Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Front Integr Neurosci ; 18: 1426219, 2024.
Article in English | MEDLINE | ID: mdl-39131599

ABSTRACT

The relationship between physical activity levels and feeding behaviors has been a focus of preclinical research for decades, yet this interaction has only recently been explored for potential sex differences. The aim of the present study was to isolate sex-dependent effects of voluntary wheel running (RUN) vs. sedentary locked wheel (SED) home cage conditions on palatability-driven feeding behavior using a 2-diet choice task between standard chow and a high-fat diet. The sex-dependent effects of physical activity on feeding behavior were examined following a within-subject novel reversal design of physical activity conditions (i.e., RUN > SED > RUN), to assess temporal sensitivity of the interaction. Following the final 2 weeks of reestablished and sustained RUN vs. SED conditions in separate groups of both males and females, reward-related opioid and dopamine gene expression within the nucleus accumbens (Acb) brain region were analyzed. Results demonstrated that the initial RUN > SED transition led to sex-dependent effects of SED condition, as males increased, and females decreased their high fat consumption, compared to their respective high fat consumption during previous RUN condition phase. Following reintroduction to the RUN condition, males decreased, and females increased their high fat consumption, compared to their separate SED control group. Last, sex-dependent shifts in ventral striatal opioid- and dopamine-related gene expression were observed to parallel the behavioral effects. The major findings of the study reveal that SED and RUN home cage conditions shift palatability-driven feeding in the opposite direction for males and females, these effects are sensitive to reversal, and these sex-dependent feeding behaviors track sex-dependent changes to critical reward-related gene expression patterns in the Acb. Considering the present high rates of sedentary behavior and obesity, furthering our understanding of the interaction between physical activity (or lack thereof) and feeding behavior should be a priority, especially in the context of these divergent sex-dependent outcomes.

2.
Int J Mol Sci ; 25(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38892320

ABSTRACT

Declining estrogen (E2) leads to physical inactivity and adipose tissue (AT) dysfunction. Mechanisms are not fully understood, but E2's effects on dopamine (DA) activity in the nucleus accumbens (NAc) brain region may mediate changes in mood and voluntary physical activity (PA). Our prior work revealed that loss of E2 robustly affected NAc DA-related gene expression, and the pattern correlated with sedentary behavior and visceral fat. The current study used a new transgenic mouse model (D1ERKO) to determine whether the abolishment of E2 receptor alpha (ERα) signaling within DA-rich brain regions affects PA and AT metabolism. Adult male and female wild-type (WT) and D1ERKO (KD) mice were assessed for body composition, energy intake (EE), spontaneous PA (SPA), and energy expenditure (EE); underwent glucose tolerance testing; and were assessed for blood biochemistry. Perigonadal white AT (PGAT), brown AT (BAT), and NAc brain regions were assessed for genes and proteins associated with DA, E2 signaling, and metabolism; AT sections were also assessed for uncoupling protein (UCP1). KD mice had greater lean mass and EE (genotype effects) and a visible change in BAT phenotype characterized by increased UCP1 staining and lipid depletion, an effect seen only among females. Female KD had higher NAc Oprm1 transcript levels and greater PGAT UCP1. This group tended to have improved glucose tolerance (p = 0.07). NAc suppression of Esr1 does not appear to affect PA, yet it may directly affect metabolism. This work may lead to novel targets to improve metabolic dysfunction following E2 loss, possibly by targeting the NAc.


Subject(s)
Adipose Tissue , Energy Metabolism , Estrogen Receptor alpha , Nucleus Accumbens , Receptors, Dopamine D1 , Animals , Female , Male , Mice , Adipose Tissue/metabolism , Adipose Tissue, Brown/metabolism , Brain/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/genetics , Gene Knockdown Techniques , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nucleus Accumbens/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D1/genetics , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics
3.
Nutrients ; 15(2)2023 Jan 14.
Article in English | MEDLINE | ID: mdl-36678314

ABSTRACT

Postmenopausal women represent an important target population in need of preventative cardiometabolic approaches. The loss of estrogen following the menopause eliminates protections against metabolic dysfunction, largely due to its role in the health and function of adipose tissue. In addition, some studies associate the menopause with reduced physical activity, which could potentially exacerbate the deleterious cardiometabolic risk profile accompanying the menopause. Meanwhile, exercise has adipocyte-specific effects that may alleviate the adverse impact of estrogen loss through the menopausal transition period and beyond. Exercise thus remains the best therapeutic agent available to mitigate menopause-associated metabolic dysfunction and represents a vital behavioral strategy to prevent and alleviate health decline in this population.


Subject(s)
Cardiovascular Diseases , Menopause , Female , Humans , Menopause/metabolism , Estrogens , Adipocytes , Exercise
4.
EBioMedicine ; 86: 104362, 2022 12.
Article in English | MEDLINE | ID: mdl-36371987
5.
Front Physiol ; 13: 920675, 2022.
Article in English | MEDLINE | ID: mdl-36213237

ABSTRACT

White adipose tissue (WAT) dysfunction independently predicts cardiometabolic disease, yet there is a lack of effective adipocyte-targeting therapeutics. B3AR agonists enhance adipocyte mitochondrial function and hold potential in this regard. Based on enhanced sensitivity to B3AR-mediated browning in estrogen receptor (ER)alpha-null mice, we hypothesized that ERß may enhance the WAT response to the B3AR ligand, CL316,243 (CL). Methods: Male and female wild-type (WT) and ERß DNA binding domain knock-out (ERßDBDKO) mice fed high-fat diet (HFD) to induce obesity were administered CL (1 mg/kg) daily for 2 weeks. Systemic physiological assessments of body composition (EchoMRI), bioenergetics (metabolic chambers), adipocyte mitochondrial respiration (oroboros) and glucose tolerance were performed, alongside perigonadal (PGAT), subcutaneous (SQAT) and brown adipose tissue (BAT) protein expression assessment (Western blot). Mechanisms were tested in vitro using primary adipocytes isolated from WT mice, and from Esr2-floxed mice in which ERß was knocked down. Statistical analyses were performed using 2 × 2 analysis of variance (ANOVA) for main effects of genotype (G) and treatment (T), as well as GxT interactions; t-tests were used to determine differences between in vitro treatment conditions (SPSS V24). Results: There were no genotype differences in HFD-induced obesity or systemic rescue effects of CL, yet ERßDBDKO females were more sensitive to CL-induced increases in energy expenditure and WAT UCP1 induction (GxT, p < 0.05), which coincided with greater WAT B3AR protein content among the KO (G, p < 0.05). Among males, who were more insulin resistant to begin with (no genotype differences before treatment), tended to be more sensitive to CL-mediated reduction in insulin resistance. With sexes combined, basal WAT mitochondrial respiration trended toward being lower in the ERßDBDKO mice, but this was completely rescued by CL (p < 0.05). Confirming prior work, CL increased adipose tissue ERß protein (T, p < 0.05, all), an effect that was enhanced in WAT and BAT the female KO (GxT, p < 0.01). In vitro experiments indicated that an inhibitor of ERß genomic function (PHTPP) synergized with CL to further increase UCP1 mRNA (p = 0.043), whereas full ERß protein was required for UCP1 expression (p = 0.042). Conclusion: Full ERß activity appears requisite and stimulatory for UCP1 expression via a mechanism involving non-classical ERß signaling. This novel discovery about the role of ERß in adipocyte metabolism may have important clinical applications.

6.
Obesity (Silver Spring) ; 30(5): 1066-1078, 2022 05.
Article in English | MEDLINE | ID: mdl-35357089

ABSTRACT

OBJECTIVE: Endothelial nitric oxide synthase (eNOS) is a potential mediator of exercise-induced hepatic mitochondrial adaptations. METHODS: Here, male and female hepatocyte-specific eNOS knockout (eNOShep-/- ) and intact hepatic eNOS (eNOSfl/fl ) mice performed voluntary wheel-running exercise (EX) or remained in sedentary cage conditions for 10 weeks. RESULTS: EX resolved the exacerbated hepatic steatosis in eNOShep-/- male mice. Elevated hydrogen peroxide emission (~50% higher in eNOShep-/- vs. eNOSfl/fl mice) was completely ablated with EX. Interestingly, EX increased [1-14 C] palmitate oxidation in eNOSfl/fl male mice, but this was blunted in the eNOShep-/- male mice. eNOShep-/- mice had lower markers of the energy sensors AMP-activated protein kinase (AMPK)/phospho- (p)AMPK and mammalian target of rapamycin (mTOR) and p-mTOR, as well as the autophagy initiators serine/threonine-protein kinase ULK1 and pULK1, compared with eNOSfl/fl mice. Females showed elevated electron transport chain protein content and markers of mitochondrial biogenesis (transcription factor A, mitochondrial, peroxisome proliferator-activated receptor-gamma coactivator 1α). CONCLUSIONS: Collectively, this study demonstrates for the first time, to the authors' knowledge, the requirement of eNOS in hepatocytes in the EX-induced increases in hepatic fatty acid oxidation in male mice. Deletion of eNOS in hepatocytes also appears to impair the energy-sensing ability of the cell and inhibit the activation of the autophagy initiating factor ULK1. These data uncover the important and novel role of hepatocyte eNOS in EX-induced hepatic mitochondrial adaptations.


Subject(s)
AMP-Activated Protein Kinases , Nitric Oxide Synthase Type III , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy/genetics , Female , Hepatocytes/metabolism , Male , Mammals/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , TOR Serine-Threonine Kinases/metabolism
7.
Cells ; 10(12)2021 12 08.
Article in English | MEDLINE | ID: mdl-34943961

ABSTRACT

Beta-3 adrenergic receptor activation via exercise or CL316,243 (CL) induces white adipose tissue (WAT) browning, improves glucose tolerance, and reduces visceral adiposity. Our aim was to determine if sex or adipose tissue depot differences exist in response to CL. Daily CL injections were administered to diet-induced obese male and female mice for two weeks, creating four groups: male control, male CL, female control, and female CL. These groups were compared to determine the main and interaction effects of sex (S), CL treatment (T), and WAT depot (D). Glucose tolerance, body composition, and energy intake and expenditure were assessed, along with perigonadal (PGAT) and subcutaneous (SQAT) WAT gene and protein expression. CL consistently improved glucose tolerance and body composition. Female PGAT had greater protein expression of the mitochondrial uncoupling protein 1 (UCP1), while SQAT (S, p < 0.001) was more responsive to CL in increasing UCP1 (S×T, p = 0.011) and the mitochondrial biogenesis induction protein, PPARγ coactivator 1α (PGC1α) (S×T, p = 0.026). Females also displayed greater mitochondrial OXPHOS (S, p < 0.05) and adiponectin protein content (S, p < 0.05). On the other hand, male SQAT was more responsive to CL in increasing protein levels of PGC1α (S×T, p = 0.046) and adiponectin (S, p < 0.05). In both depots and in both sexes, CL significantly increased estrogen receptor beta (ERß) and glucose-related protein 75 (GRP75) protein content (T, p < 0.05). Thus, CL improves systemic and adipose tissue-specific metabolism in both sexes; however, sex differences exist in the WAT-specific effects of CL. Furthermore, across sexes and depots, CL affects estrogen signaling by upregulating ERß.


Subject(s)
Adipose Tissue, Brown/metabolism , HSP70 Heat-Shock Proteins/genetics , Membrane Proteins/genetics , PPAR gamma/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Uncoupling Protein 1/genetics , Adipose Tissue/metabolism , Adipose Tissue, Brown/growth & development , Adipose Tissue, White/metabolism , Animals , Body Composition/genetics , Dioxoles/pharmacology , Energy Metabolism/genetics , Estrogen Receptor beta/genetics , Estrogens/genetics , Estrogens/metabolism , Female , Glucose Tolerance Test , Humans , Male , Mice , Mitochondria/genetics , Mitochondria/metabolism , Receptors, Adrenergic, beta-3/genetics , Receptors, Adrenergic, beta-3/metabolism , Sex Characteristics
8.
Bone Rep ; 15: 101147, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34820485

ABSTRACT

Bisphenol-A (BPA) and bisphenol-S (BPS) are endocrine disrupting chemicals (EDCs) found primarily in plastics. Estrogen is a primary hormonal regulator of skeletal growth and development; however, the impact of gestational BPA or BPS exposure on skeletal health of offspring remains relatively unknown. Here, adult female mice were randomized into three treatment groups: 200 µg BPA/kg BW (BPA), 200 µg BPS/kg BW (BPS) or control (CON). Animals were then further randomized to exercising (EX) or sedentary (SED) groups. Treatment continued through mating, gestation, and lactation. One male offspring from each dam (n = 6-8/group) was assessed at 16 weeks of age to evaluate effects of EDC exposure on the adult skeleton. Cortical geometry of the mid-diaphysis and trabecular microarchitecture of the distal femur were assessed via micro-CT. Biomechanical strength and mineral apposition rate of the femoral diaphysis were assessed via three-point bending and dynamic histomorphometry, respectively. Two-factor ANOVA or ANCOVA were used to determine the effects of maternal EX and BPA or BPS on trabecular and cortical bone outcomes. Maternal EX led to a significant decrease in body fat percentage and bone stiffness, independent of EDC exposure. Offspring exposed to BPA had significantly lower trabecular bone volume, trabecular number, connectivity density, cortical thickness, and greater trabecular spacing compared to BPS or CON animals. In conclusion, gestational BPA, but not BPS, exposure negatively impacted trabecular microarchitecture and cortical geometry in adult male offspring. If these findings translate to humans, this could have significant public health impacts on expecting women or those seeking to become pregnant.

9.
Bone Rep ; 15: 101136, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34632005

ABSTRACT

Bisphenol-A (BPA) and bisphenol-S (BPS) are estrogen disrupting chemicals (EDCs) found in the environment and common household items. Estrogen is a primary hormonal regulator of bone growth and development; however, the impact of gestational BPA or BPS exposure on skeletal health of offspring remains relatively unknown. In this longitudinal study, adult female mice were randomized into three groups: 200 µg BPA/kg BW (BPA), 200 µg BPS/kg BW (BPS) or control (CON). Animals in each group were further randomized to exercise treatment (EX) or sedentary (SED) control, resulting in six overall groups. BPA/BPS/CON and EX/SED treatment were initiated prior to mating and continued through mating, gestation, and lactation. One female offspring from each dam (n = 6/group) was assessed at 17 weeks of age to evaluate effects of EDC exposure on the adult skeleton. Cortical geometry of the mid-diaphysis and trabecular microarchitecture of the distal femur were assessed via micro-computed tomography. Biomechanical strength and mineral apposition rate of the femoral diaphysis were assessed via three-point bending and dynamic histomorphometry, respectively. Sclerostin expression was measured using immunohistochemistry. Two-factor ANOVA or ANCOVA were used to determine the effects of maternal exercise and BPA or BPS exposure on trabecular and cortical bone outcomes, respectively. Consistent with prior studies, there were no significant differences in body weight, femoral length, cortical geometry, trabecular microarchitecture, or biomechanical strength between groups in female offspring. In conclusion, gestational BPA exposure and maternal exercise have minimal impact on skeletal outcomes in female adult offspring.

10.
J Sleep Res ; 30(6): e13381, 2021 12.
Article in English | MEDLINE | ID: mdl-33949729

ABSTRACT

Sleep restriction (SR) (<6 h) and physical activity (PA) are risk factors for obesity, but little work has examined the inter-related influences of both risk factors. In a free-living environment, 13 overweight/obese adults were sleep restricted for five nights to 6 h time-in-bed each night, with and without regular exercise (45 min/65% VO2 max; counterbalanced design). Two days of recovery sleep followed SR. Subjects were measured during a mixed meal tolerance test (MMT), resting metabolic rate, cognitive testing and fat biopsy (n=8). SR increased peak glucose response (+7.3 mg/dl, p = .04), elevated fasting non-esterified fatty acid (NEFA) concentrations (+0.1 mmol/L, p = .001) and enhanced fat oxidation (p < .001) without modifying step counts or PA intensity. Inclusion of daily exercise increased step count (+4,700 steps/day, p < .001) and decreased the insulin response to a meal (p = .01) but did not prevent the increased peak glucose response or elevated NEFA levels. The weekend recovery period improved fasting glucose (p = .02), insulin (p = .02), NEFA concentrations (p = .001) and HOMA-IR (p < .01) despite reduced steps (p < .01) and increased sedentary time (p < .01). Abdominal adipose tissue (AT) samples, obtained after baseline, SR and exercise, did not differ in lipolytic capacity following SR. Fatty acid synthase protein content tended to increase following SR (p = .07), but not following exercise. In a free-living setting, SR adversely affected circulating NEFAs, fuel oxidation and peak glucose response but did not directly affect glucose tolerance or AT lipolysis. SR-associated metabolic impairments were not mitigated by exercise, yet recovery sleep completely rescued its adverse effects on glucose metabolism.


Subject(s)
Blood Glucose , Sleep , Adult , Exercise , Glucose , Humans , Insulin , Obesity
11.
J Endocrinol ; 249(3): 223-237, 2021 05 24.
Article in English | MEDLINE | ID: mdl-33877054

ABSTRACT

Estrogen receptor ß (ERb), one of the two major estrogen receptors, acts via genomic and non-genomic signaling pathways to affect many metabolic functions, including mitochondrial biogenesis and respiration. This study assessed the effect of ERb classical genomic activity on adipocyte-specific and -systemic metabolic responses to wheel running exercise in a rodent model of menopause. Female mice lacking the ERb DNA-binding domain (ERbDBDKO, n = 20) and WT (n = 21) littermate controls were fed a high-fat diet (HFD), ovariectomized (OVX), and randomized to control (no running wheel) and exercise (running wheel access) groups and were followed for 8 weeks. Wheel running did not confer protection against metabolic dysfunction associated with HFD+OVX in either ERbDBDKO or WT mice, despite increased energy expenditure. Unexpectedly, in the ERbDBDKO group, wheel running increased fasting insulin and surrogate measures of insulin resistance, and modestly increased adipose tissue inflammatory gene expression (P ≤ 0.05). These changes were not accompanied by significant changes in adipocyte mitochondrial respiration. It was demonstrated for the first time that female WT OVX mice do experience exercise-induced browning of white adipose tissue, indicated by a robust increase in uncoupling protein 1 (UCP1) (P ≤ 0.05). However, KO mice were completely resistant to this effect, indicating that full ERb genomic activity is required for exercise-induced browning. The inability to upregulate UCP1 with exercise following OVX may have resulted in the increased insulin resistance observed in KO mice, a hypothesis requiring further investigation.


Subject(s)
Estrogen Receptor beta/metabolism , Motor Activity/physiology , Ovariectomy , Adipocytes/metabolism , Adipose Tissue, Brown/physiology , Adipose Tissue, White/physiology , Animals , Diet, High-Fat , Energy Metabolism , Estrogen Receptor beta/genetics , Female , Gene Expression Regulation , Genotype , Glucose/metabolism , Lipid Metabolism , Mice , Mice, Knockout
12.
Int J Mol Sci ; 22(4)2021 Feb 09.
Article in English | MEDLINE | ID: mdl-33572215

ABSTRACT

Estrogen receptor-α knockout (ERKO) in female, but not male, mice results in an impaired osteogenic response to exercise, but the mechanisms behind this ability in males are unknown. We explored the main and interactive effects of ERKO and exercise on cortical geometry, trabecular microarchitecture, biomechanical strength, and sclerostin expression in male mice. At 12 weeks of age, male C57BL/6J ERKO and WT animals were randomized into two groups: exercise treatment (EX) and sedentary (SED) controls, until 22 weeks of age. Cortical geometry and trabecular microarchitecture were measured via µCT; biomechanical strength was assessed via three-point bending; sclerostin expression was measured via immunohistochemistry. Two-way ANOVA was used to assess sclerostin expression and trabecular microarchitecture; two-way ANCOVA with body weight was used to assess cortical geometry and biomechanical strength. ERKO positively impacted trabecular microarchitecture, and exercise had little effect on these outcomes. ERKO significantly impaired cortical geometry, but exercise was able to partially reverse these negative alterations. EX increased cortical thickness regardless of genotype. There were no effects of genotype or exercise on sclerostin expression. In conclusion, male ERKO mice retain the ability to build bone in response to exercise, but altering sclerostin expression is not one of the mechanisms involved.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cortical Bone/growth & development , Estrogen Receptor alpha/genetics , Osteogenesis/physiology , Physical Conditioning, Animal/physiology , Animals , Bone Remodeling/physiology , Cortical Bone/diagnostic imaging , Cortical Bone/metabolism , Estrogen Receptor alpha/metabolism , Male , Mice , Mice, Knockout , Models, Animal , Running/physiology , X-Ray Microtomography
13.
Obesity (Silver Spring) ; 28(9): 1698-1707, 2020 09.
Article in English | MEDLINE | ID: mdl-32734695

ABSTRACT

OBJECTIVE: The aim of this study was to examine the effects of sex and menopausal status on depot-specific estrogen signaling in white adipose tissue (AT) in age-matched men and women with morbid obesity. METHODS: A total of 28 premenopausal women, 16 postmenopausal women, and 27 age-matched men undergoing bariatric surgery were compared for omental (OM) AT (OMAT) and abdominal subcutaneous (SQ) AT (SQAT) genes and proteins. RESULTS: With the exception of fasting nonesterified fatty acids being higher in women (P < 0.01), no differences were found in other indicators of glucose and lipid metabolism. In OMAT, estrogen receptor (ER) beta (ERß) levels were higher in older women than in younger women and older men (sex-age interaction, P < 0.01), and aromatase expression was higher in older men than in older women (P < 0.05). In SQAT, women had lower expression of ERß than men (P < 0.05). Protein content of ER alpha and ERß was highly correlated with the mitochondrial protein uncoupling protein 1 across sexes and ages (P < 0.001). Age increased SQ inflammatory gene expression in both sexes. CONCLUSIONS: In morbid obesity, sex and age affect AT ERs, lipid metabolism, mitochondrial uncoupling protein 1, and inflammatory expression in an AT depot-dependent manner. The SQAT immunometabolic profile is heavily influenced by age and menopause status, more so than OMAT.


Subject(s)
Adipose Tissue/metabolism , Obesity/metabolism , Receptors, Estrogen/metabolism , Adult , Age Factors , Aged , Female , Humans , Male , Middle Aged , Obesity/epidemiology , Sex Characteristics
14.
Horm Behav ; 121: 104719, 2020 05.
Article in English | MEDLINE | ID: mdl-32081742

ABSTRACT

Aromatase catalyzes conversion of testosterone to estradiol and is expressed in a variety of tissues, including the brain. Suppression of aromatase adversely affects metabolism and physical activity behavior, but mechanisms remain uncertain. The hypothesis tested herein was that whole body aromatase deletion would cause gene expression changes in the nucleus accumbens (NAc), a brain regulating motivated behaviors such as physical activity, which is suppressed with loss of estradiol. Metabolic and behavioral assessments were performed in male and female wild-type (WT) and aromatase knockout (ArKO) mice. NAc-specific differentially expressed genes (DEGs) were identified with RNAseq, and associations between the measured phenotypic traits were determined. Female ArKO mice had greater percent body fat, reduced spontaneous physical activity (SPA), consumed less energy, and had lower relative resting energy expenditure (REE) than WT females. Such differences were not observed in ArKO males. However, in both sexes, a top DEG was Pts, a gene encoding an enzyme necessary for catecholamine (e.g., dopamine) biosynthesis. In comparing male and female WT mice, top DEGs were related to sexual development/fertility, immune regulation, obesity, dopamine signaling, and circadian regulation. SPA correlated strongly with Per3, a gene regulating circadian function, thermoregulation, and metabolism (r = -0.64, P = .002), which also correlated with adiposity (r = 0.54, P = .01). In conclusion, aromatase ablation leads to gene expression changes in NAc, which may in turn result in reduced SPA and related metabolic abnormalities. These findings may have significance to post-menopausal women and those treated with an aromatase inhibitor.


Subject(s)
Aromatase/genetics , Motor Activity/genetics , Nucleus Accumbens/metabolism , Animals , Aromatase/metabolism , Energy Metabolism/genetics , Estradiol/metabolism , Female , Gene Expression , Gene Expression Regulation, Enzymologic , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sex Characteristics , Testosterone/metabolism
15.
J Endocrinol ; 245(1): 165-178, 2020 04.
Article in English | MEDLINE | ID: mdl-32053493

ABSTRACT

Loss of ovarian hormones leads to increased adiposity and insulin resistance (IR), increasing the risk for cardiovascular and metabolic diseases. The purpose of this study was to investigate whether the molecular mechanism behind the adverse systemic and adipose tissue-specific metabolic effects of ovariectomy requires loss of signaling through estrogen receptor alpha (ERα) or estrogen receptor ß (ERß). We examined ovariectomized (OVX) and ovary-intactwild-type (WT), ERα-null (αKO), and ERß-null (ßKO) female mice (age ~49 weeks; n = 7-12/group). All mice were fed a phytoestrogen-free diet (<15 mg/kg) and either remained ovary-intact (INT) or were OVX and followed for 12 weeks. Body composition, energy expenditure, glucose tolerance, and adipose tissue gene and protein expression were analyzed. INT αKO were ~25% fatter with reduced energy expenditure compared to age-matched INT WT controls and ßKO mice (all P < 0.001). Following OVX, αKO mice did not increase adiposity or experience a further increase in IR, unlike WT and ßKO, suggesting that loss of signaling through ERα mediates OVX-induced metabolic dysfunction. In fact, OVX in αKO mice (i.e., signaling through ERß in the absence of ERα) resulted in reduced adiposity, adipocyte size, and IR (P < 0.05 for all). ßKO mice responded adversely to OVX in terms of increased adiposity and development of IR. Together, these findings challenge the paradigm that ERα mediates metabolic protection over ERß in all settings. These findings lead us to suggest that, following ovarian hormone loss, ERß may mediate protective metabolic benefits.


Subject(s)
Adiposity/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Insulin Resistance/genetics , Ovariectomy , Adiponectin/genetics , Adiponectin/metabolism , Adipose Tissue, White/metabolism , Animals , Body Composition/genetics , Energy Metabolism/genetics , Estrogen Receptor alpha/deficiency , Estrogen Receptor beta/deficiency , Female , Gene Expression , Humans , Leptin/genetics , Leptin/metabolism , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics
16.
Ann N Y Acad Sci ; 1461(1): 127-143, 2020 02.
Article in English | MEDLINE | ID: mdl-31868931

ABSTRACT

One of the leading causes for the development of adverse metabolic effects, including type 2 diabetes, dyslipidemia, and cardiovascular diseases, is the accumulation of excess body weight, often measured by body mass index (BMI). Although BMI, calculated using weight and height, is the standard measure used to determine body adiposity in clinical and public health guidelines, an inherent limitation is that BMI does not distinguish where in the body adiposity is deposited. Central obesity, characterized by greater accumulation of adiposity in the abdominal region, has been associated with a higher risk of mortality, independent of BMI. Importantly, one of the determinants of body fat distribution is sex hormones. Both estrogens and androgens appear to directly and indirectly influence body fat distribution. Our review will focus specifically on the role of estrogens and their influence in determining body fat distribution and overall health of adipose tissues, and the role of epigenetic mechanisms in regulating the production and function of estrogens.


Subject(s)
Adipose Tissue/metabolism , Estrogens/metabolism , Adipocytes/metabolism , Adipose Tissue/growth & development , Animals , Gonadal Steroid Hormones/metabolism , Humans , Menopause/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism
17.
Am J Physiol Endocrinol Metab ; 317(3): E548-E558, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31310581

ABSTRACT

Endothelin-1 (ET-1) is a potent vasoconstrictor and proinflammatory peptide that is upregulated in obesity. Herein, we tested the hypothesis that ET-1 signaling promotes visceral adipose tissue (AT) inflammation and disrupts glucose homeostasis. We also tested if reduced ET-1 is a required mechanism by which exercise ameliorates AT inflammation and improves glycemic control in obesity. We found that 1) diet-induced obesity, AT inflammation, and glycemic dysregulation were not accompanied by significantly increased levels of ET-1 in AT or circulation in wild-type mice and that endothelial overexpression of ET-1 and consequently increased ET-1 levels did not cause AT inflammation yet impaired glucose tolerance; 2) reduced AT inflammation and improved glucose tolerance with voluntary wheel running was not associated with decreased levels of ET-1 in AT or circulation in obese mice nor did endothelial overexpression of ET-1 impede such exercise-induced metabolic adaptations; 3) chronic pharmacological blockade of ET-1 receptors did not suppress AT inflammation in obese mice but improved glucose tolerance; and 4) in a cohort of human subjects with a wide range of body mass indexes, ET-1 levels in AT, or circulation were not correlated with markers of inflammation in AT. In aggregate, we conclude that ET-1 signaling is not implicated in the development of visceral AT inflammation but promotes glucose intolerance, thus representing an important therapeutic target for glycemic dysregulation in conditions characterized by hyperendothelinemia. Furthermore, we show that the salutary effects of exercise on AT and systemic metabolic function are not contingent on the suppression of ET-1 signaling.


Subject(s)
Endothelin-1/metabolism , Glucose Intolerance/metabolism , Inflammation/pathology , Intra-Abdominal Fat/pathology , Physical Conditioning, Animal/physiology , Animals , Body Mass Index , Endothelin-1/antagonists & inhibitors , Endothelin-1/genetics , Exercise/physiology , Female , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Obesity/pathology , Running
18.
Neuropharmacology ; 155: 22-30, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31100290

ABSTRACT

Palatability driven feeding and voluntary physical activity are mediated by and influence similar neural mechanisms, notably through the actions of opioids within the nucleus accumbens. Recent studies suggest that access to a voluntary running wheel results in sex dependent behavioral and physiological adaptations related to opioid mediated palatability-driven feeding. To explore this relationship, male and female Wistar rats were given either access to a voluntary running wheel (RUN group) or no access (SED group) for one week prior to being stereotaxically implanted with bilateral cannulae targeting the nucleus accumbens. Following 7 days of recovery, with RUN or SED conditions continuing the duration of the experiment, all rats were assessed daily (2 h/day) for feeding behavior of concurrently accessible high-carbohydrate and high-fat diet for one week. Following this week, all rats were administered the µ-opioid receptor agonist D-Ala2, NMe-Phe4, Glyol5-enkephalin (DAMGO) (0.0025  µg, 0.025  µg, or 0.25 µg/0.5 µl/side) or the opioid antagonist naloxone (20 µg/0.5 µl/side) into the nucleus accumbens and given concurrent access (2 h) to both diets. All groups expressed a significant baseline preference for the high-carbohydrate diet. DAMGO administration, compared to saline treatment, led to significant increased consumption of the high-carbohydrate diet in all treatment groups. While high-fat diet consumption also increased following DAMGO administration, the influence of DAMGO was much more robust for the preferred high-carbohydrate diet in all groups. Compared to males, females consumed significantly more of both diets at baseline and following DAMGO treatment. Both male and female rats in the RUN condition consumed more high-carbohydrate diet compared to rats in the SED condition. While males exhibited similar increased consumption of both diets regardless of RUN or SED condition, females in the RUN condition displayed a greater sensitivity to DAMGO-driven consumption of the preferred high-carbohydrate, compared to SED females.


Subject(s)
Analgesics, Opioid/pharmacology , Dietary Carbohydrates/administration & dosage , Dietary Fats/administration & dosage , Feeding Behavior/drug effects , Motor Activity/drug effects , Nucleus Accumbens/drug effects , Animals , Dose-Response Relationship, Drug , Exercise Test/methods , Exercise Test/psychology , Feeding Behavior/physiology , Feeding Behavior/psychology , Female , Male , Motor Activity/physiology , Nucleus Accumbens/physiology , Rats , Rats, Wistar
19.
Diabetes ; 68(9): 1717-1729, 2019 09.
Article in English | MEDLINE | ID: mdl-30862679

ABSTRACT

The prevailing dogma is that thermogenic brown adipose tissue (BAT) contributes to improvements in glucose homeostasis in obesogenic animal models, though much of the evidence supporting this premise is from thermostressed rodents. Determination of whether modulation of the BAT morphology/function drives changes in glucoregulation at thermoneutrality requires further investigation. We used loss- and gain-of-function approaches including genetic manipulation of the lipolytic enzyme Pnpla2, change in environmental temperature, and lifestyle interventions to comprehensively test the premise that a thermogenic-like BAT phenotype is coupled with enhanced glucose tolerance in female mice. In contrast to this hypothesis, we found that 1) compared to mice living at thermoneutrality, enhanced activation of BAT and its thermogenic phenotype via chronic mild cold stress does not improve glucose tolerance in obese mice, 2) silencing of the Pnpla2 in interscapular BAT causes a brown-to-white phenotypic shift accompanied with inflammation but does not disrupt glucose tolerance in lean mice, and 3) exercise and low-fat diet improve glucose tolerance in obese mice but these effects do not track with a thermogenic BAT phenotype. Collectively, these findings indicate that a thermogenic-like BAT phenotype is not linked to heightened glucose tolerance in female mice.


Subject(s)
Adipose Tissue, Brown/metabolism , Cold-Shock Response/physiology , Obesity/metabolism , Thermogenesis/physiology , Animals , Cold Temperature , Diet, High-Fat , Energy Metabolism/physiology , Female , Glucose Tolerance Test , Lipase/genetics , Lipase/metabolism , Mice , Mice, Knockout , Phenotype
20.
Front Physiol ; 10: 9, 2019.
Article in English | MEDLINE | ID: mdl-30804793

ABSTRACT

Metabolic disease risk escalates following menopause. The mechanism is not fully known, but likely involves reduced signaling through estrogen receptor alpha (ERα), which is highly expressed in brown and white adipose tissue (BAT and WAT). Objective: Test the hypothesis that uncoupling protein (UCP1) activation mitigates metabolic dysfunction caused by loss of signaling through ERα. Methods: At 8 weeks of age, female ERα knock out (KO) and wild-type mice were housed at 28°C and fed a Western-style high-fat, high sucrose diet (HFD) or a normal low-fat chow diet (NC) for 10 weeks. During the final 2 weeks, they received daily injections of CL 316,256 (CL), a selective ß3 adrenergic agonist, or vehicle control (CTRL), creating eight groups: WT-CTRL, WT-CL, KO-CTRL, and KO-CL on HFD or NC; n = 4-10/group. Results: ERαKO demonstrated exacerbated HFD-induced adiposity gain (P < 0.001) and insulin resistance (P = 0.006). CL treatment improved insulin sensitivity (P < 0.05) and normalized ERαKO-induced adiposity increase (P < 0.05). In both genotypes, CL increased resting energy expenditure (P < 0.05) and induced WAT beiging indicated by increased UCP1 protein in both perigonadal (PGAT) and subcutaneous (SQAT) depots. These effects were attenuated under HFD conditions (P < 0.05). In KO, CL reduced HFD energy consumption compared to CTRL (P < 0.05). Remarkably, CL increased WAT ERß protein levels of both WT and KO (P < 0.001), revealing CL-mediated changes in estrogen signaling may have protective metabolic effects. Conclusion: CL completely restored metabolic dysfunction in ERαKO mice. Thus, UCP1 may be a therapeutic target for treating metabolic dysfunction following loss of estrogen receptor signaling.

SELECTION OF CITATIONS
SEARCH DETAIL