Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
Add more filters










Publication year range
1.
Cell Metab ; 36(3): 575-597.e7, 2024 03 05.
Article in English | MEDLINE | ID: mdl-38237602

ABSTRACT

The glucagon receptor (GCGR) in the kidney is expressed in nephron tubules. In humans and animal models with chronic kidney disease, renal GCGR expression is reduced. However, the role of kidney GCGR in normal renal function and in disease development has not been addressed. Here, we examined its role by analyzing mice with constitutive or conditional kidney-specific loss of the Gcgr. Adult renal Gcgr knockout mice exhibit metabolic dysregulation and a functional impairment of the kidneys. These mice exhibit hyperaminoacidemia associated with reduced kidney glucose output, oxidative stress, enhanced inflammasome activity, and excess lipid accumulation in the kidney. Upon a lipid challenge, they display maladaptive responses with acute hypertriglyceridemia and chronic proinflammatory and profibrotic activation. In aged mice, kidney Gcgr ablation elicits widespread renal deposition of collagen and fibronectin, indicative of fibrosis. Taken together, our findings demonstrate an essential role of the renal GCGR in normal kidney metabolic and homeostatic functions. Importantly, mice deficient for kidney Gcgr recapitulate some of the key pathophysiological features of chronic kidney disease.


Subject(s)
Receptors, Glucagon , Renal Insufficiency, Chronic , Humans , Animals , Mice , Receptors, Glucagon/metabolism , Down-Regulation , Mice, Knockout , Kidney/metabolism , Homeostasis/physiology , Lipids
2.
Proteomics ; 24(7): e2300260, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38059784

ABSTRACT

Intrauterine growth restriction (IUGR) is associated with increased risk of cardiometabolic disease later in life and has been shown to affect female and male offspring differently, but the mechanisms remain unclear. The purpose of this study was to identify proteomic differences and metabolic risk markers in IUGR male and female neonates when compared to appropriate for gestational age (AGA) babies that will provide a better understanding of IUGR pathogenesis and its associated risks. Our results revealed alterations in IUGR cord plasma proteomes with most of the differentially abundant proteins implicated in peroxisome pathways. This effect was evident in females but not in males. Furthermore, we observed that catalase activity, a peroxisomal enzyme, was significantly increased in females (p < 0.05) but unchanged in males. Finally, we identified risk proteins associated with obesity, type-2 diabetes, and glucose intolerance such as EGF containing fibulin extracellular matrix protein 1 (EFEMP1), proprotein convertase subtilisin/kexin type 9 (PCSK9) and transforming growth factor beta receptor 3 (TGFBR3) proteins unique to females while coagulation factor IX (C9) and retinol binding protein 4 (RBP4) are unique in males. In conclusion, IUGR may display sexual dimorphism which may be associated with differences in lifelong risk for cardiometabolic disease between males and females.


Subject(s)
Cardiovascular Diseases , Fetal Growth Retardation , Infant, Newborn , Infant , Humans , Male , Female , Fetal Growth Retardation/etiology , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Proprotein Convertase 9/metabolism , Proteomics , Retinol-Binding Proteins, Plasma , Extracellular Matrix Proteins/metabolism
4.
Biomolecules ; 13(8)2023 08 06.
Article in English | MEDLINE | ID: mdl-37627289

ABSTRACT

Babies born to severe acute respiratory syndrome corona virus-2 (SARS-CoV-2)-infected mothers are at greater risk for perinatal morbidity and more likely to receive a neurodevelopmental diagnosis in the first year of life. However, the effect of maternal infection on placental function and neonatal outcomes varies depending upon the patient population. We set out to test our hypothesis that maternal SARS-CoV-2 infection in our underserved, socioeconomically disadvantaged, mostly unvaccinated, predominantly African American and Latina population in the Bronx, NY would have effects evident at birth. Under IRB approval, 56 SARS-CoV-2-positive patients infected during the "first wave" of the pandemic with alpha and beta strains of the virus, 48 patients infected during the "second wave" of the pandemic with delta and omicron strains and 61 negative third-trimester high-risk patients were randomly selected from Montefiore Medical Center (MMC), Bronx, NY. In addition, two positive cases from Yale New Haven Hospital, CT were included as controls. All 104 placentas delivered by SARS-CoV-2-positive mothers were uninfected by the virus, based on immunohistochemistry, in situ hybridization, and qPCR analysis. However, placental villous infarcts were significantly increased in first-wave cases compared to second-wave cases or negative controls. Significantly lower Apgar scores at 1 min and 5 min were observed in neonates born to infected mothers with severe symptoms. These findings suggest that even without entering the placenta, SARS-CoV-2 can affect various systemic pathways, culminating in altered placental development and function, which may adversely affect the fetus, especially in a high-risk patient population such as ours. These results underline the importance of vaccination among pregnant women, particularly in low-resource areas.


Subject(s)
COVID-19 , Female , Humans , Infant, Newborn , Pregnancy , Apgar Score , COVID-19/epidemiology , Infarction , Mothers , Placenta , Pregnant Women , SARS-CoV-2
5.
Front Pediatr ; 11: 1150216, 2023.
Article in English | MEDLINE | ID: mdl-37425276

ABSTRACT

Introduction: The effects of psychological distress/resilience on parent-child engagement (e.g., family dinners, reading) during the COVID-19 pandemic have not been well studied. Among very young children from underrepresented backgrounds enrolled in the ongoing longitudinal Bronx Mother Baby Health Study of healthy term infants, we (1) examined associations between exposures to COVID-19-related events, demographic factors and parental psychological distress and resilience; and (2) correlated these factors with parent-child engagement activities. Methods: Between June 2020-August 2021, parents of 105 Bronx Mother Baby Health Study participants aged birth-25 months completed questionnaires related to exposures to COVID-19-related events, frequency of positive parent-child engagement activities, food and housing insecurity, and parental psychological distress and resilience. Families were also asked open ended questions about the pandemic's impact. Results: 29.8% and 47.6% of parents reported food and housing insecurity, respectively. Greater exposures to COVID-19-related events were associated with increased parental psychological distress. Positive parent-child interactions were associated with demographic factors and higher levels of maternal education, but not with exposures to COVID-19-related events. Discussion: This study adds to a growing body of literature on the negative impacts of COVID-19 exposures and psychosocial stressors on families during the pandemic, supporting the need for enhanced mental health resources and social supports for families.

6.
Neoreviews ; 23(6): e363-e372, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35641462

ABSTRACT

Epidemiologic studies have shown an association between an adverse intrauterine environment (eg, exposure to malnutrition) and an increased risk of developing cardiometabolic disease in adulthood. These studies laid the foundation for the developmental origins of health and disease hypothesis, which states that limited nutrient supply to the fetus results in physiologic and metabolic adaptations that favor survival but result in unfavorable consequences in the offspring if there is excess nutrition after birth. This discrepancy in the pre- and postnatal milieus, perceived as stress by the offspring, may confer an increased risk of developing cardiometabolic disease later in life. Thus, early life exposures result in programming or changes in cellular memory that have effects on health throughout the life course. One of the mechanisms by which programming occurs is via epigenetic modifications of genes, processes that result in functionally relevant changes in genes (ie, gene expression) without an alteration in the genotype. In this review, we will describe how fetal exposures, including under- and overnutrition, affect neonatal and childhood growth and the future risk for cardiometabolic disease.


Subject(s)
Cardiovascular Diseases , Prenatal Exposure Delayed Effects , Adult , Cardiovascular Diseases/complications , Cardiovascular Diseases/genetics , Child , Epigenesis, Genetic , Female , Fetal Development/genetics , Humans , Infant, Newborn , Obesity , Prenatal Exposure Delayed Effects/genetics
7.
F S Sci ; 3(1): 95-105, 2022 02.
Article in English | MEDLINE | ID: mdl-35559999

ABSTRACT

OBJECTIVE: To examine, following perinatal exposure to a diet high in advanced glycation end products (AGEs), whether the use of standard AGE-free mouse chow during the postweaning period alters metabolism and reproduction differently than exposure to a diet low in AGEs. DESIGN: Experimental animal study. SETTING: University-based research laboratory. ANIMAL(S): Female CD1 mice. INTERVENTION(S): Seven-week-old mice were placed on a diet either low or high in AGEs perinatally, before mating and then during pregnancy and lactation. All offspring were weaned onto an AGE-free normal chow. MAIN OUTCOME MEASURE(S): Growth curve, liver and abdominal fat weight, insulin and glucose tolerance tests, vaginal opening, estrous cyclicity, and serum levels of antimüllerian hormone, leptin, and adiponectin were assessed. Ovarian histologic examination for follicular count and gene expression was also performed. RESULT(S): Compared with the mice exposed to a diet low in AGEs, the mice exposed to a diet high in AGEs showed lower body weight in pups, lower liver weight, delayed vaginal opening, higher serum antimüllerian hormone levels, lower primordial and secondary follicle pools, and higher ovarian Fshr messenger RNA levels. CONCLUSION(S): Following weaning, perinatal AGEs can target puberty onset and folliculogenesis differently to standard mouse chow.


Subject(s)
Anti-Mullerian Hormone , Glycation End Products, Advanced , Animals , Diet/adverse effects , Female , Glycation End Products, Advanced/adverse effects , Humans , Mice , Phenotype , Pregnancy , Reproduction , Weaning
8.
Mol Hum Reprod ; 26(8): 615-623, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32609365

ABSTRACT

Maternal nutrition and the intrauterine environment are important in determining susceptibility to reproductive and metabolic disturbances. Advanced glycation end products (AGEs) are widely consumed in Western diet. The purpose of this study was to determine whether perinatal exposure to a high levels of dietary AGEs affect metabolic and reproductive parameters in female mice offspring. Female CD1 mice, 7 weeks old, were placed on either a diet low (L-AGE) or high (H-AGE) in AGEs before mating and then during pregnancy and lactation. All offspring were weaned onto the L-AGE diet and studied through to 16 weeks of age; they were counted and weighed at birth and then every week for a total of 11 weeks. Vaginal opening, litter size, growth curve, liver and abdominal fat weights, serum levels of anti-Mullerian hormone, leptin and adiponectin, as well as insulin and glucose tolerance tests were compared. Ovaries were harvested for follicular count and gene expression by real-time polymerase chain reaction. Compared to perinatal exposure to the L-AGE diet, perinatal exposure to the H-AGE diet caused lower body weight at birth, and adult offspring exhibited delayed growth, lower serum leptin and adiponectin levels, delayed vaginal opening, irregular oestrous cyclicity, arrested follicular development and significant alterations in the expression of genes involved in folliculogenesis (Amh and Amhr2) and steroidogenesis (Cyp19a1). These results indicate that perinatal exposure to a diet elevated in AGEs causes deficits in perinatal growth, pubertal onset, and reproductive organ development in female mice. Whether these findings translate to humans remains to be determined in future studies.


Subject(s)
Glycation End Products, Advanced/metabolism , Ovary/metabolism , Adiponectin/metabolism , Animals , Anti-Mullerian Hormone/metabolism , Female , Leptin/metabolism , Mice , Ovary/drug effects , Reproduction/drug effects , Reproduction/physiology
9.
Syst Biol Reprod Med ; 66(4): 236-243, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32603611

ABSTRACT

Obesity, known to cause a systemic elevation in monocyte chemotactic protein-1 (MCP-1), adversely affects normal ovarian function. The aim of this study was to determine whether MCP-1 plays a role in ovarian dysfunction that is related to obesity induced by high-fat (HF) diet intake. Wild type (WT) C57BL/6J mice were fed either normal chow (NC) (Group 1, control group) or HF diet (Group 2). To assess whether MCP-1 is involved in HF-diet-induced ovarian dysfunction, MCP-1 knock-out mice were fed HF diet (Group 3). Body weight, body fat composition, number of oocytes collected following ovarian superovulation with gonadotropins, ovarian macrophage markers and expression of genes important in folliculogenesis and steroidogenesis were quantified in the 3 groups of animals. Animals in Group 2 gained significant body weight and body mass, produced the fewest number of oocytes following superovulation, and had significant alterations in ovarian genes involved in folliculogenesis and steroidogenesis as well as genes involved in inflammation. Although animals in Group 3 had the highest body weight and body fat composition, they produced similar number of oocytes compared to animals in Group 1 but had different ovarian gene expression compared to Group 2. These findings suggest that MCP-1 gene knockout could reverse some of the adverse effects of obesity induced by HF diet intake. Future studies assessing ovarian histology in MCP-1 knock out mouse model will confirm our findings. MCP-1 inhibition could represent a future therapeutic target to protect ovarian health from the adverse effects of HF diet ingestion.


Subject(s)
Chemokine CCL2/metabolism , Diet, High-Fat/adverse effects , Obesity/etiology , Ovarian Diseases/etiology , Animals , Chemokine CCL2/genetics , Female , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/complications , Obesity/metabolism , Obesity/prevention & control , Ovarian Diseases/metabolism , Ovarian Follicle/physiology , RNA, Messenger/metabolism , Steroids/metabolism
10.
Diabetes ; 69(8): 1650-1661, 2020 08.
Article in English | MEDLINE | ID: mdl-32444367

ABSTRACT

An adverse maternal in utero and lactation environment can program offspring for increased risk for metabolic disease. The aim of this study was to determine whether N-acetylcysteine (NAC), an anti-inflammatory antioxidant, attenuates programmed susceptibility to obesity and insulin resistance in offspring of mothers on a high-fat diet (HFD) during pregnancy. CD1 female mice were acutely fed a standard breeding chow or HFD. NAC was added to the drinking water (1 g/kg) of the treatment cohorts from embryonic day 0.5 until the end of lactation. NAC treatment normalized HFD-induced maternal weight gain and oxidative stress, improved the maternal lipidome, and prevented maternal leptin resistance. These favorable changes in the in utero environment normalized postnatal growth, decreased white adipose tissue (WAT) and hepatic fat, improved glucose and insulin tolerance and antioxidant capacity, reduced leptin and insulin, and increased adiponectin in HFD offspring. The lifelong metabolic improvements in the offspring were accompanied by reductions in proinflammatory gene expression in liver and WAT and increased thermogenic gene expression in brown adipose tissue. These results, for the first time, provide a mechanistic rationale for how NAC can prevent the onset of metabolic disease in the offspring of mothers who consume a typical Western HFD.


Subject(s)
Acetylcysteine/therapeutic use , Diet, High-Fat/adverse effects , Metabolic Diseases/drug therapy , Metabolic Diseases/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adiposity/drug effects , Animals , Antioxidants/metabolism , Body Temperature , Calorimetry, Indirect , Female , Glucose Tolerance Test , Inflammation/drug therapy , Inflammation/metabolism , Injections, Intraperitoneal , Insulin Resistance , Male , Mice , Weight Gain/drug effects
11.
Mol Cell Endocrinol ; 510: 110826, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32339649

ABSTRACT

Nutrition is an important source of exogenous AGEs and thermally processed foods present in western-style diets contain a large amount of these pro-inflammatory AGEs. Additionally, the intake of dietary AGEs could upregulate ovarian gene expression of inflammatory macrophage markers. The objective of this study was to investigate the effect of diet rich in AGEs on estrous cyclicity and ovarian function in a mouse model. Six-week old C57BL/6 J female mice were randomly subjected to either a diet low in AGEs (L-AGE) or a diet high in AGEs (H-AGE) for a total of 13 weeks. Experiments performed included daily vaginal smears to assess estrous cyclicity, ovarian superovulation with gonadotropins to assess the number of oocytes released, whole ovarian tissue mRNA quantification by RT-PCR to quantify genes involved in folliculogenesis, steroidogenesis, and macrophage markers, and ovarian morphology for follicle count. Outcome measures included estrous cyclicity, number of oocytes following superovulation, expression of genes involved in folliculogenesis, steroidogenesis, and macrophage infiltration as well as the number of primordial, primary, secondary, antral follicles and corpora lutea. Compared to mice on L-AGE diet, mice on H-AGE spent significantly longer time in the diestrus phase, had similar number of oocytes released following ovarian superovulation, and showed significant alterations in genes involved in steroidogenesis (increase in Star mRNA expression levels) and folliculogenesis (increase in Gdf-9 and Fshr mRNA expression levels). Mouse macrophage marker F4/80 mRNA expression was upregulated in mice on H-AGE diet compared to mice on L-AGE diet. Finally, mice on H-AGE diet had significantly fewer corpora lutea in their ovaries. These results indicate that the ingestion of high amounts of dietary AGEs could disrupt folliculogenesis and steroidogenesis that might lead to abnormal estrous cyclicity. Intake of dietary AGEs could also upregulate ovarian gene expression of inflammatory macrophage markers.


Subject(s)
Diet , Glycation End Products, Advanced/adverse effects , Ovary/physiology , Animals , Biomarkers/metabolism , Body Composition/drug effects , Body Weight/drug effects , Corpus Luteum/drug effects , Estrous Cycle/drug effects , Female , Gene Expression Regulation/drug effects , Gonadotropins/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Oocytes/drug effects , Organogenesis/drug effects , Organogenesis/genetics , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Ovary/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Steroids/biosynthesis , Superovulation/drug effects
12.
Am J Pathol ; 189(11): 2246-2257, 2019 11.
Article in English | MEDLINE | ID: mdl-31430466

ABSTRACT

The mechanism by which poor maternal nutrition can affect the long-term health of offspring is poorly understood. In mice, we previously found that maternal high-fat diet (HFD) exposure results in reduced fetal growth regardless of maternal genotype. We tested our hypothesis that maternal HFD-induced inflammation contributes to metabolic disease susceptibility of the offspring via alterations in the placenta. The effect of maternal genotype, diet, and treatment with the anti-inflammatory compound N-acetylcysteine (NAC) on placental morphologic features was investigated. Placentas from wild-type dams maintained on a HFD but not those heterozygous (+/-) for Glut4 (Slc2a4) on the same diet had an increase in decidual inflammation and vasculopathy occurring together. NAC administration resulted in amelioration of HFD-induced decidual vasculopathy independent of offspring genotype and sex. Consistent with these morphologic improvements, placentas from HFD dams treated with NAC had decreased mRNA and immunostaining of IL-1ß and monocyte chemoattractant protein-1, decreased mRNA of inflammatory genes, and increased mRNA of Vegfa. These results strongly suggest consumption of an HFD results in vascular changes in placenta reflected by alterations in expression of pivotal vascular developmental markers and inflammatory genes all of which are ameliorated by NAC. These placental changes play a key role in the increased programed metabolic disease of HFD-exposed offspring.


Subject(s)
Acetylcysteine/therapeutic use , Diet, High-Fat/adverse effects , Inflammation/prevention & control , Placenta/drug effects , Pregnancy Complications/prevention & control , Vascular Diseases/prevention & control , Animals , Disease Models, Animal , Female , Inflammation/complications , Inflammation/pathology , Male , Maternal Nutritional Physiological Phenomena/physiology , Mice , Mice, Transgenic , Placenta/pathology , Pregnancy , Pregnancy Complications/etiology , Vascular Diseases/complications , Vascular Diseases/pathology
14.
Med Sci Sports Exerc ; 51(6): 1116-1125, 2019 06.
Article in English | MEDLINE | ID: mdl-30694975

ABSTRACT

INTRODUCTION: Follistatin (FST) is a protein with numerous biological roles and was recently identified as an exercise-inducible hepatokine; however, the signals that regulate this are not well understood. The purpose of this study was to delineate potential endocrine factors that may regulate hepatic FST at rest and during exercise. METHODS: This study used four experiments. First, male and female C57BL/6J mice remained sedentary or were subjected to a single bout of exercise at moderate or exhaustive intensity with liver collected immediately post. Second, mice were injected with glucagon (1 mg·kg, 60 min), epinephrine (2 mg·kg, 30 min), glucagon then epinephrine, or saline. Third, mice were pretreated with propranolol (20-60 mg·kg, 30 min) before epinephrine injection. Fourth, glucagon receptor wild type (Gcgr) or knockout (Gcgr) mice were pretreated with saline or propranolol (20 mg·kg, 30 min) and were subjected to a single bout of exhaustive exercise with liver collected immediately post or after 2 h recovery. In all experiments liver FST mRNA expression was measured, and in experiment four FST protein content was measured. RESULTS: A single bout of treadmill exercise performed at an exhaustive but not moderate-intensity increased FST expression, as did injection of glucagon or epinephrine alone and when combined. Pretreatment of mice with propranolol attenuated the epinephrine-induced increase in FST expression. The exercise-induced increase in FST expression was attenuated in Gcgr mice, with no effect of propranolol. Gcgr mice had higher protein content of FST, but there was no effect of exercise or propranolol. CONCLUSIONS: These data suggest that both glucagon and epinephrine regulate hepatic FST expression at rest; however, only glucagon is required for the exercise-induced increase.


Subject(s)
Epinephrine/physiology , Follistatin/metabolism , Glucagon/physiology , Liver/metabolism , Physical Conditioning, Animal , Rest , Adrenergic beta-Antagonists/pharmacology , Animals , Epinephrine/administration & dosage , Epinephrine/antagonists & inhibitors , Female , Gene Expression , Glucagon/administration & dosage , Injections , Male , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Propranolol/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism
15.
FASEB J ; 33(4): 4824-4835, 2019 04.
Article in English | MEDLINE | ID: mdl-30615494

ABSTRACT

Various endocrine factors contribute to cold-induced white adipose tissue (WAT) browning, but glucagon has largely been ignored. The purpose of the current investigation was to determine if glucagon was required for the effects of cold on WAT browning. Utilizing whole-body glucagon receptor knockout (Gcgr-/-) mice and their wild-type (WT) littermate controls, we examined the response of inguinal WAT (iWAT) and interscapular brown adipose tissue (BAT) to an acute (48 h) cold stress or challenge with the ß3-adrenergic agonist CL316,243. The effects of glucagon alone on the induction of thermogenic genes in adipose tissue from C57BL6/J mice were also examined. Gcgr-/- mice displayed modest increases in indices of browning at room temperature while displaying a blunted induction of Ucp1, Cidea, and Ffg21 mRNA expression in iWAT following cold exposure. Similarly, cold induced increases in mitochondrial DNA copy number, and the protein content of mitochondrial respiratory chain complexes, UCP1, and PGC1α were attenuated in iWAT from Gcgr-/- mice. In BAT, the induction of thermogenic markers following cold exposure was reduced, but the effect was less pronounced than in iWAT. Glucagon treatment increased the expression of thermogenic genes in both iWAT and BAT of C57BL6/J mice. In response to CL316,243, circulating fatty acids, glycerol, and the phosphorylation of hormone-sensitive lipase were attenuated in iWAT of Gcgr-/- mice. We provide evidence that glucagon is sufficient for the induction of thermogenic genes in iWAT, and the absence of intact glucagon signaling blunts the cold-induced browning of WAT, possibly due, in part, to impaired adrenergic signaling.-Townsend, L. K., Medak, K. D., Knuth, C. M., Peppler, W. T., Charron, M. J., Wright, D. C. Loss of glucagon signaling alters white adipose tissue browning.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Glucagon/metabolism , Receptors, Glucagon/metabolism , Adipose Tissue/metabolism , Animals , Dioxoles/pharmacology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Glucagon/genetics , Signal Transduction/drug effects , Signal Transduction/physiology
16.
Mol Metab ; 16: 45-54, 2018 10.
Article in English | MEDLINE | ID: mdl-29937214

ABSTRACT

OBJECTIVE: Therapeutic interventions that improve glucose homeostasis such as attenuation of glucagon receptor (Gcgr) signaling and bariatric surgery share common metabolic features conserved in mice and humans. These include increased circulating levels of bile acids (BA) and the proglucagon-derived peptides (PGDPs), GLP-1 and GLP-2. Whether BA acting through TGR5 (Gpbar1) increases PGDP levels in these scenarios has not been examined. Furthermore, although the importance of GLP-1 action has been interrogated in Gcgr-/- mice and after bariatric surgery, whether GLP-2 contributes to the metabolic benefits of these interventions is not known. METHODS: To assess whether BA acting through Gpbar1 mediates improved glucose homeostasis in Gcgr-/- mice we generated and characterized Gcgr-/-:Gpbar1-/- mice. The contribution of GLP-2 receptor (GLP-2R) signaling to intestinal and metabolic adaptation arising following loss of the Gcgr was studied in Gcgr-/-:Glp2r-/- mice. The role of the GLP-2R in the metabolic improvements evident after bariatric surgery was studied in high fat-fed Glp2r-/- mice subjected to vertical sleeve gastrectomy (VSG). RESULTS: Circulating levels of BA were markedly elevated yet similar in Gcgr-/-:Gpbar1+/+ vs. Gcgr-/-:Gpbar1-/- mice. Loss of GLP-2R lowered levels of BA in Gcgr-/- mice. Gcgr-/-:Glp2r-/- mice also exhibited shifts in the proportion of circulating BA species. Loss of Gpbar1 did not impact body weight, intestinal mass, or glucose homeostasis in Gcgr-/- mice. In contrast, small bowel growth was attenuated in Gcgr-/-:Glp2r-/- mice. The improvement in glucose tolerance, elevated circulating levels of GLP-1, and glucose-stimulated insulin levels were not different in Gcgr-/-:Glp2r+/+ vs. Gcgr-/-:Glp2r-/- mice. Similarly, loss of the GLP-2R did not attenuate the extent of weight loss and improvement in glucose control after VSG. CONCLUSIONS: These findings reveal that GLP-2R controls BA levels and relative proportions of BA species in Gcgr-/- mice. Nevertheless, the GLP-2R is not essential for i) control of body weight or glucose homeostasis in Gcgr-/- mice or ii) metabolic improvements arising after VSG in high fat-fed mice. Furthermore, despite elevations of circulating levels of BA, Gpbar1 does not mediate elevated levels of PGDPs or major metabolic phenotypes in Gcgr-/- mice. Collectively these findings refine our understanding of the relationship between Gpbar1, elevated levels of BA, PGDPs, and the GLP-2R in amelioration of metabolic derangements arising following loss of Gcgr signaling or after vertical sleeve gastrectomy.


Subject(s)
Bile Acids and Salts/metabolism , Glucagon-Like Peptide-2 Receptor/metabolism , Receptors, Glucagon/metabolism , Animals , Blood Glucose/metabolism , Body Weight/physiology , Diet, High-Fat , Gastrectomy/methods , Glucagon , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 2/metabolism , Glucagon-Like Peptide-1 Receptor/genetics , Glucose/metabolism , Glucose Tolerance Test , Homeostasis/physiology , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proglucagon/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Glucagon/genetics , Signal Transduction , Weight Loss/physiology
17.
Am J Reprod Immunol ; 80(3): e12868, 2018 09.
Article in English | MEDLINE | ID: mdl-29736947

ABSTRACT

PROBLEM: Complications from prematurity are the leading cause of death among children under 5 years of age. Although clinical studies have shown a positive correlation between maternal high-fat diet (HFD) and preterm birth (PTB), the underlying mechanisms remain to be elucidated. Furthermore, it remains unclear how fatty acid type influences the effects of bacterial endotoxins. METHOD OF STUDY: HTR-8/SVneo trophoblasts were cultured in either 0.5 mmol L-1 palmitic acid (PA) or linoleic acid (LA) in the absence or presence of 100 µg mL-1 of lipopolysaccharide (LPS) or lipoteichoic acid (LTA). Murine placental explants were cultured in either 2 mmol L-1 PA or LA, and cell viability, total antioxidant capacity (TAC), lipid peroxidation, H2 O2 , heme oxygenase-1 (HO-1), and nuclear erythroid 2-related factor 2 (Nrf-2) and nuclear factor-kappa light-chain enhancer of activated B cells (NF-κB) transcription factor activity assays were assessed. RESULTS: Palmitic acid significantly (i) increased cell death, (ii) decreased TAC, and (iii) increased lipid peroxidation; but did not significantly increase HO-1. In contrast, LA maintained cell viability and significantly increased TAC and HO-1. In addition, incubating placental explants with PA significantly increased NF-κB activity. Co-incubating cells with PA and LPS or LTA significantly potentiated H2 O2 production and increased lipid peroxidation. Co-incubating cells with PA and LTA synergistically impaired TAC, and LTA decreased TAC more so than LPS. Co-incubation with PA/LA and LPS/LTA decreased HO-1 levels compared to treatment with either fatty acid alone. CONCLUSION: Our findings suggest that saturated and unsaturated fats differentially regulate placental viability, antioxidant capacity, and inflammation and the actions of gram-positive and gram-negative endotoxins.


Subject(s)
Linoleic Acid/metabolism , Palmitic Acid/metabolism , Placenta/physiology , Premature Birth/metabolism , Trophoblasts/physiology , Animals , Antioxidants/metabolism , Cell Survival , Cells, Cultured , Female , Humans , Lipid Peroxidation , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Oxidative Stress , Pregnancy
18.
Am J Physiol Endocrinol Metab ; 314(1): E93-E103, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28978545

ABSTRACT

Glucagon secreted from the pancreatic alpha-cells is essential for regulation of blood glucose levels. However, glucagon may play an equally important role in the regulation of amino acid metabolism by promoting ureagenesis. We hypothesized that disruption of glucagon receptor signaling would lead to an increased plasma concentration of amino acids, which in a feedback manner stimulates the secretion of glucagon, eventually associated with compensatory proliferation of the pancreatic alpha-cells. To address this, we performed plasma profiling of glucagon receptor knockout ( Gcgr-/-) mice and wild-type (WT) littermates using liquid chromatography-mass spectrometry (LC-MS)-based metabolomics, and tissue biopsies from the pancreas were analyzed for islet hormones and by histology. A principal component analysis of the plasma metabolome from Gcgr-/- and WT littermates indicated amino acids as the primary metabolic component distinguishing the two groups of mice. Apart from their hyperaminoacidemia, Gcgr-/- mice display hyperglucagonemia, increased pancreatic content of glucagon and somatostatin (but not insulin), and alpha-cell hyperplasia and hypertrophy compared with WT littermates. Incubating cultured α-TC1.9 cells with a mixture of amino acids (Vamin 1%) for 30 min and for up to 48 h led to increased glucagon concentrations (~6-fold) in the media and cell proliferation (~2-fold), respectively. In anesthetized mice, a glucagon receptor-specific antagonist (Novo Nordisk 25-2648, 100 mg/kg) reduced amino acid clearance. Our data support the notion that glucagon secretion and hepatic amino acid metabolism are linked in a close feedback loop, which operates independently of normal variations in glucose metabolism.


Subject(s)
Amino Acids/adverse effects , Amino Acids/blood , Cell Communication , Glucagon-Secreting Cells/physiology , Hepatocytes/physiology , Receptors, Glucagon/genetics , Animals , Cell Communication/drug effects , Cell Communication/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Electrolytes/adverse effects , Electrolytes/blood , Female , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/pathology , Glucose/adverse effects , Hepatocytes/drug effects , Hyperplasia/genetics , Hyperplasia/metabolism , Hyperplasia/pathology , Liver/drug effects , Liver/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics , Solutions/adverse effects
19.
Endocrinology ; 158(9): 2860-2872, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28911167

ABSTRACT

Exposure to a high-fat (HF) diet in utero is associated with increased incidence of cardiovascular disease, diabetes, and metabolic syndrome later in life. However, the molecular basis of this enhanced susceptibility for metabolic disease is poorly understood. Gene expression microarray and genome-wide DNA methylation analyses of mouse liver revealed that exposure to a maternal HF milieu activated genes of immune response, inflammation, and hepatic dysfunction. DNA methylation analysis revealed 3360 differentially methylated loci, most of which (76%) were hypermethylated and distributed preferentially to hotspots on chromosomes 4 [atherosclerosis susceptibility quantitative trait loci (QTLs) 1] and 18 (insulin-dependent susceptibility QTLs 21). Interestingly, we found six differentially methylated genes within these hotspot QTLs associated with metabolic disease that maintain altered gene expression into adulthood (Arhgef19, Epha2, Zbtb17/Miz-1, Camta1 downregulated; and Ccdc11 and Txnl4a upregulated). Most of the hypermethylated genes in these hotspots are associated with cardiovascular system development and function. There were 140 differentially methylated genes that showed a 1.5-fold increase or decrease in messenger RNA levels. Many of these genes play a role in cell signaling pathways associated with metabolic disease. Of these, metalloproteinase 9, whose dysregulation plays a key role in diabetes, obesity, and cardiovascular disease, was upregulated 1.75-fold and hypermethylated in the gene body. In summary, exposure to a maternal HF diet causes DNA hypermethylation, which is associated with long-term gene expression changes in the liver of exposed offspring, potentially contributing to programmed development of metabolic disease later in life.


Subject(s)
DNA Methylation , Diet, High-Fat , Gene Expression Regulation , Liver/metabolism , Maternal Nutritional Physiological Phenomena , Metabolic Syndrome/etiology , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/metabolism , Animals , Body Weight/genetics , Female , Male , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Mice , Pregnancy , Sex Characteristics
20.
Kidney Blood Press Res ; 42(3): 468-482, 2017.
Article in English | MEDLINE | ID: mdl-28750406

ABSTRACT

BACKGROUND/AIMS: Altered nutrients during the in utero (IU) and/or lactation (L) period predispose offspring to cardio-renal diseases in adulthood. This study investigates the effect of a high fat diet (HFD) fed to female mice during IU/L on gene expression patterns associated with heart and kidney failure and hypertension in male offspring. METHODS: Female wild type (WT) mice were fed either a HFD or control chow (C) prior to mating with males with a genetic heterozygous deletion of GLUT4 (G4+/-, a model of peripheral insulin resistance and hypertension) and throughout IU/L. After weaning male offspring were placed on a standard rodent chow until 24 weeks of age. RESULTS: All offspring exposed to a maternal HFD showed increased heart and kidney weight and reduced cardiac insulin responsiveness. G4+/- offspring on a HFD displayed early hypertension associated with increased renal gene expression of renin and the AT1- receptors compared to G4+/- on a C diet. This group showed decreased cardiac expression of key genes involved in fatty acid oxidation compared to WT on a C diet. CONCLUSIONS: These results indicate an interaction between a HFD diet and genotype during early life development that can enhance susceptibility to cardio-renal diseases later in life.


Subject(s)
Diet, High-Fat/adverse effects , Genotype , Glucose Transporter Type 4/genetics , Lactation , Animals , Female , Genetic Predisposition to Disease , Heart Diseases/genetics , Hypertension , Kidney Diseases/genetics , Male , Mice , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...