregnancy and lactation (CON), or a calorie-restricted (50% of control) diet (UN) during pregnancy. Offspring reproductive phenotype was investigated at postnatal days 4, 27, and 65. Maternal UN resulted in young adult (P65) irregular estrous cyclicity due to persistent estrus, a significant loss of antral follicles, corpora lutea, and an increase in atretic follicles. This decrease in growing follicles in UN offspring appears to be due to increased apoptosis as seen by immunopositive staining of pro-apoptotic factor CASP3 (caspase 3) in ovaries of young adult offspring. UN prepubertal offspring had reduced expression levels of Fshr in antral follicles, which may contribute to a decrease in PI3K/AKT activation evident as a decrease in pAKT immunolocalization in prepubertal antral follicles. Moreover, neonatal ovaries of UN offspring show decreased levels of immunopositive staining for AMHR2 (anti-mullerian hormone receptor 2). Collectively, these data demonstrate that maternal UN during pregnancy impacts ovarian function in offspring as early as P65 and provides a model for understanding the mechanisms driving early life UN-induced follicle loss and reproductive dysfunction.


Assuntos
Restrição Calórica , Ciclo Estral/metabolismo , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , Folículo Ovariano/metabolismo , Ovário/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Reprodução/fisiologia , Animais , Apoptose/fisiologia , Caspase 3/metabolismo , Feminino , Hormônio Foliculoestimulante/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Gravidez , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Receptores do FSH/metabolismo , Receptores de Peptídeos/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais/fisiologia
6.
Biol Reprod ; 94(4): 94, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26962114

RESUMO

Maternal obesity predisposes offspring to metabolic and reproductive dysfunction. We have shown previously that female rat offspring born to mothers fed a high-fat (HF) diet throughout pregnancy and lactation enter puberty early and display aberrant reproductive cyclicity. The mechanisms driving this reproductive phenotype are currently unknown thus we investigated whether changes in ovarian function were involved. Wistar rats were mated and randomized to: dams fed a control diet (CON) or dams fed a HF diet from conception until the end of lactation (HF). Ovaries were collected from fetuses at Embryonic Day (E) 20, and neonatal ovaries at Day 4 (P4), prepubertal ovaries at P27 and adult ovaries at P120. In a subset of offspring, the effects of a HF diet fed postweaning were evaluated. The present study shows that fetuses of mothers fed a HF diet had significantly fewer oocytes at E20, and in neonates, have reduced AMH signaling that may facilitate an increased number of assembled primordial follicles. Both prepubertally and in adulthood, ovaries show increased follicular atresia. As adults, offspring have reduced FSH responsiveness, low expression levels of estrogen receptor alpha (Eralpha), the oocyte-secreted factor, Gdf9, oocyte-specific RNA binding protein, Dazl, and high expression levels of the granulosa-cell derived factor, AMH, in antral follicles. Together, these data suggest that ovarian compromise in offspring born to HF-fed mothers may arise from changes already observable in the fetus and neonate and in the long term, associated with increased follicular atresia through adulthood.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Doenças Fetais/etiologia , Folículo Ovariano/crescimento & desenvolvimento , Efeitos Tardios da Exposição Pré-Natal/etiologia , Animais , Animais Recém-Nascidos , Feminino , Doenças Fetais/patologia , Oócitos , Folículo Ovariano/patologia , Fenótipo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/patologia , Distribuição Aleatória , Ratos , Ratos Wistar
7.
Biol Reprod ; 92(4): 110, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25810471

RESUMO

Maternal nutritional restriction has been shown to induce impairments in a number of organ systems including the ovary. We have previously shown that maternal undernutrition induces fetal growth restriction and low birth weight, and results in an offspring ovarian phenotype characteristic of premature ovarian aging with reduced ovarian reserve. In the present study, we set out to investigate the underlying mechanisms that lead offspring of undernourished mothers to premature ovarian aging. Pregnant dams were randomized to 1) a standard diet throughout pregnancy and lactation (control), 2) a calorie-restricted (50% of control) diet during pregnancy, 3) a calorie-restricted (50% of control) diet during pregnancy and lactation, or 4) a calorie-restricted (50% of control) diet during lactation alone. The present study shows that early life undernutrition-induced reduction of adult ovarian follicles may be mediated by increased ovarian endoplasmic reticulum stress in a manner that increased follicular apoptosis but not autophagy. These changes were associated with a loss of ovarian vessel density and are consistent with an accelerated ovarian aging phenotype. Whether these changes are mediated specifically by a reduction in the local antioxidant environment is unclear, although our data suggest the possibility that ovarian melatonin may play a part in early life nutritional undernutrition and impaired offspring folliculogenesis.


Assuntos
Apoptose , Estresse do Retículo Endoplasmático , Desnutrição/fisiopatologia , Neovascularização Fisiológica , Ovário/irrigação sanguínea , Animais , Antioxidantes/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia/fisiologia , Proteína Beclina-1 , Restrição Calórica , Feminino , Fenômenos Fisiológicos da Nutrição Materna , Folículo Ovariano/metabolismo , Folículo Ovariano/fisiologia , Ovário/crescimento & desenvolvimento , Gravidez , Ratos , Ratos Wistar , Fluxo Sanguíneo Regional , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
8.
Pediatr Res ; 77(1-2): 196-204, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25314580

RESUMO

Chronic disease risk is inextricably linked to our early-life environment, where maternal, fetal, and childhood factors predict disease risk later in life. Currently, maternal obesity is a key predictor of childhood obesity and metabolic complications in adulthood. Although the mechanisms are unclear, new and emerging evidence points to our microbiome, where the bacterial composition of the gut modulates the weight gain and altered metabolism that drives obesity. Over the course of pregnancy, maternal bacterial load increases, and gut bacterial diversity changes and is influenced by pre-pregnancy- and pregnancy-related obesity. Alterations in the bacterial composition of the mother have been shown to affect the development and function of the gastrointestinal tract of her offspring. How these microbial shifts influence the maternal-fetal-infant relationship is a topic of hot debate. This paper will review the evidence linking nutrition, maternal obesity, the maternal gut microbiome, and fetal gut development, bringing together clinical observations in humans and experimental data from targeted animal models.


Assuntos
Trato Gastrointestinal/microbiologia , Fenômenos Fisiológicos da Nutrição Materna , Troca Materno-Fetal , Microbiota , Obesidade/complicações , Efeitos Tardios da Exposição Pré-Natal/microbiologia , Feminino , Trato Gastrointestinal/crescimento & desenvolvimento , Humanos , Obesidade/microbiologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/imunologia
9.
Sci Rep ; 12(1): 4581, 2022 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-35301416

RESUMO

The heterogeneity of clinical responses to antifungals in aspergillosis is partially understood. We hypothesized that besides direct antifungal effects, these discrepancies may be related to different immunomodulatory profiles. Human THP-1 monocytes were coincubated in vitro with Aspergillus fumigatus and variable concentrations of voriconazole (0.5, 1 and 2 mg/l), caspofungin (1 and 2 mg/l), amphotericin B deoxycholate (0.25, 0.5 and 1 mg/l) and liposomal amphotericin B (1, 2 and 3 mg/l). After 6 h of coincubation, total cellular RNA was extracted, converted into cDNA, and transcription of 84 genes involved in antifungal immunity was measured through RT-qPCR. The presence of A. fumigatus was the main driver of the global immune-related transcriptomic response. After Aspergillus infection, thirty genes were upregulated, while 19 genes were downregulated. Discrepancies across antifungals were also evident; voriconazole-containing conditions showed similar reaction to natural infection, while the use of liposomal Amphotericin B significantly decreased the inflammatory response. Chemokines (notably CCL20 and CXCL2) and pro-inflammatory cytokines (IL1A, IL1B, IL23, G-CSF) exhibited the most pronounced differences across antifungals. Pattern recognition receptors and adaptor protein transcription were minimally affected. Protein-protein-interaction network analysis showed that IL23A played a dominant role in upregulated genes. Pathway enrichment analysis indicated that cytokine-cytokine receptor integration, TNF signaling pathways and Toll-like receptor pathways were highly involved. This exploratory study confirms the heterogeneous immunomodulatory role of antifungals. Overall, voriconazole appears to maintain an early pro-inflammatory response seen in natural infection. Assessment of immunomodulatory response with clinical response may provide a better rationale for differences observed across antifungals.


Assuntos
Antifúngicos , Aspergilose , Antifúngicos/farmacologia , Antifúngicos/uso terapêutico , Aspergilose/tratamento farmacológico , Aspergillus fumigatus , Humanos , Testes de Sensibilidade Microbiana , Monócitos , Transcriptoma , Voriconazol/farmacologia
10.
Sci Rep ; 10(1): 9399, 2020 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-32523064

RESUMO

The placenta is a metabolically active interfacial organ that plays crucial roles in fetal nutrient delivery, gas exchange and waste removal reflecting dynamic maternal and fetal interactions during gestation. There is growing evidence that the sex of the placenta influences fetal responses to external stimuli in utero, such as changes in maternal nutrition and exposure to environmental stressors. However, the exact biochemical mechanisms associated with sex-specific metabolic adaptations during pregnancy and its link to placental function and fetal development remain poorly understood. Herein, multisegment injection-capillary electrophoresis-mass spectrometry is used as a high throughput metabolomics platform to characterize lyophilized placental tissue (~2 mg dried weight) from C57BL/6J mice fed a standardized diet. Over 130 authentic metabolites were consistently measured from placental extracts when using a nontargeted metabolomics workflow with stringent quality control and robust batch correction. Our work revealed distinct metabolic phenotype differences that exist between male (n = 14) and female (n = 14) placentae collected at embryonic day E18.5. Intracellular metabolites associated with fatty acid oxidation and purine degradation were found to be elevated in females as compared to male placentae (p < 0.05, effect size >0.40), including uric acid, valerylcarnitine, hexanoylcarnitine, and 3-hydroxyhexanolycarnitine. This murine model sheds new insights into sex-specific differences in placental mitochondrial function and protective mechanisms against deleterious oxidative stress that may impact fetal growth and birth outcomes later in life.


Assuntos
Desenvolvimento Fetal/fisiologia , Feto/metabolismo , Metaboloma/fisiologia , Placenta/metabolismo , Animais , Peso Corporal/fisiologia , Ácidos Graxos/metabolismo , Feminino , Masculino , Metabolômica/métodos , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Purinas/metabolismo , Caracteres Sexuais
11.
J Heart Lung Transplant ; 39(11): 1228-1237, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32771440

RESUMO

BACKGROUND: MicroRNAs (miRNAs) are small non-coding RNAs of ∼22 nucleotides that play a crucial role in post-transcriptional regulation of gene expression. Dysregulation of miRNA expression has been shown during microbial infections. We sought to identify miRNAs that distinguish invasive aspergillosis (IA) from non-IA in lung transplant recipients (LTRs). METHODS: We used NanoString nCounter Human miRNA, version 3, panel to measure miRNAs in bronchoalveolar lavage (BAL) samples from LTRs with Aspergillus colonization (ASP group) (n = 10), those with Aspergillus colonization and chronic lung allograft dysfunction (CLAD) (ASPCLAD group) (n = 7), those with IA without CLAD (IA group) (n = 10), those who developed IA with CLAD (IACLAD group) (n = 9), and control patients (controls) (n = 9). The miRNA profile was compared using the permutation test of 100,000 trials for each of the comparisons. We used mirDIP to obtain their gene targets and pathDIP to determine the pathway enrichment. RESULTS: We performed pairwise comparisons between patient groups to identify differentially expressed miRNAs. A total of 5 miRNAs were found to be specific to IA, including 4 (miR-145-5p, miR-424-5p, miR-99b-5p, and miR-4488) that were upregulated and the pair (miR-4454 + miR-7975) that was downregulated in IA group vs controls. The expression change for these miRNAs was specific to patients with IA; they were not significantly differentiated between IACLAD and IA groups. Signaling pathways associated with an immunologic response to IA were found to be significantly enriched. CONCLUSIONS: We report a set of 5 differentially expressed miRNAs in the BAL of LTRs with IA that might help in the development of diagnostic and prognostic tools for IA in LTRs. However, further investigation is needed in a larger cohort to validate the findings.


Assuntos
Aspergillus/isolamento & purificação , Líquido da Lavagem Broncoalveolar/química , Transplante de Pulmão , MicroRNAs/genética , Disfunção Primária do Enxerto/genética , Aspergilose Pulmonar/genética , Transplantados , Adulto , Líquido da Lavagem Broncoalveolar/microbiologia , Feminino , Seguimentos , Humanos , Masculino , MicroRNAs/biossíntese , Pessoa de Meia-Idade , Disfunção Primária do Enxerto/metabolismo , Prognóstico , Aspergilose Pulmonar/metabolismo , Estudos Retrospectivos
12.
Gut Microbes ; 6(5): 310-20, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26322500

RESUMO

Shifts in the maternal gut microbiome have been implicated in metabolic adaptations to pregnancy. We investigated how pregnancy and diet interact to influence the composition of the maternal gut microbiota. Female C57BL/6 mice were fed either a control or a high fat diet for 8 weeks prior to mating. After confirmation of pregnancy, maternal weight gain and food intake were recorded. Fecal pellets were collected at 2 timepoints prior to mating (at the beginning of the experiment, and after 6 weeks of the specified diet) and at 4 timepoints during pregnancy (gestation day 0.5, 5.5, 10.5, and 15.5). The microbial composition and predicted metabolic functionality of the non-pregnant and pregnant gut was determined via sequencing of the variable 3 region of the 16S rRNA gene. Upon conception, differences in gut microbial communities were observed in both control and high fat-fed mice, including an increase in mucin-degrading bacteria. Control versus high fat-fed pregnant mice possessed the most profound changes to their maternal gut microbiota as indicated by statistically significant taxonomic differences. High fat-fed pregnant mice, when compared to control-fed animals, were found to be significantly enriched in microbes involved in metabolic pathways favoring fatty acid, ketone, vitamin, and bile synthesis. We show that pregnancy-induced changes in the female gut microbiota occur immediately at the onset of pregnancy, are vulnerable to modulation by diet, but are not dependent upon increases in maternal weight gain during pregnancy. High fat diet intake before and during pregnancy results in distinctive shifts in the pregnant gut microbiota in a gestational-age dependent manner and these shifts predict significant differences in the abundance of genes that favor lipid metabolism, glycolysis and gluconeogenic metabolic pathways over the course of pregnancy.


Assuntos
Dieta/métodos , Microbioma Gastrointestinal , Animais , DNA Ribossômico/química , DNA Ribossômico/genética , Fezes/microbiologia , Feminino , Camundongos Endogâmicos C57BL , Gravidez , RNA Ribossômico 16S/genética , Análise de Sequência de DNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA