Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Am J Physiol Endocrinol Metab ; 324(2): E120-E134, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36516471

ABSTRACT

Estrogen signaling is protective against chronic liver diseases, although men and a subset of women are contraindicated for chronic treatment with 17ß-estradiol (17ß-E2) or combination hormone replacement therapies. We sought to determine if 17α-estradiol (17α-E2), a naturally occurring diastereomer of 17ß-E2, could attenuate liver fibrosis. We evaluated the effects of 17α-E2 treatment on collagen synthesis and degradation rates using tracer-based labeling approaches in male mice subjected to carbon tetrachloride (CCl4)-induced liver fibrosis. We also assessed the effects of 17α-E2 on markers of hepatic stellate cell (HSC) activation, collagen cross-linking, collagen degradation, and liver macrophage content and polarity. We found that 17α-E2 significantly reduced collagen synthesis rates and increased collagen degradation rates, which was mirrored by declines in transforming growth factor ß1 (TGF-ß1) and lysyl oxidase-like 2 (LOXL2) protein content in liver. These improvements were associated with increased matrix metalloproteinase 2 (MMP2) activity and suppressed stearoyl-coenzyme A desaturase 1 (SCD1) protein levels, the latter of which has been linked to the resolution of liver fibrosis. We also found that 17α-E2 increased liver fetuin-A protein, a strong inhibitor of TGF-ß1 signaling, and reduced proinflammatory macrophage activation and cytokines expression in the liver. We conclude that 17α-E2 reduces fibrotic burden by suppressing HSC activation and enhancing collagen degradation mechanisms. Future studies will be needed to determine if 17α-E2 acts directly in hepatocytes, HSCs, and/or immune cells to elicit these benefits.


Subject(s)
Matrix Metalloproteinase 2 , Transforming Growth Factor beta1 , Male , Mice , Female , Animals , Transforming Growth Factor beta1/pharmacology , Matrix Metalloproteinase 2/metabolism , Estradiol/pharmacology , Estradiol/metabolism , Longevity , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Liver/metabolism , Collagen/metabolism
2.
Mech Ageing Dev ; 194: 111425, 2021 03.
Article in English | MEDLINE | ID: mdl-33383072

ABSTRACT

Decline in ovarian reserve with advancing age is associated with reduced fertility and the emergence of metabolic disturbances, osteoporosis, and neurodegeneration. Recent studies have provided insight into connections between ovarian insufficiency and systemic aging, although the basic mechanisms that promote ovarian reserve depletion remain unknown. Here, we sought to determine if chronological age is linked to changes in ovarian cellular senescence, transcriptomic, and epigenetic mechanisms in a mouse model. Histological assessments and transcriptional analyses revealed the accumulation of lipofuscin aggresomes and senescence-related transcripts (Cdkn1a, Cdkn2a, Pai-1 and Hmgb1) significantly increased with advancing age. Transcriptomic profiling and pathway analyses following RNA sequencing, revealed an upregulation of genes related to pro-inflammatory stress and cell-cycle inhibition, whereas genes involved in cell-cycle progression were downregulated; which could be indicative of senescent cell accumulation. The emergence of these senescence-related markers preceded the dramatic decline in primordial follicle reserve observed. Whole Genome Oxidative Bisulfite Sequencing (WGoxBS) found no genome-wide or genomic context-specific DNA methylation and hydroxymethylation changes with advancing age. These findings suggest that cellular senescence may contribute to ovarian aging, and thus, declines in ovarian follicular reserve. Cell-type-specific analyses across the reproductive lifespan are needed to fully elucidate the mechanisms that promote ovarian insufficiency.


Subject(s)
Aging/pathology , Cellular Senescence , Ovarian Follicle/pathology , Ovarian Reserve , Ovary/pathology , Primary Ovarian Insufficiency/pathology , Age Factors , Aging/genetics , Aging/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cytokines/genetics , Cytokines/metabolism , DNA Methylation , Epigenesis, Genetic , Female , Gene Expression Profiling , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Ovarian Follicle/metabolism , Ovarian Follicle/physiopathology , Ovary/metabolism , Ovary/physiopathology , Primary Ovarian Insufficiency/genetics , Primary Ovarian Insufficiency/metabolism , Primary Ovarian Insufficiency/physiopathology , Transcriptome
3.
J Diabetes ; 13(6): 506-511, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33247879

ABSTRACT

BACKGROUND: Studies evaluating endocrine and exocrine functions in fibrocalculous pancreatic diabetes (FCPD) are scarce. METHODS: Insulin, C-peptide, glucagon, incretin hormones (glucagon-like peptide 1 [GLP-1] and gastric inhibitory peptide [GIP]), and dipeptidyl peptidase IV (DPP-IV) were estimated in patients with FCPD (n = 20), type 2 diabetes mellitus (T2DM) (n = 20), and controls (n = 20) in fasting and 60 minutes after 75 g glucose. RESULTS: Fasting and post-glucose C-peptide and insulin in FCPD were lower than that of T2DM and controls. Plasma glucagon decreased after glucose load in controls (3.72, 2.29), but increased in T2DM (4.01, 5.73), and remained unchanged in FCPD (3.44, 3.44). Active GLP-1 (pmol/L) after glucose load increased in FCPD (6.14 to 9.72, P = <.001), in T2DM (2.87 to 4.62, P < .001), and in controls (3.91 to 6.13, P < .001). Median active GLP-1 in FCPD, both in fasting and post-glucose state (6.14, 9.72), was twice that of T2DM (2.87, 4.62) and 1.5 times that of controls (3.91, 6.13) (P < .001 for all). Post-glucose GIP (pmol/L) increased in all: FCPD (15.83 to 94.14), T2DM (21.85 to 88.29), and control (13.00 to 74.65) (P < .001 for all). GIP was not different between groups. DPP-IV concentration (ng/mL) increased in controls (1578.54, 3012.00) and FCPD (1609.95, 1995.42), but not in T2DM (1204.50, 1939.50) (P = .131). DPP-IV between the three groups was not different. Fecal elastase was low in FCPD compared with T2DM controls. CONCLUSIONS: In FCPD, basal C-peptide and glucagon are low, and glucagon does not increase after glucose load. GLP-1, but not GIP, in FCPD increases 1.5 to 2 times as compared with T2DM and controls (fasting and post glucose) without differences in DPP-IV.


Subject(s)
Calcinosis/blood , Diabetes Mellitus, Type 2/blood , Incretins/blood , Pancreatitis, Chronic/blood , Adolescent , Adult , Biomarkers/blood , Blood Glucose/metabolism , C-Peptide/blood , Calcinosis/diagnosis , Calcinosis/drug therapy , Case-Control Studies , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl Peptidase 4/blood , Female , Fibrosis , Gastric Inhibitory Polypeptide/blood , Glucagon/blood , Glucagon-Like Peptide 1/blood , Humans , Hypoglycemic Agents/therapeutic use , Insulin/blood , Male , Middle Aged , Pancreatitis, Chronic/diagnosis , Pancreatitis, Chronic/drug therapy , Time Factors , Young Adult
4.
Elife ; 92020 12 08.
Article in English | MEDLINE | ID: mdl-33289482

ABSTRACT

Metabolic dysfunction underlies several chronic diseases, many of which are exacerbated by obesity. Dietary interventions can reverse metabolic declines and slow aging, although compliance issues remain paramount. 17α-estradiol treatment improves metabolic parameters and slows aging in male mice. The mechanisms by which 17α-estradiol elicits these benefits remain unresolved. Herein, we show that 17α-estradiol elicits similar genomic binding and transcriptional activation through estrogen receptor α (ERα) to that of 17ß-estradiol. In addition, we show that the ablation of ERα completely attenuates the beneficial metabolic effects of 17α-E2 in male mice. Our findings suggest that 17α-E2 may act through the liver and hypothalamus to improve metabolic parameters in male mice. Lastly, we also determined that 17α-E2 improves metabolic parameters in male rats, thereby proving that the beneficial effects of 17α-E2 are not limited to mice. Collectively, these studies suggest ERα may be a drug target for mitigating chronic diseases in male mammals.


Subject(s)
Estradiol/physiology , Estrogen Receptor alpha/physiology , Longevity , Animals , Female , Gene Expression Regulation/physiology , Hypothalamus/metabolism , Hypothalamus/physiology , Insulin Resistance/physiology , Liver/metabolism , Liver/physiology , Longevity/physiology , Male , Mice , Mice, Knockout , Rats
SELECTION OF CITATIONS
SEARCH DETAIL