151
|
Obesity and the role of nonsurgical and surgical weight reduction in idiopathic intracranial hypertension. Int Ophthalmol Clin 2014; 54:27-41. [PMID: 24296369 DOI: 10.1097/iio.0b013e3182aabf2e] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
152
|
Rodrigues MH, Bruno AS, Nahas-Neto J, Santos MES, Nahas EAP. Nonalcoholic fatty liver disease and metabolic syndrome in postmenopausal women. Gynecol Endocrinol 2014; 30:325-9. [PMID: 24460502 DOI: 10.3109/09513590.2013.875992] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is considered the most common cause of chronic liver disease in the Western countries. NAFLD includes a spectrum ranging from a simple steatosis to a nonalcoholic steatohepatitis (NASH) which is defined by the presence of inflammatory infiltrate, cellular necrosis, hepatocyte ballooning, and fibrosis and cirrhosis that can eventually develop into hepatocellular carcinoma. Studies emphasize the role of insulin resistance, oxidative stress, pro-inflammatory cytokines, adipokines in the development and progression of NAFLD. It seems to be independently associated with type II diabetes mellitus, increased triglycerides, decreased HDL-cholesterol, abdominal obesity and insulin resistance. These findings are in accordance with the criteria used in the diagnosis of metabolic syndrome (MetS). Here, we will discuss the current knowledge on the epidemiology, pathophysiology and diagnosis of NAFLD and the association of metabolic syndrome in postmenopausal women.
Collapse
Affiliation(s)
- Marcio H Rodrigues
- Department of Gynecology and Obstetrics, Botucatu Medical School-Sao Paulo State University , Botucatu, Sao Paulo , Brazil
| | | | | | | | | |
Collapse
|
153
|
Rodrigues MH, Bruno AS, Nahas-Neto J, Sandrim VC, Muniz LG, Nahas EAP. Evaluation of clinical and inflammatory markers of nonalcoholic fatty liver disease in postmenopausal women with metabolic syndrome. Metab Syndr Relat Disord 2014; 12:330-8. [PMID: 25051240 DOI: 10.1089/met.2013.0140] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The aim of this study was to assess clinical and inflammatory markers in nonalcoholic fatty liver disease (NAFLD) in postmenopausal women with metabolic syndrome. METHODS This cross-sectional study included 180 Brazilian women (age ≥45 years and amenorrhea ≥12 months). Metabolic syndrome was diagnosed by the presence of at least three of the following indicators: Waist circumference (WC) >88 cm, triglycerides (TGs) ≥150 mg/dL, high-density lipoprotein (HDL) <50 mg/dL; blood pressure ≥130/85 mmHg; and glucose ≥100 mg/dL. NAFLD was diagnosed by abdominal ultrasound. Participants were divided into three groups: Metabolic syndrome alone (n=53); metabolic syndrome+NAFLD (n=67); or absence of metabolic syndrome or NAFLD (control, n=60). Clinical, anthropometric, and biochemical variables were quantified. The inflammatory profile included adiponectin, interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). Data were submitted to statistical analysis using a Tukey test, analysis of variance (ANOVA), chi-squared, Pearson correlation, and logistic regression (odds ratio, OR). RESULTS Women with metabolic syndrome+NAFLD, abdominal obesity, high glucose, and insulin resistance by HOMA-IR were compared to women with metabolic syndrome alone and controls (P<0.05). High values of IL-6 and TNF-α and low values of adiponectin were observed among women with metabolic syndrome alone or metabolic syndrome+NAFLD when compared to controls (P<0.05). In multivariate analysis, the variables considered as risk of NAFLD development were: High systolic blood pressure (SBP) [(OR 1.02, 95% confidence interval (CI) 1.0-1.04]; large WC (OR 1.07, 95% CI 1.01-1.13); insulin resistance (OR 3.81, 95% CI 2.01-7.13); and metabolic syndrome (OR 8.68, 95% CI 3.3-24.1). Adiponectin levels reduced NAFLD risk (OR 0.88, 95% CI 0.80-0.96). CONCLUSION In postmenopausal women, metabolic syndrome, abdominal obesity, and insulin resistance were risk markers for the development of NAFLD, whereas higher adiponectin values indicated a protection marker.
Collapse
Affiliation(s)
- Marcio Hipolito Rodrigues
- 1 Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University-UNESP , Sao Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
154
|
Chaudhari HN, Kim SW, Yun JW. Gender-dimorphic regulation of antioxidant proteins in response to high-fat diet and sex steroid hormones in rats. Free Radic Res 2014; 48:587-98. [DOI: 10.3109/10715762.2014.896003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
155
|
Gettler LT, McDade TW, Feranil AB, Agustin SS, Kuzawa CW. Salivary estradiol and testosterone in Filipino men: Diurnal patterns and relationships with adiposity. Am J Hum Biol 2014; 26:376-83. [PMID: 24573919 DOI: 10.1002/ajhb.22528] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 01/25/2014] [Accepted: 02/01/2014] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVES We used detailed saliva sampling procedures to test for diurnal changes in men's salivary estradiol (E2) and testosterone (T) and assessed whether greater adiposity predicted higher E2 and T. METHODS We drew on a subsample of young adults enrolled in a long-running birth cohort study in Metro Cebu, Philippines. Subjects provided saliva samples at four time points during the day (waking, waking +40 min, early evening, and bedtime), which were assayed for E2 and T. Using these detailed hormonal data, we calculated E2 (n = 29) and T (n = 44) area-under-the-curve values, which provide insights on hormonal production over the study period. RESULTS While T declined immediately after waking and reached a nadir in the early evening, E2 did not show significant diurnal change (P ≥ 0.1) but was positively correlated to T at multiple time points (P ≤ 0.05). Subjects with higher adiposity (BMI, waist circumference, skinfolds) had elevated E2 secretion throughout the day (P ≤ 0.01), but adiposity was not related to salivary T. CONCLUSIONS Consistent with past research, our results indicate that adipose tissue is a significant site of E2 production in males but differ from a limited number of prior studies of young men in that we did not find lower T with increasing adiposity. Given E2's role in male hypothalamic-pituitary-gonadal function and complex interfaces with the immune system, these results have important implications for models of male life history as rates of overweight and obesity rise in populations around the world.
Collapse
Affiliation(s)
- Lee T Gettler
- Department of Anthropology, University of Notre Dame, Indiana
| | | | | | | | | |
Collapse
|
156
|
Abstract
Estrogen sulfotransferase (EST/SULT1E1) is known to catalyze the sulfoconjugation and deactivation of estrogens. The goal of this study is to determine whether and how EST plays a role in human adipogenesis. By using human primary adipose-derived stem cells (ASCs) and whole-fat tissues from the abdominal subcutaneous fat of obese and nonobese subjects, we showed that the expression of EST was low in preadipocytes but increased upon differentiation. Overexpression and knockdown of EST in ASCs promoted and inhibited differentiation, respectively. The proadipogenic activity of EST in humans was opposite to the antiadipogenic effect of the same enzyme in rodents. Mechanistically, EST promoted adipogenesis by deactivating estrogens. The proadipogenic effect of EST can be recapitulated by using an estrogen receptor (ER) antagonist or ERα knockdown. In contrast, activation of ER in ASCs inhibited adipogenesis by decreasing the recruitment of the adipogenic peroxisome proliferator-activated receptor γ (PPARγ) onto its target gene promoters, whereas ER antagonism increased the recruitment of PPARγ to its target gene promoters. Linear regression analysis revealed a positive correlation between the expression of EST and body mass index (BMI), as well as a negative correlation between ERα expression and BMI. We conclude that EST is a proadipogenic factor which may serve as a druggable target to inhibit the turnover and accumulation of adipocytes in obese patients.
Collapse
|
157
|
Kararigas G, Nguyen BT, Jarry H. Estrogen modulates cardiac growth through an estrogen receptor α-dependent mechanism in healthy ovariectomized mice. Mol Cell Endocrinol 2014; 382:909-14. [PMID: 24275180 DOI: 10.1016/j.mce.2013.11.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 11/15/2013] [Accepted: 11/16/2013] [Indexed: 10/26/2022]
Abstract
The modulation of cardiac growth by estrogen in healthy mice is not completely understood. The aim was to investigate the effects of estrogen on cardiac growth in healthy mice lacking either estrogen receptor (ER) α or β. Wild-type (WT), ERα knockout (ERKO) and ERβ knockout (BERKO) 2-month-old mice were ovariectomized and randomly assigned to groups receiving an estradiol (E2)-containing or soy-free (control, CON) diet (n=5-7/group). After three months of E2 administration, WT and BERKO mice had significantly lower body weight, higher relative uterus and heart weight than CON mice, while there was no major E2 effect in ERKO mice. Furthermore, there was a higher concentration of E2-responsive genes Igf1 and Myocd in WT and BERKO but not in ERKO mice. Together, these findings indicate that the estrogenic regulation of cardiac growth in healthy mice is primarily mediated through ERα and not ERβ.
Collapse
Affiliation(s)
- Georgios Kararigas
- Institute of Gender in Medicine and Center for Cardiovascular Research, Charite University Hospital, Berlin, Germany
| | - Ba Tiep Nguyen
- Department of Endocrinology, Goettingen University Hospital, Goettingen, Germany; Faculty of Veterinary Medicine, Hanoi University of Agriculture, Hanoi, Viet Nam
| | - Hubertus Jarry
- Department of Endocrinology, Goettingen University Hospital, Goettingen, Germany.
| |
Collapse
|
158
|
Bruno ADS, Rodrigues MH, Alvares MCB, Nahas-Neto J, Nahas EAP. Non-alcoholic fatty liver disease and its associated risk factors in Brazilian postmenopausal women. Climacteric 2014; 17:465-71. [PMID: 24517420 DOI: 10.3109/13697137.2014.881353] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To evaluate the prevalence and risk factors of non-alcoholic fatty liver disease (NAFLD) in postmenopausal women. METHODS A cross-sectional study was carried involving 188 women (age ≥ 45 years and amenorrhea ≥ 12 months) attending the outpatient unit in south-eastern Brazil. Exclusion criteria were liver disease (hepatitis B and C, cholestatic disease, liver insufficiency), use of drugs that affect liver metabolism; alcoholics; AIDS or cancer history; and morbid obesity. NAFLD was diagnosed by abdominal ultrasound. Clinical, anthropometric (body mass index, waist circumference) and biochemical variables were measured. RESULTS Of the 188 women, 73 (38.8%) had NAFLD. Blood pressure, waist circumference, body mass index, LDL cholesterol, triglycerides and glucose were significantly higher in NAFLD patients when compared with women without NAFLD (control group) (p < 0.05). HOMA-IR values indicated insulin resistance only in the NAFLD group (6.1 ± 4.6 vs. 2.4 ± 1.4 in control group, p < 0.05). Metabolic syndrome was detected in 93.1% of the women affected by NAFLD, and 46.1% of the control group (p < 0.05). In multivariate analysis, adjusted for age and weight, the variables considered at risk for the development of NAFLD, were: high waist circumference (odds ratio (OR) 1.07, 95% confidence interval (CI) 1.01-1.13), insulin resistance (OR 3.81, 95% CI 2.01-7.13), and presence of metabolic syndrome (OR 8.68, 95% CI 3.3-24.1). CONCLUSION NAFLD showed a high prevalence among postmenopausal women. The presence of metabolic syndrome, abdominal obesity and IR were indicators of risk for the development of NAFLD.
Collapse
Affiliation(s)
- A de Souza Bruno
- * Department of Gynecology and Obstetrics, Botucatu Medical School-Sao Paulo State University , Botucatu, Sao Paulo
| | | | | | | | | |
Collapse
|
159
|
Wang N, Luo LL, Xu JJ, Xu MY, Zhang XM, Zhou XL, Liu WJ, Fu YC. Obesity accelerates ovarian follicle development and follicle loss in rats. Metabolism 2014; 63:94-103. [PMID: 24135502 DOI: 10.1016/j.metabol.2013.09.001] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 09/04/2013] [Accepted: 09/04/2013] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Studies have shown that excess body fat negatively affects reproductive functions in females. However, whether obesity affects the ovarian follicle development and ovarian lifespan and the underlying mechanism has not been well elucidated. The aim of the present study was to investigate the association between obesity and ovarian follicle development. METHODS Adult female Sprague-Dawley rats (n = 36) were randomly divided into three groups: the normal control (NC) group, the caloric restriction (CR) group (fed 70% food of the NC group) and the high-fat diet (HF) group. They were maintained on these regimens for 18 weeks. RESULTS The body weight, ovary weight and visceral fat in the HF group were significantly higher than those in the NC group and the CR group at the end of treatment. Histological analysis showed that the HF rats had significantly less number and percentage of primordial follicles, but greater number and percentage of developing and atretic follicles than the NC rats and CR rats. Western blot analysis demonstrated that the level of mTORC1 and p-S6K1 proteins significantly increased in the ovaries of HF rats, whereas that of SIRT1, SIRT6, FOXO3a and NRF-1 decreased compared to the NC rats. In contrast, the expression of mTORC1 and p-S6K1 dramatically declined, while that of SIRT1, SIRT6, FOXO3a and NRF1 increased in the ovaries of CR rats. CONCLUSIONS Our study suggests that the HF diet induced obesity may accelerate the ovarian follicle development and rate of follicle loss through activating mTOR and suppressing SIRT1 signaling, thus leading to POF, and that CR may inhibit the activation of primordial follicles, follicular development and loss, thus extending the ovarian lifespan through suppressing mTOR and activating SIRT1 signaling.
Collapse
Affiliation(s)
- Na Wang
- Laboratory of Cell Senescence, Shantou University Medical College, Xinling Road 22, Shantou 515041, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
160
|
Zhang H, Sairam MR. Sex hormone imbalances and adipose tissue dysfunction impacting on metabolic syndrome; a paradigm for the discovery of novel adipokines. Horm Mol Biol Clin Investig 2014; 17:89-97. [DOI: 10.1515/hmbci-2014-0002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 02/24/2014] [Indexed: 01/13/2023]
Abstract
AbstractSex hormone imbalance is causally related with visceral adipose tissue (AT) dysfunction and visceral obesity – an etiological component of metabolic syndrome (MetS), associated with high risk of both cardiovascular disease (CVD) and type 2 diabetes. In general, premenopausal women appear to be protected from CVD and the dramatic decline in sex steroid hormone occurring during menopausal transitions or other sex-related disorders influence the regional distribution, function, and metabolism of AT and increase the risk of CVD. Visceral AT dysfunction, manifesting as abnormality of fatty acid metabolism, increased oxidative stress, endothelial dysfunction, and excessive production of adipokines have been proposed in the pathogenesis of MetS. However, direct evidence of molecular mechanisms of depot-specific AT alterations, and dysfunction causally related to MetS is limited in studies on postmenopausal women due to difficulty in collecting discrete AT specimens at different ages and repeated sampling from different fat depots. This can be overcome using animal models that can mimic the cluster of pathology leading to MetS and help establish the molecular basis of links between loss of gonadal function on various AT depots and their contribution to MetS. Our group used sex hormone imbalance FSH receptor knock out (FORKO) female mice to recapitulate different aspects of the MetS and addressed the mechanism of visceral obesity related to MetS and discover two novel sex steroid hormone-regulated deep mesenteric estrogen-dependent adipose (MEDAs) genes. Taken together, such recent studies raise hopes for pharmacologic intervention strategies targeting sex steroid hormone signaling in AT to provide protection against AT dysfunction.
Collapse
|
161
|
Varlamov O, Bethea CL, Roberts CT. Sex-specific differences in lipid and glucose metabolism. Front Endocrinol (Lausanne) 2014; 5:241. [PMID: 25646091 PMCID: PMC4298229 DOI: 10.3389/fendo.2014.00241] [Citation(s) in RCA: 209] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 12/19/2014] [Indexed: 12/12/2022] Open
Abstract
Energy metabolism in humans is tuned to distinct sex-specific functions that potentially reflect the unique requirements in females for gestation and lactation, whereas male metabolism may represent a default state. These differences are the consequence of the action of sex chromosomes and sex-specific hormones, including estrogens and progesterone in females and androgens in males. In humans, sex-specific specialization is associated with distinct body-fat distribution and energy substrate-utilization patterns; i.e., females store more lipids and have higher whole-body insulin sensitivity than males, while males tend to oxidize more lipids than females. These patterns are influenced by the menstrual phase in females, and by nutritional status and exercise intensity in both sexes. This minireview focuses on sex-specific mechanisms in lipid and glucose metabolism and their regulation by sex hormones, with a primary emphasis on studies in humans and the most relevant pre-clinical model of human physiology, non-human primates.
Collapse
Affiliation(s)
- Oleg Varlamov
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center, Beaverton, OR, USA
- Division of Developmental and Reproductive Sciences, Oregon National Primate Research Center, Beaverton, OR, USA
- *Correspondence: Oleg Varlamov, Divisions of Diabetes, Obesity, and Metabolism and Developmental and Reproductive Sciences, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006, USA e-mail:
| | - Cynthia L. Bethea
- Division of Developmental and Reproductive Sciences, Oregon National Primate Research Center, Beaverton, OR, USA
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, USA
| | - Charles T. Roberts
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center, Beaverton, OR, USA
- Division of Developmental and Reproductive Sciences, Oregon National Primate Research Center, Beaverton, OR, USA
- Department of Medicine, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
162
|
Marinou K, Hodson L, Vasan SK, Fielding BA, Banerjee R, Brismar K, Koutsilieris M, Clark A, Neville MJ, Karpe F. Structural and functional properties of deep abdominal subcutaneous adipose tissue explain its association with insulin resistance and cardiovascular risk in men. Diabetes Care 2014; 37:821-9. [PMID: 24186879 DOI: 10.2337/dc13-1353] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Fat distribution is an important variable explaining metabolic heterogeneity of obesity. Abdominal subcutaneous adipose tissue (SAT) is divided by the Scarpa's fascia into a deep subcutaneous adipose tissue (dSAT) and a superficial subcutaneous adipose tissue (sSAT) layer. This study sought to characterize functional differences between the two SAT layers to explore their relative contribution to metabolic traits and cardiovascular risk (CVR) profile. RESEARCH DESIGN AND METHODS We recruited 371 Caucasians consecutively from a local random, population-based screening project in Oxford and 25 Asian Indians from the local community. The depth of the SAT layers was determined by ultrasound (US), and adipose tissue (AT) biopsies were performed under US guidance in a subgroup of 43 Caucasians. Visceral adipose tissue (VAT) mass was quantified by dual-energy X-ray absorptiometry scan. RESULTS Male adiposity in both ethnic groups was characterized by a disproportionate expansion of dSAT, which was strongly correlated with VAT mass. dSAT depth was a strong predictor of global insulin resistance (IR; homeostatic model assessment of IR), liver-specific IR (insulin-like growth factor binding protein-1), and Framingham risk score independently of other measures of adiposity in men. Moreover, dSAT had higher expression of proinflammatory, lipogenic, and lipolytic genes and contained higher proportions of saturated fatty acids. There was increased proportion of small adipocytes in dSAT. CONCLUSIONS SAT is heterogeneous; dSAT expands disproportionally more than sSAT with increasing obesity in Caucasian males (confirmed also in Asian Indians). Its expansion is related to increased CVR independent of other adiposity measures, and it has biological properties suggestive of higher metabolic activity contributing to global IR.
Collapse
|
163
|
Arnetz L, Ekberg NR, Alvarsson M. Sex differences in type 2 diabetes: focus on disease course and outcomes. Diabetes Metab Syndr Obes 2014; 7:409-20. [PMID: 25258546 PMCID: PMC4172102 DOI: 10.2147/dmso.s51301] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Women with type 2 diabetes (T2D) are less likely to reach the goals for hemoglobin A1c compared with men, and have higher all-cause mortality. The risk of cardiovascular disease is elevated among both men and women with T2D, however, the risk has declined among men over recent years while it remains stationary in women. Reasons for these sex differences remain unclear, and guidelines for diabetes treatment do not differentiate between sexes. Possible causes for varying outcome include differences in physiology, treatment response, and psychological factors. This review briefly outlines sex differences in hormonal pathophysiology, and thereafter summarizes the literature to date on sex differences in disease course and outcome. METHODS Systematic searches were performed on PubMed using "sex", "gender", and various glucose-lowering therapies as keywords. Earlier reviews are summarized and results from individual studies are reported. Reference lists from studies were used to augment the search. RESULTS There is an increased risk of missing the diagnosis of T2D when screening women with only fasting plasma glucose instead of with an oral glucose tolerance test. The impact of various risk factors for complications may differ by sex. Efficacy and side effects of some glucose-lowering drugs differ between men and women. Men with T2D appear to suffer more microvascular complications, while women have higher morbidity and mortality in cardiovascular disease and also fare worse psychologically. CONCLUSION Few studies to date have focused on sex differences in T2D. Several questions demand further study, such as whether risk factors and treatment guidelines should be sex-specific. There is a need for clinical trials designed specifically to evaluate sex differences in efficacy and outcome of the available treatments.
Collapse
Affiliation(s)
- Lisa Arnetz
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Endocrinology, Metabolism and Diabetes, Stockholm, Sweden
| | - Neda Rajamand Ekberg
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Endocrinology, Metabolism and Diabetes, Stockholm, Sweden
| | - Michael Alvarsson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Endocrinology, Metabolism and Diabetes, Stockholm, Sweden
- Correspondence: Michael Alvarsson, Department of Endocrinology, Diabetes and Metabolism, D2:04, Karolinska University Hospital Solna, 17176 Stockholm, Sweden, Tel +46 8 5177 2862, Fax +46 8 5177 3096, Email
| |
Collapse
|
164
|
Rettberg JR, Yao J, Brinton RD. Estrogen: a master regulator of bioenergetic systems in the brain and body. Front Neuroendocrinol 2014; 35:8-30. [PMID: 23994581 PMCID: PMC4024050 DOI: 10.1016/j.yfrne.2013.08.001] [Citation(s) in RCA: 328] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 08/09/2013] [Accepted: 08/10/2013] [Indexed: 01/12/2023]
Abstract
Estrogen is a fundamental regulator of the metabolic system of the female brain and body. Within the brain, estrogen regulates glucose transport, aerobic glycolysis, and mitochondrial function to generate ATP. In the body, estrogen protects against adiposity, insulin resistance, and type II diabetes, and regulates energy intake and expenditure. During menopause, decline in circulating estrogen is coincident with decline in brain bioenergetics and shift towards a metabolically compromised phenotype. Compensatory bioenergetic adaptations, or lack thereof, to estrogen loss could determine risk of late-onset Alzheimer's disease. Estrogen coordinates brain and body metabolism, such that peripheral metabolic state can indicate bioenergetic status of the brain. By generating biomarker profiles that encompass peripheral metabolic changes occurring with menopause, individual risk profiles for decreased brain bioenergetics and cognitive decline can be created. Biomarker profiles could identify women at risk while also serving as indicators of efficacy of hormone therapy or other preventative interventions.
Collapse
Affiliation(s)
- Jamaica R Rettberg
- Neuroscience Department, University of Southern California, Los Angeles, CA 90033, United States
| | - Jia Yao
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033, United States
| | - Roberta Diaz Brinton
- Neuroscience Department, University of Southern California, Los Angeles, CA 90033, United States; Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033, United States; Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States.
| |
Collapse
|
165
|
Kargi AY, Iacobellis G. Adipose tissue and adrenal glands: novel pathophysiological mechanisms and clinical applications. Int J Endocrinol 2014; 2014:614074. [PMID: 25018768 PMCID: PMC4075085 DOI: 10.1155/2014/614074] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 05/24/2014] [Indexed: 12/22/2022] Open
Abstract
Hormones produced by the adrenal glands and adipose tissues have important roles in normal physiology and are altered in many disease states. Obesity is associated with changes in adrenal function, including increase in adrenal medullary catecholamine output, alterations of the hypothalamic-pituitary-adrenal (HPA) axis, elevations in circulating aldosterone together with changes in adipose tissue glucocorticoid metabolism, and enhanced adipocyte mineralocorticoid receptor activity. It is unknown whether these changes in adrenal endocrine function are in part responsible for the pathogenesis of obesity and related comorbidities or represent an adaptive response. In turn, adipose tissue hormones or "adipokines" have direct effects on the adrenal glands and interact with adrenal hormones at several levels. Here we review the emerging evidence supporting the existence of "cross talk" between the adrenal gland and adipose tissue, focusing on the relevance and roles of their respective hormones in health and disease states including obesity, metabolic syndrome, and primary disorders of the adrenals.
Collapse
Affiliation(s)
- Atil Y. Kargi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- *Atil Y. Kargi:
| | - Gianluca Iacobellis
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
166
|
Varlamov O, Chu MP, McGee WK, Cameron JL, O'Rourke RW, Meyer KA, Bishop CV, Stouffer RL, Roberts CT. Ovarian cycle-specific regulation of adipose tissue lipid storage by testosterone in female nonhuman primates. Endocrinology 2013; 154:4126-35. [PMID: 24008344 PMCID: PMC3800767 DOI: 10.1210/en.2013-1428] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Previous studies in rodents and humans suggest that hyperandrogenemia causes white adipose tissue (WAT) dysfunction in females, although the underlying mechanisms are poorly understood. In light of the differences in the length of the ovarian cycle between humans and rodents, we used a nonhuman primate model to elucidate the effects of chronic hyperandrogenemia on WAT function in vivo. Female rhesus macaques implanted with testosterone capsules developed insulin resistance and altered leptin secretion on a high-fat, Western-style diet. In control visceral WAT, lipolysis and hormone-sensitive lipase expression were upregulated during the luteal phase compared with the early follicular (menses) phase of the ovarian cycle. Hyperandrogenemia attenuated elevated lipolysis and hormone-sensitive lipase activity in visceral WAT during the luteal phase but not during menses. Under control conditions, insulin-stimulated Akt and Erk activation and fatty acid uptake in WAT were not significantly affected by the ovarian cycle. In contrast, testosterone treatment preferentially increased fatty acid uptake and insulin signaling at menses. The fatty acid synthase and glucose transporter-4 genes were upregulated by testosterone during the luteal phase. In summary, this study reveals ovarian stage-specific fluctuations in adipocyte lipolysis and suggests that male sex hormones increase and female sex hormones decrease lipid storage in female WAT.
Collapse
Affiliation(s)
- Oleg Varlamov
- Jr, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, Oregon 97006.
| | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Zhu P, Yuen JML, Sham KWY, Cheng CHK. GPER mediates the inhibitory actions of estrogen on adipogenesis in 3T3-L1 cells through perturbation of mitotic clonal expansion. Gen Comp Endocrinol 2013; 193:19-26. [PMID: 23871778 DOI: 10.1016/j.ygcen.2013.07.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 06/21/2013] [Accepted: 07/04/2013] [Indexed: 11/27/2022]
Abstract
G-protein-coupled estrogen receptor 1 (GPER) mediates non-genomic signaling of estrogenic events. Here we showed for the first time that Gper/GPER is expressed in Swiss 3T3 mouse embryo preadipocytes 3T3-L1, and that Gper/GPER is up-regulated during differentiation of the cells induced by monocyte differentiation-inducing (MDI) cocktail. Activation of GPER by the natural ligand 17β-estradiol (E2), and the specific agonist G1, was shown to inhibit lipid accumulation in 3T3-L1 cells, while such inhibition was reversed upon knockdown of GPER using specific siRNA. GPER was also found to mediate perturbation of mitotic clonal expansion (MCE) in these cells by inhibiting cell cycle arrest during MDI cocktail-induced differentiation. Persistent activation of cell cycle regulating factors cyclin-dependant kinase (CDK) 4, CDK6 and cyclin D1, and phosphorylation of retinoblastoma (Rb) protein at serine 795 was observed in the G1-treated cells. Taken together, our results indicate that E2-GPER signaling leads to an inhibition of adipogenesis in 3T3-L1 cells via perturbation of MCE.
Collapse
Affiliation(s)
- Pei Zhu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | | | | | | |
Collapse
|
168
|
Effects of estrogen on food intake, serum leptin levels and leptin mRNA expression in adipose tissue of female rats. Lab Anim Res 2013; 29:168-73. [PMID: 24106512 PMCID: PMC3791351 DOI: 10.5625/lar.2013.29.3.168] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 07/29/2013] [Accepted: 08/20/2013] [Indexed: 11/21/2022] Open
Abstract
The integration of metabolism and reproduction involves complex interactions of hypothalamic neuropeptides with metabolic hormones, fuels, and sex steroids. Of these, estrogen influences food intake, body weight, and the accumulation and distribution of adipose tissue. In this study, the effects of estrogen on food intake, serum leptin levels, and leptin mRNA expression were evaluated in ovariectomized rats. Seven-week-old female Wistar-Imamichi rats were ovariectomized and divided into three treatment groups: group 1 (the control group) received sesame oil, group 2 was given 17β-estradiol benzoate, and group 3 received 17β-estradiol benzoate plus progesterone. The body weight and food consumption of each rat were determined daily. Serum leptin levels and leptin mRNA expression were measured by ELISA and quantitative RT-PCR, respectively. Food consumption in the control group was significantly higher (P<0.05) than that in groups 2 and 3, although body weight did not significantly differ among the three groups. The serum leptin concentration and leptin mRNA expression were significantly higher (P<0.05) in groups 2 and 3 than in group 1, but no significant difference existed between groups 2 and 3. In conclusion, estrogen influenced food intake via the modulation of leptin signaling pathway in ovariectomized rats.
Collapse
|
169
|
Murphy ZC, Pezuk P, Menaker M, Sellix MT. Effects of ovarian hormones on internal circadian organization in rats. Biol Reprod 2013; 89:35. [PMID: 23843233 DOI: 10.1095/biolreprod.113.109322] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The circadian clock in the suprachiasmatic nucleus (SCN) of the hypothalamus is the central pacemaker driving rhythms in endocrine physiology. Gonadal steroid hormones affect behavioral rhythms and clock gene expression. However, the impact of fluctuating ovarian steroid levels during the estrous cycle on internal circadian organization remains to be determined. Further, it is not known if steroid hormone depletion, as in menopause, affects the timing system. To determine the influence of estrous cycle stage and steroid depletion on circadian organization, we measured clock gene expression in the SCN and peripheral tissues from cycling and ovariectomized (OVX) period1-luciferase (per1-luc) transgenic rats. The estrous cycle had modest effects on mean phase and phase distribution of per1-luc expression in the SCN. Surprisingly, peak per1-luc expression in the SCN was widely distributed mainly at night, regardless of cycle stage, an effect eliminated by OVX. Treatment of SCN tissue explants with ovarian steroids did not significantly affect per1-luc expression, suggesting that brain regions outside the SCN mediate the phasic effects of steroids. Our data demonstrate that estrous cycle stage has tissue-dependent effects on the phase of per1-luc expression, phase synchrony among oscillators, and the phase relationship between some peripheral clocks and the light-dark cycle. They also reveal that steroid hormone depletion following OVX alters the timing system, suggesting that the decline in hormone levels, common during the transition to menopause, may be associated with irregular internal circadian organization. This effect on the timing system could contribute to the behavioral and physiological changes associated with this transition.
Collapse
Affiliation(s)
- Zachary C Murphy
- Department of Medicine, Division of Endocrinology and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | | | | | | |
Collapse
|
170
|
Gavin KM, Cooper EE, Hickner RC. Estrogen receptor protein content is different in abdominal than gluteal subcutaneous adipose tissue of overweight-to-obese premenopausal women. Metabolism 2013; 62:1180-8. [PMID: 23557590 DOI: 10.1016/j.metabol.2013.02.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 02/01/2013] [Accepted: 02/25/2013] [Indexed: 12/01/2022]
Abstract
OBJECTIVE Premenopausal women demonstrate a distinctive gynoid body fat distribution and circulating estrogen status is associated with the maintenance of this adiposity patterning. Estrogen's role in modulation of regional adiposity may occur through estrogen receptors (ERs), which are present in human adipose tissue. The purpose of this study was to determine regional differences in the protein content of ERα, ERβ, and the G protein-coupled estrogen receptor (GPER) between the abdominal (AB) and gluteal (GL) subcutaneous adipose tissue of overweight-to-obese premenopausal women. MATERIALS/METHODS Biopsies of the subcutaneous AB and GL adipose tissue were performed in 15 premenopausal women (7 Caucasian/8 African American, 25.1 ± 1.8 years, BMI 29.5 ± 0.5kg/m(2)). Adipose tissue protein content was measured by western blot analysis and correlation analyses were conducted to assess the relationship between ER protein content and anthropometric indices/body composition measurements. RESULTS We found that ERα protein was higher in AB than GL (AB 1.0 ± 0.2 vs GL 0.67 ± 0.1 arbitrary units [AU], P=0.02), ERβ protein was higher in GL than AB (AB 0.78 ± 0.12 vs GL 1.3 ± 0.2 AU, P=0.002), ERα/ERβ ratio was higher in AB than GL (AB 1.9 ± 0.4 vs GL 0.58 ± 0.08 AU, P=0.007), and GPER protein content was similar in AB and GL (P=0.80) subcutaneous adipose tissue. Waist-to-hip ratio was inversely related to gluteal ERβ (r(2)=0.315, P=0.03) and positively related to gluteal ERα/ERβ ratio (r(2)=0.406, P=0.01). CONCLUSIONS These results indicate that depot specific ER content may be an important underlying determinant of regional effects of estrogen in upper and lower body adipose tissue of overweight-to-obese premenopausal women.
Collapse
Affiliation(s)
- Kathleen M Gavin
- Human Performance Laboratory, East Carolina University, Greenville, NC 27858, USA.
| | | | | |
Collapse
|
171
|
Effect of APOE polymorphism on obesity and lipid profile in women of differing reproductive status. Open Life Sci 2013. [DOI: 10.2478/s11535-013-0192-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractThe aim of this study was to investigate whether the effect of apolipoprotein E polymorphism (APOE) on somatic and lipid risk parameters varies in women of differing reproductive status. We analyzed 447 Slovak women aged between 39 and 90 years. APOE genotypes were determined by PCR-RFLP. Regression analysis confirmed the effect of the APOE genotype on the levels of LDL-cholesterol, apolipoprotein B (apoB), nonHDL-cholesterol and on the three atherogenic indices: apoB-to-apoA1, TC-to-HDLcholesterol, LDL-C-to-HDL-cholesterol. Here, lower mean levels were registered in the E2 carriers than in the E3 and E4 subgroups. However, the impact of menopausal status on lipid parameters was not confirmed. Bonferroni correction showed that systolic blood pressure was significantly lower in the E4 carriers compared to the E3 group (P=0.017). Univariate analysis of covariance revealed a significant interaction between the menopausal group and the APOE group, and their common effect on waist-to-hipratio (WHR). Bonferroni correction in early postmenopausal women showed that the mean WHR values were significantly different between E2 and E4 groups (P=0.008). This study demonstrates that the E*2 allele has a protective effect against higher blood lipid levels. Moreover, the results suggest that E*2 could have a partial negative effect on WHR in early postmenopausal Slovak women.
Collapse
|
172
|
Gaikwad NW. Metabolomic profiling unravels DNA adducts in human breast that are formed from peroxidase mediated activation of estrogens to quinone methides. PLoS One 2013; 8:e65826. [PMID: 23762435 PMCID: PMC3675060 DOI: 10.1371/journal.pone.0065826] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 04/30/2013] [Indexed: 12/18/2022] Open
Abstract
Currently there are three major hypotheses that have been proposed for estrogen induced carcinogenicity, however exact etiology remains unknown. Based on the chemical logic, studies were undertaken to investigate if estrogens could generate quinone methides in an oxidative environment which then could cause DNA damage in humans. In presence of MnO2 estrogens were oxidized to quinone methides. Surprisingly quinone methides were found to be stable with t1/2 of 20.8 and 4.5 min respectively. Incubation of estrogens with lactoperoxidase (LPO) and H2O2 resulted in formation of respective quinone methides (E1(E2)-QM). Subsequent addition of adenine to the assay mixture lead to trapping of E1(E2)-QM, resulting in formation of adenine adducts of estrogens, E1(E2)-9-N-Ade. Targeted ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) based metabolomic analysis of the breast tissue extracts showed the presence of adenine adducts of estrogens, E1(E2)-9-N-Ade, along with other estrogen related metabolites. Identity of E1(E2)-N-Ade in LPO assay extracts and breast tissue extracts were confirmed by comparing them to pure synthesized E1(E2)-9-N-Ade standards. From these results, it is evident that peroxidase enzymes or peroxidase-like activity in human breast tissue could oxidize estrogens to electrophilic and stable quinone methides in a single step that covalently bind to DNA to form adducts. The error prone repair of the damaged DNA can result in mutation of critical genes and subsequently cancer. This article reports evidence for hitherto unknown estrogen metabolic pathway in human breast, catalyzed by peroxidase, which could initiate cancer.
Collapse
Affiliation(s)
- Nilesh W. Gaikwad
- Department of Nutrition and Department of Environmental Toxicology, University of California Davis, Davis, California, United States of America
| |
Collapse
|
173
|
Borrell LN, Nguyen EA, Roth LA, Oh SS, Tcheurekdjian H, Sen S, Davis A, Farber HJ, Avila PC, Brigino-Buenaventura E, Lenoir MA, Lurmann F, Meade K, Serebrisky D, Rodriguez-Cintron W, Kumar R, Rodriguez-Santana JR, Thyne SM, Burchard EG. Childhood obesity and asthma control in the GALA II and SAGE II studies. Am J Respir Crit Care Med 2013; 187:697-702. [PMID: 23392439 DOI: 10.1164/rccm.201211-2116oc] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
RATIONALE Obesity is associated with increased asthma morbidity, lower drug responsiveness to inhaled corticosteroids, and worse asthma control. However, most prior investigations on obesity and asthma control have not focused on pediatric populations, considered environmental exposures, or included minority children. OBJECTIVES To examine the association between body mass index categories and asthma control among boys and girls; and whether these associations are modified by age and race/ethnicity. METHODS Children and adolescents ages 8-19 years (n = 2,174) with asthma were recruited from the Genes-environments and Admixture in Latino Americans (GALA II) Study and the Study of African Americans, Asthma, Genes, and Environments (SAGE II). Ordinal logistic regression was used to estimate odds ratios (OR) and their confidence intervals (95% CI) for worse asthma control. MEASUREMENTS AND MAIN RESULTS In adjusted analyses, boys who were obese had a 33% greater chance of having worse asthma control than their normal-weight counterparts (OR, 1.33; 95% CI, 1.04-1.71). However, for girls this association varied with race and ethnicity (P interaction = 0.008). When compared with their normal-weight counterparts, obese African American girls (OR, 0.65; 95% CI, 0.41-1.05) were more likely to have better controlled asthma, whereas Mexican American girls had a 1.91 (95% CI, 1.12-3.28) greater odds of worse asthma control. CONCLUSIONS Worse asthma control is uniformly associated with increased body mass index in boys. Among girls, the direction of this association varied with race/ethnicity.
Collapse
Affiliation(s)
- Luisa N Borrell
- Department of Health Sciences, Graduate Program in Public Health, Lehman College, City University of New York, Bronx, NY, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
174
|
White UA, Tchoukalova YD. Sex dimorphism and depot differences in adipose tissue function. Biochim Biophys Acta Mol Basis Dis 2013; 1842:377-92. [PMID: 23684841 DOI: 10.1016/j.bbadis.2013.05.006] [Citation(s) in RCA: 192] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 04/18/2013] [Accepted: 05/03/2013] [Indexed: 02/06/2023]
Abstract
Obesity, characterized by excessive adiposity, is a risk factor for many metabolic pathologies, such as type 2 diabetes mellitus (T2DM). Numerous studies have shown that adipose tissue distribution may be a greater predictor of metabolic health. Upper-body fat (visceral and subcutaneous abdominal) is commonly associated with the unfavorable complications of obesity, while lower-body fat (gluteal-femoral) may be protective. Current research investigations are focused on analyzing the metabolic properties of adipose tissue, in order to better understand the mechanisms that regulate fat distribution in both men and women. This review will highlight the adipose tissue depot- and sex-dependent differences in white adipose tissue function, including adipogenesis, adipose tissue developmental patterning, the storage and release of fatty acids, and secretory function. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.
Collapse
Affiliation(s)
- Ursula A White
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Yourka D Tchoukalova
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA.
| |
Collapse
|
175
|
Gaikwad NW. Ultra performance liquid chromatography-tandem mass spectrometry method for profiling of steroid metabolome in human tissue. Anal Chem 2013; 85:4951-60. [PMID: 23597399 DOI: 10.1021/ac400016e] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In humans, steroids play a broad and vital role in regulation of gene expression, secondary sexual characteristics, maturation, reproduction, cardiovascular health, neurological functions, etc., but imbalance in steroid metabolism is also linked to development and progression of many diseases, such as cancer, neurodegenerative diseases, and cardiovascular diseases. Hence, measurement of steroids in biological samples is essential to monitor human health. Currently, there is radioimmunoassay, gas chromatography-mass spectrometry (GC/MS), and liquid chromatography-mass spectrometry (LC-MS) methods developed for steroid measurements in biological samples. However, these methods require elaborate sample preparation procedures and have concerns(s) related to reproducibility, dynamic range, time, costs, and most importantly the total coverage of steroids. Also currently, there is no method available for comprehensive steroid profiling in a single LC-MS run that includes androgens, corticosteroids, progestogens, estrogens, estrogen metabolites, estrogen conjugates, and estrogen-DNA adducts as well as exogenous steroid derivatives. Here, I present a global steroid metabolic profiling method based on liquid-liquid extraction (LLE) followed by ultra performance liquid chromatography (UPLC)-tandem mass spectrometry (MS/MS) for simultaneous measurement of over 100 indigenous as well as exogenous steroids in about 12 min, without derivatization. The method was successfully applied to determine steroid hormone levels in the breast tissue of healthy women. Overall presence of all major classes of steroids as well as estrogen derivatives was detected in breast tissue.
Collapse
Affiliation(s)
- Nilesh W Gaikwad
- Departments of Nutrition and Environmental Toxicology, University of California, Davis, California 95616, United States.
| |
Collapse
|
176
|
Abstract
The alarming prevalence of obesity has led to a better understanding of the molecular mechanisms controlling energy homeostasis. Regulation of energy intake and expenditure is more complex than previously thought, being influenced by signals from many peripheral tissues. In this sense, a wide variety of peripheral signals derived from different organs contributes to the regulation of body weight and energy expenditure. Besides the well-known role of insulin and adipokines, such as leptin and adiponectin, in the regulation of energy homeostasis, signals from other tissues not previously thought to play a role in body weight regulation have emerged in recent years. The role of fibroblast growth factor 21 (FGF21), insulin-like growth factor 1 (IGF-I), and sex hormone-binding globulin (SHBG) produced by the liver in the regulation of body weight and insulin sensitivity has been recently described. Moreover, molecules expressed by skeletal muscle such as myostatin have also been involved in adipose tissue regulation. Better known is the involvement of ghrelin, cholecystokinin, glucagon-like peptide 1 (GLP-1) and PYY(3-36), produced by the gut, in energy homeostasis. Even the kidney, through the production of renin, appears to regulate body weight, with mice lacking this hormone exhibiting resistance to diet-induced obesity. In addition, the skeleton has recently emerged as an endocrine organ, with effects on body weight control and glucose homeostasis through the actions of bone-derived factors such as osteocalcin and osteopontin. The comprehension of these signals will help in a better understanding of the aetiopathology of obesity, contributing to the potential development of new therapeutic targets aimed at tackling excess body fat accumulation.
Collapse
|
177
|
Ukkola O, Huttunen T, Puurunen VP, Piira OP, Niva J, Lepojärvi S, Tulppo M, Huikuri H. Total testosterone levels, metabolic parameters, cardiac remodeling and exercise capacity in coronary artery disease patients with different stages of glucose tolerance. Ann Med 2013; 45:206-12. [PMID: 23548167 DOI: 10.3109/07853890.2012.711951] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE AND METHODS The correlation between total testosterone levels, exercise capacity, and metabolic and echocardiographic parameters was studied in 1097 male subjects with coronary artery disease (CAD) and different stages of glucose tolerance. RESULTS Testosterone level was the lowest among diabetics as compared to prediabetics or controls (P < 0.001). Total and abdominal adiposity were the highest in the subjects with the lowest testosterone. Independent of adiposity, fasting glucose, insulin, and leptin were higher (P < 0.03 to < 0.001) among diabetic and control groups in the lowest, and HbA1c values (P < 0.001) higher among diabetics in the lowest, than in the highest testosterone tertile. Controls and prediabetic subjects with the lowest testosterone levels had the lowest HDL-cholesterol levels, and controls also the highest triglycerides. An association between low testosterone level and low maximal exercise capacity was observed in diabetics (P < 0.001) and controls (P < 0.03). Independent of adiposity and metabolic parameters, low testosterone levels were associated with the highest septal wall thickness (P < 0.03) among diabetics. CONCLUSION A negative correlation between low testosterone and dysmetabolic features was observed. Independent of metabolic status, low plasma testosterone seems to be an indicator of impaired maximal exercise capacity and cardiac hypertrophy among CAD patients with type II diabetes.
Collapse
Affiliation(s)
- Olavi Ukkola
- Institute of Clinical Medicine, Department of Internal Medicine, University of Oulu and Clinical Research Center of Oulu University Hospital, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
178
|
Choi KO, Lee I, Paik SYR, Kim DE, Lim JD, Kang WS, Ko S. Ultrafine Angelica gigas powder normalizes ovarian hormone levels and has antiosteoporosis properties in ovariectomized rats: particle size effect. J Med Food 2013; 15:863-72. [PMID: 23039111 DOI: 10.1089/jmf.2011.2047] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
The root of Angelica gigas (Korean angelica) is traditionally used to treat women's ailments that are caused by an impairment of menstrual blood flow and cycle irregularities. This study evaluated the effect particle size of Korean angelica powder on its efficacy for treating estrogen-related symptoms of menopause. Initially, Korean angelica roots were pulverized into ultrafine powder, and orally administered to the rats at a concentration of 500 mg/kg body weight for 8 weeks. The effects of Korean angelica powder particle size on extraction yield, contents of bioactive compounds (decursin and decursinol angelate), levels of serum ovarian hormones (estradiol and progesterone), reproductive hormones (luteinizing hormone and follicle-stimulating hormone), and experimental osteoporosis parameters (mineral density, strength, and histological features) were determined. A significant increase (fivefold) in the contents of decursin and decursinol angelate in the extract of the ultrafine Korean angelica powder was observed compared to coarse Korean angelica powder. Rats were divided into sham-operated or ovariectomized (OVX) groups that were fed coarse (CRS) or ultrafine (UF) ground Korean angelica root. The serum levels of estradiol in the OVX_UF group were 19.2% and 54.1% higher than that of OVX_CRS group. Serum bone-alkaline phosphatase/total-alkaline phosphatase index in the OVX_UF group was half that of the OVX_CRS group. In addition, less trabecular bone loss and thick cortical areas were observed in rats administered ultrafine powder. Therefore, ultrafine grinding may enhance the bioactivity of herbal medicines and be especially useful when their extracted forms lose bioactivity during processing, storage, and oral intake.
Collapse
Affiliation(s)
- Kyeong-Ok Choi
- Department of Food Science and Technology, Sejong University, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
179
|
The preventive effect of biochanin a on bone loss in ovariectomized rats: involvement in regulation of growth and activity of osteoblasts and osteoclasts. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:594857. [PMID: 23533501 PMCID: PMC3594937 DOI: 10.1155/2013/594857] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 12/27/2012] [Accepted: 01/17/2013] [Indexed: 11/18/2022]
Abstract
Biochanin A (BCA) is a major isoflavone abundant in red clover (Trifolium pretense). The protective effect of BCA on bone loss in an ovariectomized (OVX) animal model has never been clarified. The objective of this study was to investigate the biological effects of BCA on bone loss in OVX rats in vivo and on the development of osteoblasts and osteoclasts in vitro. Ovariectomy resulted in a marked increase in body weight and a decrease in femoral bone mineral density and trabecular bone volume that was prevented by BCA or 17 β -estradiol (E2) treatment. However, an increase in uterine weight was observed in E2-treated OVX rats, but not in response to BCA treatment. Treatment with BCA increased the mRNA expression of osterix, collagen type I, alkaline phosphatase (ALP), and osteocalcin and decreased the mRNA expression of tartrate-resistant acid phosphatase (TRAP) and the receptor activator of nuclear factor- κ B ligand (RANKL)/osteoprotegerin (OPG) ratio in the femur of OVX rats. Treatment with BCA or E2 prevented the OVX-induced increase in urinary deoxypyridinoline (DPD) and serum tumor necrosis factor α (TNF- α ) and interleukin-1 β (IL-1 β ). In vitro, BCA induced preosteoblasts to differentiate into osteoblasts and increased osteoblast mineralization. BCA inhibited preosteoclasts and osteoclast proliferation and decreased osteoclast bone resorption. These findings suggest that BCA treatment can effectively prevent the OVX-induced increase in bone loss and bone turnover possibly by increasing osteoblastic activities and decreasing osteoclastic activities.
Collapse
|
180
|
Yang N, Ginsburg GS, Simmons LA. Personalized medicine in women's obesity prevention and treatment: implications for research, policy and practice. Obes Rev 2013; 14:145-61. [PMID: 23114034 DOI: 10.1111/j.1467-789x.2012.01048.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 08/30/2012] [Accepted: 08/30/2012] [Indexed: 12/20/2022]
Abstract
The prevalence of obesity in America has reached epidemic proportions, and obesity among women is particularly concerning. Severe obesity (body mass index ≥35 kg m(-2) ) is more prevalent in women than men. Further, women have sex-specific risk factors that must be considered when developing preventive and therapeutic interventions. This review presents personalized medicine as a dynamic approach to obesity prevention, management and treatment for women. First, we review obesity as a complex health issue, with contributing sex-specific, demographic, psychosocial, behavioural, environmental, epigenetic and genetic/genomic risk factors. Second, we present personalized medicine as a rapidly advancing field of health care that seeks to quantify these complex risk factors to develop more targeted and effective strategies that can improve disease management and/or better minimize an individual's likelihood of developing obesity. Third, we discuss how personalized medicine can be applied in a clinical setting with current and emerging tools, including health risk assessments, personalized health plans, and strategies for increasing patient engagement. Finally, we discuss the need for additional research, training and policy that can enhance the practice of personalized medicine in women's obesity, including further advancements in the '-omics' sciences, physician training in personalized medicine, and additional development and standardization of innovative targeted therapies and clinical tools.
Collapse
Affiliation(s)
- N Yang
- Duke Center for Research on Prospective Health Care, Duke University School of Medicine, Durham, North Carolina, USA
| | | | | |
Collapse
|
181
|
Ha J, Cohen JI, Tirsi A, Convit A. Association of obesity-mediated insulin resistance and hypothalamic volumes: possible sex differences. DISEASE MARKERS 2013; 35:249-59. [PMID: 24344399 PMCID: PMC3810672 DOI: 10.1155/2013/531736] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 07/25/2013] [Accepted: 08/07/2013] [Indexed: 11/18/2022]
Abstract
The hypothalamus is important in hunger and metabolism. Although a lot is known about the basic role of the human hypothalamus, less is known about how the in vivo volume is affected in obesity, particularly among adolescents. Based on pediatric body mass index percentiles, 95 participants were assigned to lean or obese groups. All subjects had medical evaluations, including fasting blood tests, to assess insulin sensitivity and circulating CRP and neurotrophins (NGF and BDNF) and an MRI of the brain. Hypothalamic volumes were measured by a segmentation method combining manual and automated steps. Overall, obese participants had descriptively smaller hypothalamic volumes, although this difference did not reach statistical significance; however, among obese participants, females had significantly smaller hypothalamic volumes than their male counterparts. There was a significant interaction between insulin resistance and sex on hypothalamus volume; obese females with significant insulin resistance have smaller hypothalamic volumes than obese males. Obese adolescents had higher circulating CRP and neurotrophin levels. Furthermore, among obese females, BDNF concentrations were inversely associated with hypothalamus volumes (r = −0.48). Given this negative association between BDNF and hypothalamus volumes among obese insulin-resistant females, elevated neurotrophin levels may suggest an attempt at protective compensation.
Collapse
Affiliation(s)
- Jenny Ha
- Brain, Obesity, and Diabetes Laboratory (BODyLab), Department of Psychiatry, New York University School of Medicine, 145 East 32nd Street, 8th Floor, New York, NY 10016, USA
| | - Jessica I. Cohen
- Brain, Obesity, and Diabetes Laboratory (BODyLab), Department of Psychiatry, New York University School of Medicine, 145 East 32nd Street, 8th Floor, New York, NY 10016, USA
| | - Aziz Tirsi
- Brain, Obesity, and Diabetes Laboratory (BODyLab), Department of Psychiatry, New York University School of Medicine, 145 East 32nd Street, 8th Floor, New York, NY 10016, USA
| | - Antonio Convit
- Brain, Obesity, and Diabetes Laboratory (BODyLab), Department of Psychiatry, New York University School of Medicine, 145 East 32nd Street, 8th Floor, New York, NY 10016, USA
- Department of Medicine, New York University School of Medicine, 145 East 32nd Street, 8th Floor, New York, NY 10016, USA
- Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY 10962, USA
| |
Collapse
|
182
|
Clarke SD, Clarke IJ, Rao A, Evans RG, Henry BA. Differential effects of acute and chronic estrogen treatment on thermogenic and metabolic pathways in ovariectomized sheep. Endocrinology 2013. [PMID: 23183185 DOI: 10.1210/en.2012-1758] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Estrogen is protective against weight gain, but the underlying mechanisms are not fully elucidated. We sought to characterize the effects of estrogen on energy expenditure in skeletal muscle and adipose tissue in ovariectomized sheep. Temperature probes were implanted into sc (gluteal) and visceral (retroperitoneal) fat depots and skeletal muscle of the hind limb (vastus lateralis). Food was available from 1100-1600 h to entrain postprandial thermogenesis. We characterized the effects of single (50 μg estradiol benzoate, im) and repeated (25 μg estradiol-17β, iv) injections as well as chronic (3 × 3 cm estradiol-17β implants for 7 d) treatment on heat production. A single injection of estrogen increased heat production in visceral fat and skeletal muscle, without an effect on food intake. Increased heat production in skeletal muscle was sustained by repeated estradiol-17β injections. On the other hand, continuous treatment reduced food intake but had no effect on thermogenesis. To determine possible mechanisms that underpin estradiol-17β-induced heat production, we measured femoral artery blood flow, the expression of uncoupling protein (UCP) mRNA and the phosphorylation of AMP-activated protein kinase and Akt in fat and muscle. There was little effect of either single or repeated injections of estradiol-17β on the expression of UCP1, -2, or -3 mRNA in visceral fat or skeletal muscle. Acute injection of estradiol-17β increased the phosphorylation of AMP-activated protein kinase and Akt in muscle only. Estradiol-17β treatment did not alter femoral artery blood flow. Thus, the stimulatory effect of estradiol-17β on thermogenesis in female sheep is dependent upon a pulsatile pattern of treatment and not constant continuous exposure.
Collapse
Affiliation(s)
- Scott D Clarke
- Research Fellow, Monash University, Physiology, Building 13F, Wellington Road, Clayton, Victoria 3168, Australia
| | | | | | | | | |
Collapse
|
183
|
Christoffersen B, Golozoubova V, Pacini G, Svendsen O, Raun K. The young Göttingen minipig as a model of childhood and adolescent obesity: influence of diet and gender. Obesity (Silver Spring) 2013; 21:149-58. [PMID: 23505180 DOI: 10.1002/oby.20249] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2012] [Accepted: 05/28/2012] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Gender and sex hormones influence the development of obesity and metabolic syndrome in humans and Göttingen minipigs. The aim of this study was to investigate possible gender differences in the metabolic response to a high energy diet in young Göttingen minipigs as a model of childhood/adolescent obesity. DESIGN AND METHODS Nine-week-old male and female Göttingen minipigs were fed restrictedly on either a low energy diet (LED) or a high energy diet (HED) for 4 months (n = 5-7). Parameters of interest were fat percentage, visceral fat mass, plasma lipids and glucose tolerance, insulin resistance, and β-cell function measured by oral and intravenous glucose tolerance tests. RESULTS At 11 to 12 weeks of age, after 2 weeks diet feeding, both genders on HED had increased fat percentage, glucose intolerance, decreased insulin sensitivity, and increased plasma levels of cholesterol and triglycerides (TGs). There was no gender difference in body weight (BW) or fat percentage, but males had lower glucose tolerance than females. After 3.5 to 4 months on the diets, the pigs on HED had increased BW, fat percentage, and visceral fat mass and were more glucose intolerant and insulin resistant than pigs on LED. Also increases in plasma cholesterol and TG levels were observed in the pigs on HED. Females had higher fat percentage and more visceral fat, were more insulin resistant, and had a more unfavorable lipid profile compared with males independent of diet. CONCLUSION In conclusion, the young Göttingen minipig, and especially the female gender, seems to be a potential model for diet induced childhood/adolescent obesity and metabolic syndrome.
Collapse
|
184
|
Shi H, Kumar SPDS, Liu X. G protein-coupled estrogen receptor in energy homeostasis and obesity pathogenesis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 114:193-250. [PMID: 23317786 PMCID: PMC3632385 DOI: 10.1016/b978-0-12-386933-3.00006-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Obesity and its related metabolic diseases have reached a pandemic level worldwide. There are sex differences in the prevalence of obesity and its related metabolic diseases, with men being more vulnerable than women; however, the prevalence of these disorders increases dramatically in women after menopause, suggesting that sex steroid hormone estrogens play key protective roles against development of obesity and metabolic diseases. Estrogens are important regulators of several aspects of metabolism, including body weight and body fat, caloric intake and energy expenditure, and glucose and lipid metabolism in both males and females. Estrogens act in complex ways on their nuclear estrogen receptors (ERs) ERα and ERβ and transmembrane ERs such as G protein-coupled estrogen receptor. Genetic tools, such as different lines of knockout mouse models, and pharmacological agents, such as selective agonists and antagonists, are available to study function and signaling mechanisms of ERs. We provide an overview of the evidence for the physiological and cellular actions of ERs in estrogen-dependent processes in the context of energy homeostasis and body fat regulation and discuss its pathology that leads to obesity and related metabolic states.
Collapse
Affiliation(s)
- Haifei Shi
- Department of Biology, Center for Physiology and Neuroscience, Miami University, Oxford, Ohio, USA
| | | | | |
Collapse
|
185
|
Kwok S, Canoy D, Soran H, Ashton DW, Lowe GDO, Wood D, Humphries SE, Durrington PN. Body fat distribution in relation to smoking and exogenous hormones in British women. Clin Endocrinol (Oxf) 2012; 77:828-33. [PMID: 23137104 DOI: 10.1111/j.1365-2265.2012.04331.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Both cigarette smoking and use of exogenous hormones are associated with changes in regional distribution of body fat, but their combined effects are less investigated. We examined the interrelation between smoking, exogenous hormones and fat distribution in premenopausal and postmenopausal women. METHOD We used data from 20, 962 women without known cardiovascular disease (CVD) who were employees of a major department store in Britain. They completed a health questionnaire and attended a clinical examination that included waist and hip circumference measurements. The cross-sectional analyses were conducted using linear regression models. RESULTS Cigarette smoking, particularly smoking ≥20 cigarettes/day, was associated with larger waist circumference and higher waist/hip ratio (WHR) in pre- and postmenopausal women after adjusting for potential confounding factors (all P < 0·001). Premenopausal women using combined oral contraceptive (COC) and postmenopausal women using oestrogen-only hormone replacement therapy (HRT) had lower WHR than non-hormone users in both smokers and nonsmokers. However, smokers had higher WHR than nonsmokers in both groups of hormone users and nonusers. There was no significant interaction between smoking and hormone use in premenopausal and postmenopausal women (P > 0·05). CONCLUSION Although exogenous hormones use was related to a more favourable fat distribution in women, smoking was associated with greater abdominal fat accumulation.
Collapse
Affiliation(s)
- See Kwok
- School of Biomedicine, University of Manchester, Manchester, UK.
| | | | | | | | | | | | | | | |
Collapse
|
186
|
Male mice that lack the G-protein-coupled receptor GPR41 have low energy expenditure and increased body fat content. Br J Nutr 2012; 109:1755-64. [PMID: 23110765 DOI: 10.1017/s0007114512003923] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
SCFA are produced in the gut by bacterial fermentation of undigested carbohydrates. Activation of the Gαi-protein-coupled receptor GPR41 by SCFA in β-cells and sympathetic ganglia inhibits insulin secretion and increases sympathetic outflow, respectively. A possible role in stimulating leptin secretion by adipocytes is disputed. In the present study, we investigated energy balance and glucose homoeostasis in GPR41 knockout mice fed on a standard low-fat or a high-fat diet. When fed on the low-fat diet, body fat mass was raised and glucose tolerance was impaired in male but not female knockout mice compared to wild-type mice. Soleus muscle and heart weights were reduced in the male mice, but total body lean mass was unchanged. When fed on the high-fat diet, body fat mass was raised in male but not female GPR41 knockout mice, but by no more in the males than when they were fed on the low-fat diet. Body lean mass and energy expenditure were reduced in male mice but not in female knockout mice. These results suggest that the absence of GPR41 increases body fat content in male mice. Gut-derived SCFA may raise energy expenditure and help to protect against obesity by activating GPR41.
Collapse
|
187
|
Two different isomers of vitamin e prevent bone loss in postmenopausal osteoporosis rat model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 2012:161527. [PMID: 23118785 PMCID: PMC3484319 DOI: 10.1155/2012/161527] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 09/15/2012] [Indexed: 11/17/2022]
Abstract
Postmenopausal osteoporotic bone loss occurs mainly due to cessation of ovarian function, a condition associated with increased free radicals. Vitamin E, a lipid-soluble vitamin, is a potent antioxidant which can scavenge free radicals in the body. In this study, we investigated the effects of alpha-tocopherol and pure tocotrienol on bone microarchitecture and cellular parameters in ovariectomized rats. Three-month-old female Wistar rats were randomly divided into ovariectomized control, sham-operated, and ovariectomized rats treated with either alpha-tocopherol or tocotrienol. Their femurs were taken at the end of the four-week study period for bone histomorphometric analysis. Ovariectomy causes bone loss in the control group as shown by reduction in both trabecular volume (BV/TV) and trabecular number (Tb.N) and an increase in trabecular separation (Tb.S). The increase in osteoclast surface (Oc.S) and osteoblast surface (Ob.S) in ovariectomy indicates an increase in bone turnover rate. Treatment with either alpha-tocopherol or tocotrienol prevents the reduction in BV/TV and Tb.N as well as the increase in Tb.S, while reducing the Oc.S and increasing the Ob.S. In conclusion, the two forms of vitamin E were able to prevent bone loss due to ovariectomy. Both tocotrienol and alpha-tocopherol exert similar effects in preserving bone microarchitecture in estrogen-deficient rat model.
Collapse
|
188
|
Abstract
Diabetic men have benefited in the last 30 years from a significant improvement in total and cardiovascular mortality, whereas diabetic women have had no improvement at all. Moreover, recent research focused on the role of sex hormones in glucose homeostasis, and might account for different pathophysiologic mechanisms in the development of diabetes-related complications. Thus, care of diabetic women is a challenge that requires particular attention. The available data regarding gender-specific care of diabetes mellitus are uneven, rich in some domains but very poor in others. The large prospective trials performed in the last 20 years have assumed that the natural history of diabetes mellitus in men and women, as well as the efficiency of glucose-lowering therapies and management of hyperglycemic-related complications, could be attributable without distinction to men and women. We propose in this paper to analyze the published medical literature according to the specific management of diabetes mellitus in women, and to try to distinguish some particular features. We found important distinctions between diabetic men and women regarding the patterns of abnormalities of glucose regulation, epidemiology, development of diabetes-related complications, ischemic heart disease, morbidity and mortality, impact of cardiovascular risk factors, development of the metabolic syndrome, depression and osteoporosis, as well as the impact of lifestyle modifications or primary and secondary preventions on cardiovascular risk factors, and finally medical therapeutics. Moreover, special considerations were given to some particular aspects of the medical life in diabetic women, such as the features of gestational diabetes mellitus and the management of pregnancy in pregestational diabetic women, use of contraception, hormone-replacement therapy and polycystic ovary syndrome.
Collapse
Affiliation(s)
- Auryan Szalat
- Hadassah Hebrew University Hospital, Internal Medicine, Endocrinology and Metabolism, Jerusalem, Israel.
| | | |
Collapse
|
189
|
Hammarstedt A, Graham TE, Kahn BB. Adipose tissue dysregulation and reduced insulin sensitivity in non-obese individuals with enlarged abdominal adipose cells. Diabetol Metab Syndr 2012; 4:42. [PMID: 22992414 PMCID: PMC3523053 DOI: 10.1186/1758-5996-4-42] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 09/17/2012] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Obesity contributes to Type 2 diabetes by promoting systemic insulin resistance. Obesity causes features of metabolic dysfunction in the adipose tissue that may contribute to later impairments of insulin action in skeletal muscle and liver; these include reduced insulin-stimulated glucose transport, reduced expression of GLUT4, altered expression of adipokines, and adipocyte hypertrophy. Animal studies have shown that expansion of adipose tissue alone is not sufficient to cause systemic insulin resistance in the absence of adipose tissue metabolic dysfunction. To determine if this holds true for humans, we studied the relationship between insulin resistance and markers of adipose tissue dysfunction in non-obese individuals. METHOD 32 non-obese first-degree relatives of Type 2 diabetic patients were recruited. Glucose tolerance was determined by an oral glucose tolerance test and insulin sensitivity was measured with the hyperinsulinaemic-euglycaemic clamp. Blood samples were collected and subcutaneous abdominal adipose tissue biopsies obtained for gene/protein expression and adipocyte cell size measurements. RESULTS Our findings show that also in non-obese individuals low insulin sensitivity is associated with signs of adipose tissue metabolic dysfunction characterized by low expression of GLUT4, altered adipokine profile and enlarged adipocyte cell size. In this group, insulin sensitivity is positively correlated to GLUT4 mRNA (R = 0.49, p = 0.011) and protein (R = 0.51, p = 0.004) expression, as well as with circulating adiponectin levels (R = 0.46, 0 = 0.009). In addition, insulin sensitivity is inversely correlated to circulating RBP4 (R = -0.61, 0 = 0.003) and adipocyte cell size (R = -0.40, p = 0.022). Furthermore, these features are inter-correlated and also associated with other clinical features of the metabolic syndrome in the absence of obesity. No association could be found between the hypertrophy-associated adipocyte dysregulation and HIF-1alpha in this group of non-obese individuals. CONCLUSIONS In conclusion, these findings support the concept that it is not obesity per se, but rather metabolic dysfunction of adipose tissue that is associated with systemic insulin resistance and the metabolic syndrome.
Collapse
Affiliation(s)
- Ann Hammarstedt
- The Lundberg Laboratory for Diabetes Research, Center of Excellence for Metabolic and Cardiovascular Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, SE-413 45, Sweden
| | - Timothy E Graham
- Division of Endocrinology, Diabetes and Medicine, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Barbara B Kahn
- Division of Endocrinology, Diabetes and Medicine, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
190
|
Pohjanvirta R, Miettinen H, Sankari S, Hegde N, Lindén J. Unexpected gender difference in sensitivity to the acute toxicity of dioxin in mice. Toxicol Appl Pharmacol 2012; 262:167-76. [DOI: 10.1016/j.taap.2012.04.032] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 04/25/2012] [Accepted: 04/26/2012] [Indexed: 10/28/2022]
|
191
|
Given breast cancer, does breast size matter? Data from a prospective breast cancer cohort. Cancer Causes Control 2012; 23:1307-16. [DOI: 10.1007/s10552-012-0008-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 05/31/2012] [Indexed: 10/28/2022]
|
192
|
Zhang H, Chen X, Sairam MR. Novel genes of visceral adiposity: identification of mouse and human mesenteric estrogen-dependent adipose (MEDA)-4 gene and its adipogenic function. Endocrinology 2012; 153:2665-76. [PMID: 22510272 DOI: 10.1210/en.2011-2008] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Visceral adiposity represents a high risk factor for type 2 diabetes, metabolic syndrome, and cardiovascular disease as well as various cancers. While studying sex hormone imbalance-induced early obesity and late onset of insulin resistance in FSH receptor knock out female mice, we identified a novel mesenteric estrogen-dependent adipose gene (MEDA-4) selectively up-regulated in a depot-specific manner in mesenteric adipose tissue. Meda-4 cloned from both mouse and human adipose tissue codes for a 34-kDa cytosolic protein with 91% homology. Mouse Meda-4 mRNA is expressed highest in visceral adipose tissue and localizes predominantly in the adipocyte fraction. Human MEDA-4 is also more abundant in omental fat than sc depot in obese patients. In 3T3-L1 cells endogenous Meda-4 expression increases early during differentiation, and its overexpression promotes differentiation of preadipocytes into adipocytes and enhances glucose uptake. Conversely, short hairpin RNA-mediated knockdown of Meda-4 reduces both adipogenic and glucose uptake potential. In promoting adipogenesis, Meda-4 up-regulates transcription factor peroxisome proliferator-activated receptor-γ2. Meda-4 promotes lipid accumulation in adipocytes, regulating adipocyte fatty acid-binding protein 2, CD36, lipoprotein lipase, hormone-sensitive lipase, acyl-Coenzyme A oxidase-1, perilipin-1, and fatty acid synthase expression. 17β-Estradiol reduced Meda-4 expression in mesenteric adipose tissue of ovariectomized mice and in 3T3-L1 adipocytes. Thus our study identifies Meda-4 as a novel adipogenic gene, capable of promoting differentiation of preadipocytes into adipocytes, increasing lipid content and glucose uptake in adipocytes. Therefore it might play an important role in adipose tissue expansion in normal and aberrant hormonal conditions and pathophysiological states.
Collapse
Affiliation(s)
- H Zhang
- Molecular Endocrinology Laboratory, Institut de Recherches Cliniques de Montréal, Montréal, Quebec, Canada H2W 1R7
| | | | | |
Collapse
|
193
|
Cao J, Chen TM, Hao WJ, Li J, Liu L, Zhu BP, Li XY. Correlation between sex hormone levels and obesity in the elderly male. Aging Male 2012; 15:85-9. [PMID: 22452321 DOI: 10.3109/13685538.2012.666585] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To investigate the levels of sex hormones and androgen receptor (AR) in elderly male patients and to explore a possible correlation with obesity. METHODS The cross-sectional study included 314 Elderly males (age ≥ 65 year). Of these subjects, 104 were healthy (age range 65-92 year; mean 71.38 ± 5.154 year), 74 were obese (65-87 year; 71.32 ± 4.74 year), and 111 were overweight (65-85 year; 71.43 ± 5.03 year). The following parameters were measured: total testosterone (TT), free testosterone, dehydroepiandrosterone sulfate, sex hormone-binding globulin (SHBG), estradiol (E2), luteinizing hormone, follicle-stimulating hormone and AR. RESULTS (i) The levels of TT and SHBG in the obesity group were significantly lower than those in non-obese subjects. (ii) Body mass index (BMI) negatively correlated with TT and SHBG. (iii) Multiple regression analysis revealed that TT (β: -0.230; p = 0.045) and SHBG (β: -0.163; p = 0.02) were statistically correlated with BMI. CONCLUSION Testosterone levels in the obese population were significantly lower than in the non-obese population and there is a significant association between testosterone levels and the extent of obesity.
Collapse
Affiliation(s)
- Jian Cao
- First Geriatric Cardiology Division, Chinese PLA General Hospital, Beijing, PR China.
| | | | | | | | | | | | | |
Collapse
|
194
|
|
195
|
Dietary supplementation with methyl donors reduces fatty liver and modifies the fatty acid synthase DNA methylation profile in rats fed an obesogenic diet. GENES AND NUTRITION 2012; 8:105-13. [PMID: 22648174 DOI: 10.1007/s12263-012-0300-z] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 05/11/2012] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the first hepatic manifestations of metabolic syndrome, whose progression can lead to cirrhosis and hepatic carcinoma. Interestingly, methyl donor supplementation could improve obesogenic diet-induced hepatic triglyceride accumulation. The aim of this research is to describe methyl donor effects on a high-fat-sucrose (HFS) diet in both sexes and epigenetic changes induced on fatty acid synthase (FASN) promoter methylation pattern as well as gene expression of NAFLD key metabolic genes. Twenty-four male and 28 female Wistar rats were assigned to three dietary groups: control, HFS, and HFS supplemented with methyl donors (choline, betaine, vitamin B12, and folic acid). After 8 weeks of treatment, somatic, biochemical, mRNA, and epigenetic measurements were performed. Rats fed the HFS diet presented an overweight phenotype and alterations in plasma biochemical measurements. Methyl donor supplementation reverted the HFS-diet-induced hepatic triglyceride accumulation. Analysis of FASN promoter cytosine methylation showed changes in both sexes due to the obesogenic diet at -1,096, -780, -778, and -774 CpG sites with respect to the transcriptional start site. Methyl donor supplementation modified DNA methylation at -852, -833, -829, -743, and -733 CpGs depending on the sex. RT-PCR analysis confirmed that FASN expression tended to be altered in males. Our findings reinforce the hypothesis that methyl donor supplementation can prevent hepatic triglyceride accumulation induced by obesogenic diets in both sexes. Changes in liver gene expression profile and epigenetic-mediated mechanisms related to FASN DNA hypermethylation could be involved in methyl donor-induced NAFLD improvement.
Collapse
|
196
|
Wang SJ, Li XF, Jiang LS, Dai LY. Estrogen stimulates leptin receptor expression in ATDC5 cells via the estrogen receptor and extracellular signal-regulated kinase pathways. J Endocrinol 2012; 213:163-72. [PMID: 22396455 DOI: 10.1530/joe-11-0353] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Regulation of the physiological processes of endochondral bone formation during long bone growth is controlled by various factors including the hormones estrogen and leptin. The effects of estrogen are mediated not only through the direct activity of estrogen receptors (ERs) but also through cross talk with other signaling systems implicated in chondrogenesis. The receptors of both estrogen and leptin (OBR (LEPR)) are detectable in growth plate chondrocytes of all zones. In this study, the expression of mRNA and protein of OBR in chondrogenic ATDC5 cells and the effect of 17β-estradiol (E(2)) stimulation were assessed using quantitative PCR and western blotting. We have found that the mRNA of Obr was dynamically expressed during the differentiation of ATDC5 cells over 21 days. Application of E(2) (10(-7) M) at day 14 for 48 h significantly upregulated OBR mRNA and protein levels (P<0.05). The upregulation of Obr mRNA by E(2) was shown to take place in a concentration-dependent manner, with a concentration of 10(-7) M E(2) having the greatest effect. Furthermore, we have confirmed that E(2) affected the phosphorylation of ERK1/2 (MAPK1/MAPK3) in a time-dependent manner where a maximal fourfold change was observed at 10 min following application of E(2). Finally, pretreatment of the cells with either U0126 (ERK1/2 inhibitor) or ICI 182 780 (ER antagonist) blocked the upregulation of OBR by E(2) and prevented the E(2)-induced phosphorylation of ERK. These data demonstrate, for the first time, the existence of cross talk between estrogen and OBR in the regulation of bone growth whereby estrogen regulates the expression of Obr in growth plate chondrocytes via ERs and the activation of ERK1/2 signaling pathways.
Collapse
Affiliation(s)
- Shan-Jin Wang
- Department of Orthopedic Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China
| | | | | | | |
Collapse
|
197
|
Amengual-Cladera E, Lladó I, Gianotti M, Proenza AM. Retroperitoneal white adipose tissue mitochondrial function and adiponectin expression in response to ovariectomy and 17β-estradiol replacement. Steroids 2012; 77:659-65. [PMID: 22402114 DOI: 10.1016/j.steroids.2012.02.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 02/16/2012] [Accepted: 02/17/2012] [Indexed: 01/12/2023]
Abstract
Sexual dimorphism has been previously found both in mitochondrial biogenesis and function and in adiponectin expression of retroperitoneal WAT. However, little is known about the E2 effects on WAT mitochondrial function. Accordingly, the aim of this study was to examine in greater depth the role of estrogens in sexual dimorphism. This was accomplished by studying the effects of ovariectomy and E2 replacement on retroperitoneal WAT mitochondrial function. Fourteen-week-old female and ovariectomized (OVX) female Wistar rats were used in this study. The ovariectomy was performed at 5 weeks of age and at 10 weeks of age OVX rats were divided into two experimental groups: OVX, and OVX treated with 17β-estradiol (E2) (OVX+E2). Subcutaneous injections of E2 (10 μg/kg/48 h) were administered to the OVX+E2 rats for 4 weeks previous to the sacrifice whereas OVX rats were treated only with the vehicle. Levels of the main markers for mitochondrial biogenesis and function and those representatives of the antioxidant defense system and insulin sensitivity were determined. Additionally, the mRNA levels of the α and β estrogen receptors and of some adipocyte differentiation markers were studied. Our results indicate that retroperitoneal WAT was able to adapt itself to ovariectomy without any changes in mitochondrial function markers or for the adiponectin levels. However, E2 supplementation led to an unexpected decrease in: TFAM protein levels, in LPL, PPARγ and adiponectin gene expression and in the systemic HMW adiponectin levels. This decrease is probably due to the down-regulation of the ERα mRNA expression to avoid an over-stimulation by E2.
Collapse
Affiliation(s)
- Emilia Amengual-Cladera
- Grup Metabolisme Energètic i Nutrició, Departament de Biologia Fonamental i Ciències de la Salut, Institut Universitari d'Investigació en Ciències de la Salut, Universitat de les Illes Balears, Palma de Mallorca, Spain.
| | | | | | | |
Collapse
|
198
|
Wiren KM, Hashimoto JG, Zhang XW. Stem cell activation in adults can reverse detrimental changes in body composition to reduce fat and increase lean mass in both sexes. J Cell Biochem 2012; 112:3638-47. [PMID: 21793043 DOI: 10.1002/jcb.23288] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Detrimental changes in body composition are often associated with declining levels of testosterone. Here, we evaluated the notion that multipotent mesenchymal stem cells, that give rise to both fat and muscle tissue, can play a significant role to alter existing body composition in the adult. Transgenic mice with targeted androgen receptor (AR) overexpression in stem cells were employed. Wild-type littermate and AR-transgenic male and female mice were gonadectomized and left untreated for 2 months. After the hypogonadal period, mice were then treated with 5α-dihydrotestosterone (DHT) for 6 weeks. After orchidectomy (ORX), wild-type males have reduced lean mass and increased fat mass compared to shams. DHT treatment was beneficial to partially restore body composition. In wild-type females, ovariectomy (OVX) produced a similar change but there was no improvement with DHT. In targeted AR transgenic mice, DHT treatment increased lean and reduced fat mass to sham levels. In contrast to wild-type females, DHT treatment in female transgenic mice significantly ameliorated the increased fat and decreased lean mass changes that result after OVX. Our results show that DHT administration reduces fat mass and increases lean mass in wild-type males but not females, indicating that wild-type females are not as sensitive to androgen treatment. Because both male and female transgenic mice are more responsive than wild-type, results suggest that body composition remains linked to stem cell fate in the adult and that targeted androgen signaling in stem cells can play a significant role to reverse detrimental changes in body composition in both sexes.
Collapse
Affiliation(s)
- Kristine M Wiren
- Research Service, Veterans Affairs Medical Center, Portland, OR 97239, USA.
| | | | | |
Collapse
|
199
|
Mastaglia SR, Solis F, Bagur A, Mautalen C, Oliveri B. Increase in android fat mass with age in healthy women with normal body mass index. J Clin Densitom 2012; 15:159-64. [PMID: 22402116 DOI: 10.1016/j.jocd.2011.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 12/08/2011] [Accepted: 12/21/2011] [Indexed: 11/17/2022]
Abstract
Body fat distribution is gender specific: men tend to accumulate adipose tissue in the android region, whereas women tend to do so in the gynoid region. The aim of the study was to assess total fat mass (TFM), android fat (AF), and gynoid fat (GF) mass in a selected group of healthy adult women with normal body mass index (BMI) to evaluate variations in fat distribution. Seventy-seven women (20--69yr of age) with BMI values between ≥18.5 and ≤24.9kg/m(2) were included. TMF, AF, GF, and the AF to GF ratio (A:G) were assessed using dual-energy X-ray absorptiometry. Results showed an increase in AF after the fifth decade of life (D), which reached statistical significance in the sixth and seventh decades (p<0.05--0.008), a 33% increase in kg of AF between the fourth and seventh and a 20% increase in A:G between the third and the seventh, with no significant changes in TFM and GF. In normal BMI women, age appears to be associated with changes in fat mass distribution with an increase in AF, which might have potential deleterious health consequences, after the fifth D.
Collapse
Affiliation(s)
- Silvina R Mastaglia
- Sección Osteopatías Médicas, Hospital de Clínicas, Universidad de Buenos Aires, Buenos Aires, Argentina.
| | | | | | | | | |
Collapse
|
200
|
High-dose testosterone propionate treatment reverses the effects of endurance training on myocardial antioxidant defenses in adolescent male rats. Cardiovasc Toxicol 2012; 11:118-27. [PMID: 21312070 PMCID: PMC3085793 DOI: 10.1007/s12012-011-9105-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This study was aimed at evaluation of changes in activities of selected antioxidant enzymes (superoxide dismutase, catalase, glutathione peroxidase, and glutathione reductase) and contents of key nonenzymatic antioxidants (glutathione, protein thiol groups, and α- and γ-tocopherols) in the left heart ventricle of young male Wistar rats subjected to endurance training (treadmill running, 1 h daily, 5 days a week, for 6 weeks) or/and testosterone propionate treatment (8 or 80 mg/kg body weight, intramuscularly, once a week, for 6 weeks) during adolescence. The training alone increased the activities of key antioxidant enzymes, but lowered the pool of nonenzymatic antioxidants and enhanced myocardial oxidative stress as evidenced by elevation of the lipid peroxidation biomarker malondialdehyde. The lower-dose testosterone treatment showed mixed effects on the individual components of the antioxidant defense system, but markedly enhanced lipid peroxidation. The higher-dose testosterone treatment decreased the activities of the antioxidant enzymes, lowered the contents of the nonenzymatic antioxidants, except for that of γ-tocopherol, reversed the effect of endurance training on the antioxidant enzymes activities, and enhanced lipid peroxidation more than the lower-dose treatment. These data demonstrate the potential risk to cardiac health from exogenous androgen use, either alone or in combination with endurance training, in adolescents.
Collapse
|