Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep ; 35(13): 109301, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34192532

ABSTRACT

Hepatic lipid accumulation is a hallmark of type II diabetes (T2D) associated with hyperinsulinemia, insulin resistance, and hyperphagia. Hepatic synthesis of GABA, catalyzed by GABA-transaminase (GABA-T), is upregulated in obese mice. To assess the role of hepatic GABA production in obesity-induced metabolic and energy dysregulation, we treated mice with two pharmacologic GABA-T inhibitors and knocked down hepatic GABA-T expression using an antisense oligonucleotide. Hepatic GABA-T inhibition and knockdown decreased basal hyperinsulinemia and hyperglycemia and improved glucose intolerance. GABA-T knockdown improved insulin sensitivity assessed by hyperinsulinemic-euglycemic clamps in obese mice. Hepatic GABA-T knockdown also decreased food intake and induced weight loss without altering energy expenditure in obese mice. Data from people with obesity support the notion that hepatic GABA production and transport are associated with serum insulin, homeostatic model assessment for insulin resistance (HOMA-IR), T2D, and BMI. These results support a key role for hepatocyte GABA production in the dysfunctional glucoregulation and feeding behavior associated with obesity.


Subject(s)
Hyperphagia/metabolism , Hyperphagia/physiopathology , Liver/metabolism , Liver/physiopathology , Obesity/metabolism , Obesity/physiopathology , gamma-Aminobutyric Acid/metabolism , 4-Aminobutyrate Transaminase/metabolism , Animals , Biomarkers/metabolism , Diet, High-Fat , Energy Metabolism , Feeding Behavior , Glucose/metabolism , Glucose Clamp Technique , Homeostasis , Humans , Hyperinsulinism/complications , Hyperinsulinism/metabolism , Hyperinsulinism/physiopathology , Hyperphagia/complications , Insulin Resistance , Liver/innervation , Male , Mice, Inbred C57BL , Mice, Obese , Obesity/complications , Vagotomy , Vagus Nerve/physiopathology
2.
Cell Rep ; 35(13): 109298, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34192533

ABSTRACT

Hepatic lipid accumulation in obesity correlates with the severity of hyperinsulinemia and systemic insulin resistance. Obesity-induced hepatocellular lipid accumulation results in hepatocyte depolarization. We have established that hepatocyte depolarization depresses hepatic afferent vagal nerve firing, increases GABA release from liver slices, and causes hyperinsulinemia. Preventing hepatic GABA release or eliminating the ability of the liver to communicate to the hepatic vagal nerve ameliorates the hyperinsulinemia and insulin resistance associated with diet-induced obesity. In people with obesity, hepatic expression of GABA transporters is associated with glucose infusion and disposal rates during a hyperinsulinemic euglycemic clamp. Single-nucleotide polymorphisms in hepatic GABA re-uptake transporters are associated with an increased incidence of type 2 diabetes mellitus. Herein, we identify GABA as a neuro-hepatokine that is dysregulated in obesity and whose release can be manipulated to mute or exacerbate the glucoregulatory dysfunction common to obesity.


Subject(s)
Hepatocytes/metabolism , Insulin Resistance , Insulin/blood , Liver/metabolism , Membrane Potentials , gamma-Aminobutyric Acid/metabolism , Animals , Blood Glucose/metabolism , Diet , Female , Humans , Hyperinsulinism/blood , Male , Mice, Inbred C57BL , Middle Aged , Models, Biological , Obesity/blood , Vagotomy , Vagus Nerve/physiopathology
3.
Int J Mol Sci ; 22(8)2021 Apr 13.
Article in English | MEDLINE | ID: mdl-33924461

ABSTRACT

Signaling through GPR109a, the putative receptor for the endogenous ligand ß-OH butyrate, inhibits adipose tissue lipolysis. Niacin, an anti-atherosclerotic drug that can induce insulin resistance, activates GPR109a at nM concentrations. GPR109a is not essential for niacin to improve serum lipid profiles. To better understand the involvement of GPR109a signaling in regulating glucose and lipid metabolism, we treated GPR109a wild-type (+/+) and knockout (-/-) mice with repeated overnight injections of saline or niacin in physiological states characterized by low (ad libitum fed) or high (16 h fasted) concentrations of the endogenous ligand, ß-OH butyrate. In the fed state, niacin increased expression of apolipoprotein-A1 mRNA and decreased sterol regulatory element-binding protein 1 mRNA independent of genotype, suggesting a possible GPR109a independent mechanism by which niacin increases high-density lipoprotein (HDL) production and limits transcriptional upregulation of lipogenic genes. Niacin decreased fasting serum non-esterified fatty acid concentrations in both GPR109a +/+ and -/- mice. Independent of GPR109a expression, niacin blunted fast-induced hepatic triglyceride accumulation and peroxisome proliferator-activated receptor α mRNA expression. Although unaffected by niacin treatment, fasting serum HDL concentrations were lower in GPR109a knockout mice. Surprisingly, GPR109a knockout did not affect glucose or lipid homeostasis or hepatic gene expression in either fed or fasted mice. In turn, GPR109a does not appear to be essential for the metabolic response to the fasting ketogenic state or the acute effects of niacin.


Subject(s)
Fasting , Feeding Behavior , Liver/metabolism , Niacin/pharmacology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Cholesterol/metabolism , Feeding Behavior/drug effects , Gene Expression Regulation/drug effects , Glucose/metabolism , Glycogen/metabolism , Homeostasis/drug effects , Liver/drug effects , Male , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects
4.
Am J Physiol Gastrointest Liver Physiol ; 316(5): G623-G631, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30767679

ABSTRACT

Ketosis is a metabolic adaptation to fasting, nonalcoholic fatty liver disease (NAFLD), and prolonged exercise. ß-OH butyrate acts as a transcriptional regulator and at G protein-coupled receptors to modulate cellular signaling pathways in a hormone-like manner. While physiological ketosis is often adaptive, chronic hyperketonemia may contribute to the metabolic dysfunction of NAFLD. To understand how ß-OH butyrate signaling affects hepatic metabolism, we compared the hepatic fasting response in control and 3-hydroxy-3-methylglutaryl-CoA synthase II (HMGCS2) knockdown mice that are unable to elevate ß-OH butyrate production. To establish that rescue of ketone metabolic/endocrine signaling would restore the normal hepatic fasting response, we gave intraperitoneal injections of ß-OH butyrate (5.7 mmol/kg) to HMGCS2 knockdown and control mice every 2 h for the final 9 h of a 16-h fast. In hypoketonemic, HMGCS2 knockdown mice, fasting more robustly increased mRNA expression of uncoupling protein 2 (UCP2), a protein critical for supporting fatty acid oxidation and ketogenesis. In turn, exogenous ß-OH butyrate administration to HMGCS2 knockdown mice decreased fasting UCP2 mRNA expression to that observed in control mice. Also supporting feedback at the transcriptional level, ß-OH butyrate lowered the fasting-induced expression of HMGCS2 mRNA in control mice. ß-OH butyrate also regulates the glycemic response to fasting. The fast-induced fall in serum glucose was absent in HMGCS2 knockdown mice but was restored by ß-OH butyrate administration. These data propose that endogenous ß-OH butyrate signaling transcriptionally regulates hepatic fatty acid oxidation and ketogenesis, while modulating glucose tolerance. NEW & NOTEWORTHY Ketogenesis regulates whole body glucose metabolism and ß-OH butyrate produced by the liver feeds back to inhibit hepatic ß-oxidation and ketogenesis during fasting.


Subject(s)
Fasting/physiology , Fatty Acids/metabolism , Ketone Bodies/biosynthesis , Ketones/metabolism , Liver/metabolism , Adaptation, Physiological , Animals , Blood Glucose/metabolism , Butyrates/metabolism , Gene Expression Regulation , Hydroxymethylglutaryl-CoA Synthase/metabolism , Ketosis/metabolism , Mice , Mice, Knockout , Oxidation-Reduction , Signal Transduction , Uncoupling Protein 2/metabolism
5.
Evol Ecol Res ; 16(3): 203-221, 2014.
Article in English | MEDLINE | ID: mdl-31354387

ABSTRACT

BACKGROUND: The evolution of mortal somatic cells was a critical step in the evolution of complex body plans and the major radiations of multicellular life. In the volvocine green algae, somatic cells are hypothesized to mitigate an increasing cost of reproduction as colony size increases, primarily by providing motility to the colony during reproduction. QUESTIONS: Does selection on colony size cause an evolutionary response in proportion of somatic cells? Does the effect of selection on colony size differ in environments that differ in the importance of motility? METHODS: We subjected an outcrossed population of the volvocine alga Pleodorina starrii to selection on colony size in still and mixed environments. After approximately 40 generations with periodic selection, we estimated the relationship between colony size and proportion of soma in evolved colonies from both environments. RESULTS: In the largest size category, colonies selected in the still environment (in which motility is hypothesized to be more important) had a higher proportion of soma than those from the mixed environment. Within-strain variation in cell number was surprisingly large: up to 16-fold for some genotypes. The positive among-species relationship between colony size and proportion of soma was paralleled within the larger (16- to 64-celled) colonies of P. starrii, but not within the smaller (4- and 8-celled) colonies, which had the highest proportions of soma, suggesting the existence of an evolutionary constraint preventing optimization of soma in the smallest size classes.

SELECTION OF CITATIONS
SEARCH DETAIL
...