Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Pediatr Res ; 95(3): 647-659, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37935884

ABSTRACT

BACKGROUND: Fetal growth restriction (FGR) increases risk for development of obesity and type 2 diabetes. Using a mouse model of FGR, we tested whether metabolic outcomes were exacerbated by high-fat diet challenge or associated with fecal microbial taxa. METHODS: FGR was induced by maternal calorie restriction from gestation day 9 to 19. Control and FGR offspring were weaned to control (CON) or 45% fat diet (HFD). At age 16 weeks, offspring underwent intraperitoneal glucose tolerance testing, quantitative MRI body composition assessment, and energy balance studies. Total microbial DNA was used for amplification of the V4 variable region of the 16 S rRNA gene. Multivariable associations between groups and genera abundance were assessed using MaAsLin2. RESULTS: Adult male FGR mice fed HFD gained weight faster and had impaired glucose tolerance compared to control HFD males, without differences among females. Irrespective of weaning diet, adult FGR males had depletion of Akkermansia, a mucin-residing genus known to be associated with weight gain and glucose handling. FGR females had diminished Bifidobacterium. Metabolic changes in FGR offspring were associated with persistent gut microbial changes. CONCLUSION: FGR results in persistent gut microbial dysbiosis that may be a therapeutic target to improve metabolic outcomes. IMPACT: Fetal growth restriction increases risk for metabolic syndrome later in life, especially if followed by rapid postnatal weight gain. We report that a high fat diet impacts weight and glucose handling in a mouse model of fetal growth restriction in a sexually dimorphic manner. Adult growth-restricted offspring had persistent changes in fecal microbial taxa known to be associated with weight, glucose homeostasis, and bile acid metabolism, particularly Akkermansia, Bilophilia and Bifidobacteria. The gut microbiome may represent a therapeutic target to improve long-term metabolic outcomes related to fetal growth restriction.


Subject(s)
Diabetes Mellitus, Type 2 , Fetal Growth Retardation , Humans , Female , Adult , Male , Infant , Fetal Growth Retardation/metabolism , Diet, High-Fat , Weight Gain , Glucose , Fetal Development
2.
Toxicol Sci ; 185(2): 158-169, 2022 01 24.
Article in English | MEDLINE | ID: mdl-34726736

ABSTRACT

Acetaminophen (APAP) overdose results in high morbidity and mortality, with limited treatment options. Increased understanding of the cellular signaling pathways activated in response to toxic APAP exposure is needed to provide insight into novel therapeutic strategies. Toxic APAP exposure induces hepatic nuclear factor kappa B (NFκB) activation. NFκB signaling has been identified to mediate the proinflammatory response but also induces a prosurvival and regenerative response. It is currently unknown whether potentiating NFkB activation would be injurious or advantageous after APAP overdose. The NFκB inhibitory protein beta (IκBß) dictates the duration and degree of the NFκB response following exposure to oxidative injuries. Thus, we sought to determine whether IκBß/NFκB signaling contributes to APAP-induced hepatic injury. At late time points (24 h) following toxic APAP exposures, mice expressing only IκBß knock-in mice (AKBI mice) exhibited increased serologic evidence of hepatic injury. This corresponded with increased histologic injury, specifically related to sinusoidal dilatation. When compared with wild type mice, AKBI mice demonstrated sustained hepatic nuclear translocation of the NFκB subunits p65 and p50, and enhanced NFκB target gene expression. This included increased expression of interleukin-6 (Il-6), a known contributor to hepatic sinusoidal dilation. This transcriptional response corresponded with increased plasma protein content of Il-6, as well as increased activation of signal transducer and activator of transcription 3.


Subject(s)
Acetaminophen , Chemical and Drug Induced Liver Injury , Acetaminophen/metabolism , Acetaminophen/toxicity , Animals , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Dilatation , I-kappa B Proteins , Interleukin-6/genetics , Interleukin-6/metabolism , Liver/metabolism , Mice , NF-kappa B/metabolism
3.
Front Immunol ; 12: 706774, 2021.
Article in English | MEDLINE | ID: mdl-34539638

ABSTRACT

Intrauterine growth restriction (IUGR) is a relevant predictor for higher rates of neonatal sepsis worldwide and is associated with an impaired neonatal immunity and lower immune cell counts. During the perinatal period, the liver is a key immunological organ responsible for the nuclear factor kappa B (NF-κB)-mediated innate immune response to inflammatory stimuli, but whether this role is affected by IUGR is unknown. Herein, we hypothesized that the newborn liver adapts to calorie-restriction IUGR by inducing changes in the NF-κB signaling transcriptome, leading to an attenuated acute proinflammatory response to intraperitoneal lipopolysaccharide (LPS). We first assessed the hepatic gene expression of key NF-κB factors in the IUGR and normally grown (NG) newborn mice. Real-time quantitative PCR (RT-qPCR) analysis revealed an upregulation of both IκB proteins genes (Nfkbia and Nfkbib) and the NF-κB subunit Nfkb1 in IUGR vs. NG. We next measured the LPS-induced hepatic expression of acute proinflammatory genes (Ccl3, Cxcl1, Il1b, Il6, and Tnf) and observed that the IUGR liver produced an attenuated acute proinflammatory cytokine gene response (Il1b and Tnf) to LPS in IUGR vs. unexposed (CTR). Consistent with these results, LPS-exposed hepatic tumor necrosis factor alpha (TNF-α) protein concentrations were lower in IUGR vs. LPS-exposed NG and did not differ from IUGR CTR. Sex differences at the transcriptome level were observed in the IUGR male vs. female. Our results demonstrate that IUGR induces key modifications in the NF-κB transcriptomic machinery in the newborn that compromised the acute proinflammatory cytokine gene and protein response to LPS. Our results bring novel insights in understanding how the IUGR newborn is immunocompromised due to fundamental changes in NF-κB key factors.


Subject(s)
Endotoxemia/immunology , Fetal Growth Retardation/immunology , Liver/immunology , NF-kappa B/immunology , Animals , Animals, Newborn , Female , Male , Mice , Pregnancy
4.
Am J Physiol Lung Cell Mol Physiol ; 321(5): L941-L953, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34585971

ABSTRACT

Both preclinical and clinical studies have demonstrated that exposures to acetaminophen (APAP) at levels that cause hepatic injury cause pulmonary injury as well. However, whether exposures that do not result in hepatic injury have acute pulmonary implications is unknown. Thus, we sought to determine how APAP exposures at levels that do not result in significant hepatic injury impact the mature lung. Adult male ICR mice (8-12 wk) were exposed to a dose of APAP known to cause hepatotoxicity in adult mice [280 mg/kg, intraperitoneal (ip)], as well as a lower dose previously reported to not cause hepatic injury (140 mg/kg, ip). We confirm that the lower dose exposures did not result in significant hepatic injury. However, like high dose, lower exposure resulted in increased cellular content of the bronchoalveolar lavage fluid and induced a proinflammatory pulmonary transcriptome. Both the lower and higher dose exposures resulted in measurable changes in lung morphometrics, with the lower dose exposure causing alveolar wall thinning. Using RNAScope, we were able to detect dose-dependent, APAP-induced pulmonary Cyp2e1 expression. Finally, using FLIM we determined that both APAP exposures resulted in acute pulmonary metabolic changes consistent with mitochondrial overload in lower doses and a shift to glycolysis at a high dose. Our findings demonstrate that APAP exposures that do not cause significant hepatic injury result in acute inflammatory, morphometric, and metabolic changes in the mature lung. These previously unreported findings may help explain the potential relationship between APAP exposures and pulmonary-related morbidity.


Subject(s)
Acetaminophen/toxicity , Liver/drug effects , Lung Injury/drug therapy , Lung/drug effects , Acetaminophen/metabolism , Animals , Cytochrome P-450 CYP2E1/metabolism , Disease Models, Animal , Glycolysis/drug effects , Liver/metabolism , Lung/metabolism , Lung Injury/metabolism , Mice , Mice, Inbred ICR
5.
Front Immunol ; 11: 1892, 2020.
Article in English | MEDLINE | ID: mdl-32973783

ABSTRACT

Compared to adults, neonates are at increased risk of infection. There is a growing recognition that dynamic qualitative and quantitative differences in immunity over development contribute to these observations. The liver plays a key role as an immunologic organ, but whether its contribution to the acute innate immune response changes over lifetime is unknown. We hypothesized that the liver would activate a developmentally-regulated acute innate immune response to intraperitoneal lipopolysaccharide (LPS). We first assessed the hepatic expression and activity of the NF-κB, a key regulator of the innate immune response, at different developmental ages (p0, p3, p7, p35, and adult). Ontogeny of the NF-κB subunits (p65/p50) revealed a reduction in Rela (p65) and Nfkb1 (p105, precursor to p50) gene expression (p0) and p65 subunit protein levels (p0 and p3) vs. older ages. The acute hepatic innate immune response to LPS was associated by the degradation of the NF-κB inhibitory proteins (IκBα and IκBß), and nuclear translocation of the NF-κB subunit p50 in all ages, whereas nuclear translocation of the NF-κB subunit p65 was only observed in the p35 and adult mouse. Consistent with these findings, we detected NF-κB subunit p65 nuclear staining exclusively in the LPS-exposed adult liver compared with p7 mouse. We next interrogated the LPS-induced hepatic expression of pro-inflammatory genes (Tnf, Icam1, Ccl3, and Traf1), and observed a gradually increase in gene expression starting from p0. Confirming our results, hepatic NF-κB subunit p65 nuclear translocation was associated with up-regulation of the Icam1 gene in the adult, and was not detected in the p7 mouse. Thus, an inflammatory challenge induces an NF-κB-mediated hepatic innate immune response activation across all developmental ages, but nuclear translocation of the NF-κB subunit p65 and associated induction of pro-inflammatory genes occurred only after the first month of life. Our results demonstrate that the LPS-induced hepatic innate immune response is developmentally regulated by the NF-κB subunit p65 in the mouse.


Subject(s)
Endotoxemia/metabolism , Immunity, Innate , Liver/metabolism , Toll-Like Receptor 4/metabolism , Transcription Factor RelA/metabolism , Age Factors , Animals , Chemokine CCL3/genetics , Chemokine CCL3/metabolism , Disease Models, Animal , Endotoxemia/chemically induced , Endotoxemia/genetics , Endotoxemia/immunology , Gene Expression Regulation, Developmental , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Lipopolysaccharides , Liver/immunology , Male , Mice, Inbred ICR , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/metabolism , Signal Transduction , TNF Receptor-Associated Factor 1/genetics , TNF Receptor-Associated Factor 1/metabolism , Transcription Factor RelA/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
6.
Innate Immun ; 26(7): 549-564, 2020 10.
Article in English | MEDLINE | ID: mdl-32538259

ABSTRACT

Chorioamnionitis is associated with inflammatory end-organ damage in the fetus. Tissues in direct contact with amniotic fluid drive a pro-inflammatory response and contribute to this injury. However, due to a lack of direct contact with the amniotic fluid, the liver contribution to this response has not been fully characterized. Given its role as an immunologic organ, we hypothesized that the fetal liver would demonstrate an early innate immune response to an in utero inflammatory challenge. Fetal sheep (131 ± 1 d gestation) demonstrated metabolic acidosis and high cortisol and norepinephrine values within 5 h of exposure to intra-amniotic LPS. Likewise, expression of pro-inflammatory cytokines increased significantly at 1 and 5 h of exposure. This was associated with NF-κB activation, by inhibitory protein IκBα degradation, and nuclear translocation of NF-κB subunits (p65/p50). Corroborating these findings, LPS exposure significantly increased pro-inflammatory innate immune gene expression in fetal sheep hepatic macrophages in vitro. Thus, an in utero inflammatory challenge induces an early hepatic innate immune response with systemic metabolic and stress responses. Within the fetal liver, hepatic macrophages respond robustly to LPS exposure. Our results demonstrate that the fetal hepatic innate immune response must be considered when developing therapeutic approaches to attenuate end-organ injury associated with in utero inflammation.


Subject(s)
Acidosis/immunology , Chorioamnionitis/immunology , Inflammation/immunology , Liver/immunology , Macrophages/metabolism , Pregnancy/immunology , Uterus/immunology , Animals , Disease Models, Animal , Female , Fetus , Gene Expression Regulation , Humans , Hydrocortisone/metabolism , Immunity, Innate/genetics , Lipopolysaccharides/immunology , Macrophages/immunology , Norepinephrine/metabolism , Sheep
7.
Reprod Sci ; 27(11): 2063-2074, 2020 11.
Article in English | MEDLINE | ID: mdl-32542534

ABSTRACT

The transcription factor NFκB has been associated with the timing of menopause in a large human genome-wide association study. Furthermore, preclinical studies demonstrate that loss of Tumor necrosis factor alpha (Tnfα) or its receptor Tnfr2 slows primordial follicle growth activation (PFGA). Although Tnfα:receptor signaling stimulates NFκB and may mechanistically link these findings, very little is known about NFκB signaling in PFGA. Because signaling downstream of Tnfα/Tnfr2 ligand/receptor interaction has not been interrogated as relates to PFGA, we evaluated the expression of key NFκB signaling proteins in primordial and growing follicles, as well as during ovarian aging. We show that key members of the NFκB pathway, including subunits, activating kinases, and inhibitory proteins, are expressed in the murine ovary. Furthermore, the subunits p65 and p50, and the cytosolic inhibitory proteins IκBα and IκBß, are present in ovarian follicles, including at the primordial stage. Finally, we assessed PFGA in genetically modified mice (AKBI) previously demonstrated to be resistant to inflammatory stress-induced NFκB activation due to overexpression of the NFκB inhibitory protein IκBß. Consistent with the hypothesis that NFκB plays a key role in PFGA, AKBI mice exhibit slower PGFA than wild-type (WT) controls, and their ovaries contain nearly twice the number of primordial follicles as WT both at early and late reproductive ages. These data provide mechanistic insight on the control of PFGA and suggest that targeting NFκB at the level of IκB proteins may be a tractable route to slowing the rate of PFGA in women faced with early ovarian demise.


Subject(s)
NF-kappa B/metabolism , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Signal Transduction , Animals , Female , I-kappa B Proteins/metabolism , Mice, Inbred ICR , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B p50 Subunit/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
J Physiol ; 596(23): 6063-6077, 2018 12.
Article in English | MEDLINE | ID: mdl-29882596

ABSTRACT

KEY POINTS: The cerebral response to fetal asphyxia is characterized by an upregulation of nucleic acid and chromatin modification processes, as well as a downregulation of metabolic processes at 1 h post-umbilical cord occlusion (UCO). Twenty-four hours post UCO, there was an upregulation of metabolic processes and protein modifications. UCO did not alter bacterial gene expression levels, nor did it produce a robust inflammatory response compared to maternal hypoxia. The administration of ketamine produced minimal effects on the fetal response to UCO in the cerebral cortex. ABSTRACT: Umbilical cord occlusion (UCO) is known to cause neurological disorders in the neonate. Previously, we have reported that hypoxic hypoxia (HH) stimulates the appearance of bacteria in the fetal brain and upregulates the expression of inflammatory markers in fetal cerebral cortex (CTX) and also that ketamine attenuates these responses. In the present study, we aimed to test the hypothesis that UCO, similar to HH, produces an inflammatory response in the fetal CTX and also that treatment with ketamine reduces these effects. In chronically instrumented fetal sheep (∼125 days), 30 min of partial UCO decreased fetal PaO2 levels by ∼50%. Half of the fetuses received ketamine (3 mg kg-1 ) 10 min prior to UCO (n = 4 per group). Fetal brains were collected 1 and 24 h after the experiment and mRNA was extracted and hybridized for microarray analyses. Differentially-expressed genes were analysed for significant association with gene ontologies and pathways. After 1 h, UCO upregulated nucleic acid processing and chromatin modification and downregulated metabolic processes compared to control. After 24 h, UCO upregulated metabolic and protein modification processes. Ketamine produced minimal effects. UCO did not alter the abundance of bacterial DNA in fetal brain, nor did it upregulate inflammation pathways compared to HH. We conclude that UCO produced time-dependent responses that did not include bacterial invasion or upregulation of inflammation pathways in fetal CTX. This contrasts with the response to HH, which resulted in the appearance of bacteria in the CTX and upregulated inflammation pathways. These responses in fetal CTX to oxygen deprivation are therefore modified by the maternal or placental response to the stimulus.


Subject(s)
Cerebral Cortex/metabolism , Fetal Hypoxia/genetics , Fetus/metabolism , Ischemia/genetics , Transcriptome , Umbilical Cord/blood supply , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/microbiology , DNA, Bacterial , Excitatory Amino Acid Antagonists/pharmacology , Female , Fetus/drug effects , Gene Expression Regulation, Developmental/drug effects , Ketamine/pharmacology , Pregnancy , Sheep , Transcriptome/drug effects
9.
Rev. biol. trop ; 66(1): 189-203, Jan.-Mar. 2018. tab, graf
Article in Spanish | LILACS | ID: biblio-897664

ABSTRACT

Resumen El pez león (Pterois volitans) invadió la región del Caribe y tiene el potencial de alterar la composición y estructura de las comunidades de peces en los arrecifes coralinos. El objetivo de este estudio fue analizar los índices de diversidad en las comunidades de peces nativos en sitios invadidos por el pez león en dos áreas marinas protegidas (AMP) del Caribe y compararlos con datos previos a la invasión. En ambas AMP, Parque Nacional Guanahacabibes (PNG) en el occidente de Cuba y Parque Nacional Arrecifes de Xcalak (PNAX) en el S de Quintana Roo, se realizaron censos visuales de las especies de peces en hábitats durante las épocas de seca y lluvia del 2013-2015. Se evaluaron nueve sitios, mediante conteos estacionarios. Se registró mayor riqueza de especies en el PNG (43.47±5.14) que en el PNAX (40.22±4.96). No se observaron diferencias entre épocas en ninguna de las AMP. El pez león se ubicó entre las especies más abundantes del PNG. La abundancia media en el PNG (0.76 ± 1.25) fue mayor a la registrada en el PNAX (0.19±0.46). La diversidad disminuyó después de la llegada del pez león en un solo sitio del PNG y en dos sitios del PNAX, pero al parecer estos resultados están más asociados al efecto de la pesca que a la presencia del pez león. A partir de los resultados y asumiendo que los cambios en las comunidades de peces por el pez león podrían no detectarse aún, recomendamos seguir los monitoreos de los descriptores comunitarios para detectar cambios futuros en las comunidades de peces.


Abstract Lionfish (Pterois volitans) invaded the Caribbean region with the potential to alter the composition and structure of native coral reef fish communities. The objective of this study was to analyze the diversity indices of these fish communities potentially affected by lionfish predation and to compare with pre-invasion data. The study was undertaken in two Caribbean marine protected areas (MPAs): Guanahacabibes National Park (PNG) in W Cuba and Xcalak Reefs National Park (PNAX) in S Quintana Roo. We carried out visual censuses of fish species in reef habitats during the dry and rainy seasons of the period 2013-2015. For this, nine sites were defined and evaluated using stationary counts. Our results showed higher species richness (43.47 ± 5.14) and mean abundance (0.76 ± 1.25) in PNG than in PNAX (40.22 ± 4.96, 0.19 ± 0.46, respectively). Diversity decreased after the arrival of lionfish in a single site of PNG and in two sites of the PNAX, but apparently, these results are more related to the fishing activity effect than to the lionfish presence. Based on the results and assuming that changes in the native fish communities by lionfish may not yet be detected, we recommend to continue the monitoring community descriptions in order to detect future changes in native fish communities. Rev. Biol. Trop. 66(1): 189-203. Epub 2018 March 01.

10.
Asian-Australas J Anim Sci ; 31(2): 208-217, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28728358

ABSTRACT

OBJECTIVE: Examine the effects of supplementing bahiagrass hay (BG) with potentially anthelmintic quantities of hays of perennial peanut (PEA) or sericea lespedeza (LES) or seeds of velvet bean (Mucuna pruriens L.; MUC) or papaya (PAP) on the intake and nutritive value (Experiment 1), and the performance and parasite burden (Experiment 2) of goats. METHODS: In Experiment 1, 38 male goats (27.4±5.7 kg body weight) were randomly assigned to each of 5 treatments: i) BG alone and BG plus; ii) PEA; iii) LES; iv) MUC; and v) PAP. Goats were fed for ad libitum consumption and adapted to the diets for 14 d followed by 7 d of measurement. The PEA, LES, MUC (50%, 50%, and 10% of the diet dry matter [DM], respectively), and PAP (forced-fed at 10 g/d) were fed at rates that would elicit anthelmintic effects. In Experiment 2, goats remained in the same treatments but were allocated to 15 pens (3 pens per treatment) from d 22 to 63. All goats were infected with parasites by grazing an infected bahiagrass pasture from 0800 to 1500 h daily and then returned to the pens. RESULTS: Dry matter intake tended to be greater in goats fed PEA and LES than those fed BG (757 and 745 vs 612 g/d, respectively). Digestibility of DM (59.5% vs 54.9%) and organic matter (60.8% vs 56.0%) were greater in goats fed MUC vs BG, respectively. In Experiment 2, feeding PAP, LES, and PEA to goats reduced nematode fecal egg counts by 72%, 52%, and 32%, reduced abomasal adult worm counts by 78%, 52%, and 42%, and decreased plasma haptoglobin concentrations by 42%, 40%, and 45% relative to feeding BG alone, respectively. CONCLUSION: Supplementation with PEA, LES, and PAP decreased the parasite burden of goats but did not increase their performance. PAP was the most effective anthelmintic supplement.

11.
Front Physiol ; 9: 1858, 2018.
Article in English | MEDLINE | ID: mdl-30666211

ABSTRACT

The physiological response to hypoxia in the fetus has been extensively studied with regard to redistribution of fetal combined ventricular output and sparing of oxygen delivery to fetal brain and heart. Previously, we have shown that the fetal brain is capable of mounting changes in gene expression that are consistent with tissue inflammation. The present study was designed to use transcriptomics and systems biology modeling to test the hypothesis that ketamine reduces or prevents the upregulation of inflammation-related pathways in hypothalamus and hippocampus after transient hypoxic hypoxia. Chronically catheterized fetal sheep (122 ± 5 days gestation) were subjected to 30 min hypoxia (relative reduction in PaO2∼50%) caused by infusion of nitrogen into the inspired gas of the pregnant ewe. RNA was isolated from fetal hypothalamus and hippocampus collected 24 h after hypoxia, and was analyzed for gene expression using the Agilent 15.5 k ovine microarray. Ketamine, injected 10 min prior to hypoxia, reduced the cerebral immune response activation to the hypoxia in both brain regions. Genes both upregulated by hypoxia and downregulated by ketamine after hypoxia were significantly associated with gene ontology terms and KEGG pathways that are, themselves, associated with the tissue response to exposure to bacteria. We conclude that the results are consistent with interruption of the cellular response to bacteria by ketamine.

12.
Sci Rep ; 7(1): 6458, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28743956

ABSTRACT

Herein we describe an association between activation of inflammatory pathways following transient hypoxia and the appearance of the multidrug resistant bacteria Staphylococcus simulans in the fetal brain. Reduction of maternal arterial oxygen tension by 50% over 30 min resulted in a subseiuent significant over-expression of genes associated with immune responses 24 h later in the fetal brain. The activated genes were consistent with stimulation by bacterial lipopolysaccharide; an influx of macrophages and appearance of live bacteria were found in these fetal brains. S. simulans was the predominant bacterial species in fetal brain after hypoxia, but was found in placenta of all animals. Strains of S. simulans from the placenta and fetal brain were equally highly resistant to multiple antibiotics including methicillin and had identical genome sequences. These results suggest that bacteria from the placenta invade the fetal brain after maternal hypoxia.


Subject(s)
Brain/microbiology , Drug Resistance, Multiple, Bacterial , Fetal Hypoxia/complications , Placenta/microbiology , Staphylococcus/pathogenicity , Animals , Brain/embryology , Brain/pathology , Female , Fetal Hypoxia/pathology , Fetal Hypoxia/physiopathology , Gene Expression Regulation, Developmental , Macrophages/pathology , Microglia/pathology , Pregnancy , Sheep , Staphylococcus/drug effects , Staphylococcus/genetics
13.
Physiol Rep ; 4(17)2016 09.
Article in English | MEDLINE | ID: mdl-27597770

ABSTRACT

Umbilical cord occlusion (UCO) is a hypoxic insult that has been used to model birth asphyxia and umbilical cord compression in utero. UCO triggers vigorous neural and endocrine responses that include increased plasma ACTH and cortisol concentrations, increased blood pressure (BP), and decreased heart rate (HR). We have previously reported that ketamine, a noncompetitive N-methyl-D-aspartate receptor antagonist, can modify the fetal hemodynamic and ACTH responses to ventilatory hypoxia and cerebral ischemia-reperfusion. We performed the present experiments to test the hypothesis that ketamine has similar effects on the neuroendocrine and cardiovascular responses to UCO Fetal sheep were chronically catheterized at gestational day 125. Ketamine (3 mg/kg) was administered intravenously to the fetus 10 min prior to the insult. UCO was induced for 30 min by reducing the umbilical vein blood flow until fetal PaO2 levels were reduced from 17 ± 1 to 11 ± 1 mm Hg. UCO produced an initial increase on fetal BP in both control and ketamine groups (P = 0.018 time), followed by a decrease in the control group, but values remained higher with ketamine. HR decreased after UCO (P = 0.041 stimulus*time) in both groups, but the reduction was greater initially in control compared to ketamine groups. Fetal PaCO2 levels increased after UCO (P < 0.01 stimulus*time), but values were higher in the control versus ketamine groups. UCO significantly decreased fetal pH values (P < 0.01 stimulus*time) with a greater effect on the control versus ketamine group. Ketamine delayed the cortisol responses to UCO (P < 0.001 stimulus*time), and UCO produced a robust increase in ACTH levels from 19 ± 2 to 280 ± 27 pg/mL (P < 0.001 stimulus*time), but there were no differences in ACTH levels between UCO groups. We conclude that ketamine augmented the cardiovascular response to UCO, but did not alter the ACTH response to UCO.


Subject(s)
Analgesics/administration & dosage , Hemodynamics/drug effects , Ketamine/administration & dosage , Pregnancy, Animal , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Umbilical Cord/embryology , Umbilical Cord/physiopathology , Adrenocorticotropic Hormone/blood , Adrenocorticotropic Hormone/drug effects , Analgesics/pharmacology , Animals , Blood Pressure/drug effects , Carbon Dioxide/blood , Female , Fetal Blood/chemistry , Fetal Hypoxia/physiopathology , Fetus/metabolism , Hydrocortisone/blood , Hydrocortisone/metabolism , Hypoxia/chemically induced , Ketamine/pharmacology , Pregnancy , Receptors, N-Methyl-D-Aspartate/metabolism , Sheep , Sheep, Domestic , Umbilical Cord/blood supply
14.
Physiol Rep ; 4(6)2016 Mar.
Article in English | MEDLINE | ID: mdl-27033443

ABSTRACT

Transient hypoxia in pregnancy stimulates a physiological reflex response that redistributes blood flow and defends oxygen delivery to the fetal brain. We designed the present experiment to test the hypotheses that transient hypoxia produces damage of the cerebral cortex and that ketamine, an antagonist ofNMDAreceptors and a known anti-inflammatory agent, reduces the damage. Late gestation, chronically catheterized fetal sheep were subjected to a 30-min period of ventilatory hypoxia that decreased fetal PaO2from 17 ± 1 to 10 ± 1 mmHg, or normoxia (PaO217 ± 1 mmHg), with or without pretreatment (10 min before hypoxia/normoxia) with ketamine (3 mg/kg, i.v.). One day (24 h) after hypoxia/normoxia, fetal cerebral cortex was removed andmRNAextracted for transcriptomics and systems biology analysis (n = 3-5 per group). Hypoxia stimulated a transcriptomic response consistent with a reduction in cellular metabolism and an increase in inflammation. Ketamine pretreatment reduced both of these responses. The inflammation response modeled with transcriptomic systems biology was validated by immunohistochemistry and showed increased abundance of microglia/macrophages after hypoxia in the cerebral cortical tissue that ketamine significantly reduced. We conclude that transient hypoxia produces inflammation of the fetal cerebral cortex and that ketamine, in a standard clinical dose, reduces the inflammation response.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cerebral Cortex/drug effects , Fetal Hypoxia/drug therapy , Hypoxia, Brain/drug therapy , Inflammation Mediators/metabolism , Ketamine/pharmacology , Neuroprotective Agents/pharmacology , Animals , Cerebral Cortex/immunology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Female , Fetal Hypoxia/genetics , Fetal Hypoxia/immunology , Fetal Hypoxia/metabolism , Fetal Hypoxia/pathology , Gene Expression Profiling/methods , Gene Expression Regulation , Gestational Age , Hypoxia, Brain/genetics , Hypoxia, Brain/immunology , Hypoxia, Brain/metabolism , Hypoxia, Brain/pathology , Inflammation Mediators/immunology , Oligonucleotide Array Sequence Analysis , Pregnancy , RNA, Messenger/metabolism , Sheep , Systems Biology , Time Factors
15.
J Physiol ; 594(5): 1295-310, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26497972

ABSTRACT

Acute fetal hypoxia is a form of fetal stress that stimulates renal vasoconstriction and ischaemia as a consequence of the physiological redistribution of combined ventricular output. Because of the potential ischaemia-reperfusion injury to the kidney, we hypothesized that it would respond to hypoxia with an increase in the expression of inflammatory genes, and that ketamine (an N-methyl-D-aspartate receptor antagonist) would reduce or block this response. Hypoxia was induced for 30 min in chronically catheterized fetal sheep (125 ± 3 days), with or without ketamine (3 mg kg(-1)) administered intravenously to the fetus 10 min prior to hypoxia. Gene expression in fetal kidney cortex collected 24 h after the onset of hypoxia was analysed using ovine Agilent 15.5k array and validated with qPCR and immunohistochemistry in four groups of ewes: normoxic control, normoxia + ketamine, hypoxic control and hypoxia + ketamine (n = 3-4 per group). Significant differences in gene expression between groups were determined with t-statistics using the limma package for R (P ≤ 0.05). Enriched biological processes for the 427 upregulated genes were immune and inflammatory responses and for the 946 downregulated genes were metabolic processes. Ketamine countered the effects of hypoxia on upregulated immune/inflammatory responses as well as the downregulated metabolic responses. We conclude that our transcriptomics modelling predicts that hypoxia activates inflammatory pathways and reduces metabolism in the fetal kidney cortex, and ketamine blocks or ameliorates this response. The results suggest that ketamine may have therapeutic potential for protection from ischaemic renal damage.


Subject(s)
Excitatory Amino Acid Antagonists/therapeutic use , Fetal Hypoxia/drug therapy , Ketamine/therapeutic use , Kidney/physiopathology , Animals , Chemokines/genetics , Chemokines/metabolism , Female , Inflammation/drug therapy , Interleukins/genetics , Interleukins/metabolism , Kidney/blood supply , Kidney/metabolism , Pregnancy , Sheep
16.
Circ Cardiovasc Genet ; 7(4): 423-433, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25028484

ABSTRACT

BACKGROUND: Heterozygous human mutations of NKX2-5 are highly penetrant and associated with varied congenital heart defects. The heterozygous knockout of murine Nkx2-5, in contrast, manifests less profound cardiac malformations, with low disease penetrance. We sought to study this apparent discrepancy between human and mouse genetics. Because missense mutations in the NKX2-5 homeodomain (DNA-binding domain) are the most frequently reported type of human mutation, we replicated this genetic defect in a murine knockin model. METHODS AND RESULTS: We generated a murine model in a 129/Sv genetic background by knocking-in an Nkx2-5 homeodomain missense mutation previously identified in humans. The mutation was located at homeodomain position 52Arg→Gly (R52G). All the heterozygous neonatal Nkx2-5(+/R52G) mice demonstrated a prominent trabecular layer in the ventricular wall, so called noncompaction, along with diverse cardiac anomalies, including atrioventricular septal defects, Ebstein malformation of the tricuspid valve, and perimembranous and muscular ventricular septal defects. In addition, P10 Nkx2-5(+/R52G) mice demonstrated atrial sepal anomalies, with significant increase in the size of the interatrial communication and fossa ovalis, and decrease in the length of the flap valve compared with control Nkx2-5(+/+) or Nkx2-5(+/-) mice. CONCLUSIONS: The results of our study demonstrate that heterozygous missense mutation in the murine Nkx2-5 homeodomain (R52G) is highly penetrant and result in pleiotropic cardiac effects. Thus, in contrast to heterozygous Nkx2-5 knockout mice, the effects of the heterozygous knockin mimic findings in humans with heterozygous missense mutation in NKX2-5 homeodomain.


Subject(s)
Heart Defects, Congenital/genetics , Homeodomain Proteins/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Disease Models, Animal , Gene Knock-In Techniques , Heart Defects, Congenital/pathology , Heart Ventricles/pathology , Heterozygote , Homeobox Protein Nkx-2.5 , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Mutation, Missense , Phenotype , Protein Structure, Tertiary
17.
Physiol Genomics ; 46(14): 523-32, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24824211

ABSTRACT

Estradiol (E2) is a well-known modulator of fetal neuroendocrine activity and has been proposed as a critical endocrine signal readying the fetus for birth and postnatal life. To investigate the modulatory role of E2 on fetal stress responsiveness and the response of the fetal brain to asphyxic stress, we subjected chronically catheterized fetal sheep to a transient (10 min) brachiocephalic artery occlusion (BCO) or sham occlusion. Half of the fetuses received subcutaneous pellets that increased plasma E2 concentrations within the physiological range. Hypothalamic mRNA was analyzed using the Agilent 8x15k ovine array (019921), processed and annotated as previously reported by our laboratory. Analysis of the data by ANOVA revealed that E2 differentially regulated (DR) 561 genes, and BCO DR 894 genes compared with control and E2+BCO DR 1,153 genes compared with BCO alone (all P < 0.05). E2 upregulated epigenetic pathways and downregulated local steroid biosynthesis but did not significantly involve genes known to directly respond to the estrogen receptor. Brachiocephalic occlusion upregulated kinase pathways as well as genes associated with lymphocyte infiltration into the brain and downregulated neuropeptide synthesis. E2 upregulated immune- and apoptosis-related pathways after BCO and reduced kinase and epigenetic pathway responses to the BCO. Responses to BCO are different from responses to hypoxic hypoxia suggesting that mechanisms of responses to these two forms of brain hypoxia are distinct. We conclude that cerebral ischemia caused by BCO might stimulate lymphocyte infiltration into the brain and that this response appears to be modified by estradiol.


Subject(s)
Brachiocephalic Trunk/drug effects , Estradiol/pharmacology , Fetus/drug effects , Hypothalamus/drug effects , Transcriptome/drug effects , Transcriptome/genetics , Animals , Brachiocephalic Trunk/metabolism , Brain/drug effects , Brain/metabolism , Brain Ischemia/genetics , Brain Ischemia/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/genetics , Fetus/metabolism , Hypothalamus/metabolism , Hypoxia/embryology , Hypoxia/genetics , Lymphocytes/drug effects , Neuropeptides/genetics , Neuropeptides/metabolism , RNA, Messenger/genetics , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Sheep/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...