51
|
Karastergiou K, Smith SR, Greenberg AS, Fried SK. Sex differences in human adipose tissues - the biology of pear shape. Biol Sex Differ 2012; 3:13. [PMID: 22651247 PMCID: PMC3411490 DOI: 10.1186/2042-6410-3-13] [Citation(s) in RCA: 648] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 05/31/2012] [Indexed: 12/15/2022] Open
Abstract
Women have more body fat than men, but in contrast to the deleterious metabolic consequences of the central obesity typical of men, the pear-shaped body fat distribution of many women is associated with lower cardiometabolic risk. To understand the mechanisms regulating adiposity and adipose tissue distribution in men and women, significant research attention has focused on comparing adipocyte morphological and metabolic properties, as well as the capacity of preadipocytes derived from different depots for proliferation and differentiation. Available evidence points to possible intrinsic, cell autonomous differences in preadipocytes and adipocytes, as well as modulatory roles for sex steroids, the microenvironment within each adipose tissue, and developmental factors. Gluteal-femoral adipose tissues of women may simply provide a safe lipid reservoir for excess energy, or they may directly regulate systemic metabolism via release of metabolic products or adipokines. We provide a brief overview of the relationship of fat distribution to metabolic health in men and women, and then focus on mechanisms underlying sex differences in adipose tissue biology.
Collapse
Affiliation(s)
- Kalypso Karastergiou
- Department of Medicine, Section of Endocrinology, Diabetes & Nutrition, Boston University School of Medicine, Boston, MA, USA.
| | | | | | | |
Collapse
|
52
|
Genetic polymorphisms in estrogen receptors and sexual dimorphism in fat redistribution in HIV-infected patients on HAART. AIDS 2012; 26:19-26. [PMID: 22011627 DOI: 10.1097/qad.0b013e32834db3ac] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To investigate genetic single nucleotide polymorphisms (SNPs) in estrogen receptor-α (ERα) (ESR1, rs2234693, rs1801132, rs7757956 and rs2813544) and ERβ (ESR2, rs3020450, rs7154455 and rs4986938) genes and relate them to the adverse effects lipodystrophy, dyslipidemia and metabolic syndrome as well as to differences in their prevalence between sexes in HIV-infected individuals on HAART. DESIGN Cross-sectional study. METHODS Blood samples and anthropometric measurements were collected from 614 patients at reference services in the cities of Porto Alegre, Pelotas and Rio Grande in Brazil. The SNPs were genotyped by real-time PCR. RESULTS The lipodystrophy subtype frequencies in patients of different sexes showed statistically significant differences; the atrophic pattern was more prevalent in men, and the hypertrophic pattern was more prevalent in women. Furthermore, metabolic syndrome prevalence was higher in women than in men. The ESR1 rs2813544 G-allele was associated with higher measurements of several anthropometric variables in women: BMI, total subcutaneous fat and subcutaneous fat of limbs. Additionally, patients who were AA homozygous for ESR2 rs3020450 presented an increased risk for developing lipoatrophy (prevalence ratio 1.37, 95% confidence interval 1.09-1.73, P = 0.007). CONCLUSION Significant differences in lipodystrophy and metabolic syndrome prevalence were detected between sexes. Moreover, the ESR1 gene (rs2813544) presented significant sex-specific associations with anthropometric variables, and the ESR2 gene (rs3020450) was associated with an increased risk of developing lipoatrophy. Our results suggest that these genes are in part responsible for the sexual dimorphism in fat tissue redistribution and patterns of lipodystrophy.
Collapse
|
53
|
Mruk DD, Cheng CY. Environmental contaminants: Is male reproductive health at risk? SPERMATOGENESIS 2011; 1:283-290. [PMID: 22332111 PMCID: PMC3271639 DOI: 10.4161/spmg.1.4.18328] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 09/21/2011] [Accepted: 09/22/2011] [Indexed: 02/06/2023]
Abstract
Contaminants such as cadmium, bisphenol A and lead pollute our environment and affect male reproductive function. There is evidence that toxicant exposure adversely affects fertility. Cadmium and bisphenol A exert their effects in the testis by perturbing blood-testis barrier function, which in turn affects germ cell adhesion in the seminiferous epithelium because of a disruption of the functional axis between these sites. In essence, cadmium mediates its adverse effects at the blood-testis barrier by disrupting cell adhesion protein complexes, illustrating that toxicants can dismantle cell junctions in the testis. Herein, we will discuss how environmental toxicants may affect reproductive function. We will also examine how these adverse effects on fertility may be mediated in part by adipose tissue and bone. Lastly, we will briefly discuss how toxicant-induced damage may be effectively managed so that fertility can be maintained. It is hoped that this information will offer a new paradigm for future studies.
Collapse
Affiliation(s)
- Dolores D Mruk
- The Mary M. Wohlford Laboratory for Male Contraceptive Research; Center for Biomedical Research; The Population Council; New York, NY USA
| | | |
Collapse
|
54
|
Saleh J, Al-Wardy N, Farhan H, Al-Khanbashi M, Cianflone K. Acylation stimulating protein: a female lipogenic factor? Obes Rev 2011; 12:440-8. [PMID: 21348923 DOI: 10.1111/j.1467-789x.2010.00832.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Acylation stimulating protein (ASP) is a potent lipogenic factor produced from adipocytes. Plasma ASP levels were shown to increase in obesity, diabetes mellitus type II and dyslipidemia, and decrease after weight loss and fasting. Growing evidence suggests that ASP may significantly contribute to subcutaneous fat storage in females. In vitro, ASP stimulated triglyceride synthesis to a larger extent in subcutaneous compared with omental adipocytes. The ASP receptor binding affinity to plasma membranes prepared from adipose tissue showed higher binding affinity to plasma membranes from female adipose tissue compared with male adipose tissue, and was more pronounced to subcutaneous compared with omental plasma membranes. Human studies demonstrated that postprandial triglyceride clearance predicted by ASP levels was more efficient in women than in men. In mice, postprandial triglyceride clearance, with intraperitoneal ASP administration, was faster in females compared with males. The ASP deficient mice were resistant to weight gain and had reduced fat mass that was more pronounced in females. Recent findings in humans and mice point to a significant association between progesterone and ASP variations in females. In this review, we highlight findings, to date, linking ASP to physiological and hormonal alterations that may contribute to subcutaneous fat distribution typical to females.
Collapse
Affiliation(s)
- J Saleh
- Biochemistry Department, Faculty of Medicine, Sultan Qaboos University, P.O. Box 35, 123 Muscat, Oman.
| | | | | | | | | |
Collapse
|
55
|
Leung YK, Lam HM, Wu S, Song D, Levin L, Cheng L, Wu CL, Ho SM. Estrogen receptor beta2 and beta5 are associated with poor prognosis in prostate cancer, and promote cancer cell migration and invasion. Endocr Relat Cancer 2010; 17:675-89. [PMID: 20501637 PMCID: PMC2891483 DOI: 10.1677/erc-09-0294] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Estrogens play a pivotal role in the development and progression of prostate cancer (PCa). Their actions are mediated by estrogen receptors (ERs), particularly ERbeta in the prostate epithelium. With the discovery of ERbeta isoforms, data from previous studies that focused principally on the wild-type ERbeta (ERbeta1) may not be adequate in explaining the still controversial role of ERbeta(s) in prostate carcinogenesis. In this study, using newly generated isoform-specific antibodies, immunohistochemistry (IHC) was performed on a tumor microarray comprised of 144 specimens. IHC results were correlated with pathological and clinical follow-up data to delineate the distinct roles of ERbeta1, ERbeta2, and ERbeta5 in PCa. ERbeta2 was commonly found in the cytoplasm and was the most abundant isoform followed by ERbeta1 localized predominantly in the nucleus, and ERbeta5 was primarily located in the cytoplasm. Logistic regression analyses demonstrated that nuclear ERbeta2 (nERbeta2) is an independent prognostic marker for prostate specific antigen (PSA) failure and postoperative metastasis (POM). In a Kaplan-Meier analysis, the combined expression of both nERbeta2 and cytoplasmic ERbeta5 identified a group of patients with the shortest POM-free survival. Cox proportional hazard models revealed that nERbeta2 predicted shorter time to POM. In concordance with IHC data, stable, ectopic expression of ERbeta2 or ERbeta5 enhanced PCa cell invasiveness but only PCa cells expressing ERbeta5 exhibited augmented cell migration. This is the first study to uncover a metastasis-promoting role of ERbeta2 and ERbeta5 in PCa, and show that the two isoforms, singularly and conjointly, have prognostic values for PCa progression. These findings may aid future clinical management of PCa.
Collapse
Affiliation(s)
- Yuet-Kin Leung
- Division of Environmental Genetics and Molecular Toxicology, Department of Environmental HealthCollege of Medicine, University of Cincinnati Medical CenterCincinnati, Ohio, 45267USA
- Center for Environmental GeneticsCollege of Medicine, University of Cincinnati Medical CenterCincinnati, Ohio, 45267USA
- Cancer CenterCollege of Medicine, University of Cincinnati Medical CenterCincinnati, Ohio, 45267USA
| | - Hung-Ming Lam
- Division of Environmental Genetics and Molecular Toxicology, Department of Environmental HealthCollege of Medicine, University of Cincinnati Medical CenterCincinnati, Ohio, 45267USA
| | - Shulin Wu
- Department of PathologyMassachusetts General Hospital and Harvard Medical SchoolBoston, Massachusetts, 02114USA
| | - Dan Song
- Division of Environmental Genetics and Molecular Toxicology, Department of Environmental HealthCollege of Medicine, University of Cincinnati Medical CenterCincinnati, Ohio, 45267USA
| | - Linda Levin
- Division of Epidemiology and Biostatistics, Department of Environmental HealthCollege of Medicine, University of Cincinnati Medical CenterCincinnati, Ohio, 45267USA
| | - Liang Cheng
- Department of Pathology and Laboratory MedicineIndiana UniversityIndianapolis, Indiana, 46202USA
| | - Chin-Lee Wu
- Department of PathologyMassachusetts General Hospital and Harvard Medical SchoolBoston, Massachusetts, 02114USA
| | - Shuk-Mei Ho
- Division of Environmental Genetics and Molecular Toxicology, Department of Environmental HealthCollege of Medicine, University of Cincinnati Medical CenterCincinnati, Ohio, 45267USA
- Center for Environmental GeneticsCollege of Medicine, University of Cincinnati Medical CenterCincinnati, Ohio, 45267USA
- Cancer CenterCollege of Medicine, University of Cincinnati Medical CenterCincinnati, Ohio, 45267USA
- (Correspondence should be addressed to S-M Ho at Division of Environmental Genetics and Molecular Toxicology, Department of Environmental Health, College of Medicine, University of Cincinnati Medical Center; )
| |
Collapse
|
56
|
Christie DR, Grant J, Darnell BE, Chapman VR, Gastaldelli A, Sites CK. Metabolic effects of soy supplementation in postmenopausal Caucasian and African American women: a randomized, placebo-controlled trial. Am J Obstet Gynecol 2010; 203:153.e1-9. [PMID: 20435291 DOI: 10.1016/j.ajog.2010.02.058] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2009] [Revised: 01/07/2010] [Accepted: 02/24/2010] [Indexed: 10/19/2022]
Abstract
OBJECTIVE We sought to determine the effect of daily soy supplementation on abdominal fat, glucose metabolism, and circulating inflammatory markers and adipokines in obese, postmenopausal Caucasian and African American women. STUDY DESIGN In a double-blinded controlled trial, 39 postmenopausal women were randomized to soy supplementation or to a casein placebo without isoflavones. In all, 33 completed the study and were analyzed. At baseline and at 3 months, glucose disposal and insulin secretion were measured using hyperglycemic clamps, body composition and body fat distribution were measured by computed tomographic scan and dual energy x-ray absorptiometry, and serum levels of C-reactive protein, interleukin-6, tumor necrosis factor-alpha, leptin, and adiponectin were measured by immunoassay. RESULTS Soy supplementation reduced total and subcutaneous abdominal fat and interleukin-6. No difference between groups was noted for glucose metabolism, C-reactive protein, tumor necrosis factor-alpha, leptin, or adiponectin. CONCLUSION Soy supplementation reduced abdominal fat in obese postmenopausal women. Caucasians primarily lost subcutaneous and total abdominal fat, and African Americans primarily lost total body fat.
Collapse
|
57
|
Adipose tissue and reproduction in women. Fertil Steril 2010; 94:795-825. [DOI: 10.1016/j.fertnstert.2009.03.079] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2009] [Revised: 03/20/2009] [Accepted: 03/24/2009] [Indexed: 12/20/2022]
|
58
|
Mancinelli R, Onori P, DeMorrow S, Francis H, Glaser S, Franchitto A, Carpino G, Alpini G, Gaudio E. Role of sex hormones in the modulation of cholangiocyte function. World J Gastrointest Pathophysiol 2010; 1:50-62. [PMID: 21607142 PMCID: PMC3097944 DOI: 10.4291/wjgp.v1.i2.50] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Revised: 05/28/2010] [Accepted: 06/04/2010] [Indexed: 02/06/2023] Open
Abstract
Over the last years, cholangiocytes, the cells that line the biliary tree, have been considered an important object of study for their biological properties which involves bile formation, proliferation, injury repair, fibrosis and angiogenesis. Cholangiocyte proliferation occurs in all pathologic conditions of liver injury where it is associated with inflammation and regeneration. During these processes, biliary cells start to secrete different cytokines, growth factors, neuropeptides and hormones which represent potential mechanisms for cross talk with other liver cells. Several studies suggest that hormones, and in particular, sex hormones, play a fundamental role in the modulation of the growth of this compartment in the injured liver which functionally conditions the progression of liver disease. Understanding the mechanisms of action and the intracellular pathways of these compounds on cholangiocyte pathophysiology will provide new potential strategies for the management of chronic liver diseases. The purpose of this review is to summarize the recent findings on the role of sex hormones in cholangiocyte proliferation and biology.
Collapse
|
59
|
Kim WY, Chung CS. Effects of Sex Steroid Hormones on Proliferation and Differentiation of Preadipocytes from Female and Male Pigs. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2010. [DOI: 10.5187/jast.2010.52.1.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
60
|
Roh SH, Kim CY, Won YS, Park CJ, Lee SS, Lee JG. Studies on Genetic Parameter Estimation and Sire Selection to Ultrasound Measurement Traits of Hanwoo. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2010. [DOI: 10.5187/jast.2010.52.1.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
61
|
Abstract
Aromatase is the enzyme that catalyzes the last step of estrogen biosynthesis. It is expressed in many tissues such as the gonads, brain and adipose tissue. The regulation of the level and activity of aromatase determines the levels of estrogens that have endocrine, paracrine and autocrine effects on tissues. Estrogens play many roles in the body, regulating reproduction, metabolism and behavior. In the brain, cell survival and the activity of neurons are affected by estrogens and hence aromatase.
Collapse
|
62
|
Chandanos E, Lagergren J. The mystery of male dominance in oesophageal cancer and the potential protective role of oestrogen. Eur J Cancer 2009; 45:3149-55. [PMID: 19804965 DOI: 10.1016/j.ejca.2009.09.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2009] [Revised: 08/24/2009] [Accepted: 09/02/2009] [Indexed: 12/19/2022]
Abstract
Oesophageal cancer is the sixth most common form of cancer death globally with almost 400,000 deaths annually. More than 90% of all cases are either adenocarcinomas (OAC) or squamous-cell carcinomas (OSCC). There is a strong male predominance with up to 8 and 3 men for every woman affected with OAC and OSCC, respectively. It has been hypothesised that sex hormonal factors may play a role in the development of oesophageal cancer or more specifically that oestrogen prevents such development. This article reviews the available literature on this topic. Basic science studies suggest an inhibitory effect of oestrogen in the growth of oesophageal cancer cells, and a possible mechanism of any oestrogen protection might be mediated through oestrogen receptors. But from the few epidemiological studies in which the hypothesis of oestrogen protection has been tested, no firm conclusions can yet be drawn of the role of oestrogen in human oesophageal cancer aetiology. More evidence from valid and large human studies is needed before any conclusions can be drawn.
Collapse
Affiliation(s)
- Evangelos Chandanos
- Upper Gastrointestinal Research, Department of Molecular Medicine and Surgery, Karolinska Institutet, SE-171 76 Stockholm, Sweden.
| | | |
Collapse
|
63
|
Peterlin BL, Katsnelson MJ, Calhoun AH. The associations between migraine, unipolar psychiatric comorbidities, and stress-related disorders and the role of estrogen. Curr Pain Headache Rep 2009; 13:404-12. [PMID: 19728969 PMCID: PMC3972495 DOI: 10.1007/s11916-009-0066-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Migraine is a common and often disabling neurovascular disorder. It has been linked with several psychiatric disorders, such as depression and anxiety, and to stress-related disorders, such as abuse and posttraumatic stress disorder (PTSD). Epidemiological data have consistently shown a higher prevalence of migraine, depression, anxiety, abuse, and PTSD in women as compared with men. The increased vulnerability of women to migraine and psychiatric disorders often occurs during periods of marked hormonal fluctuations of ovarian hormones. One consequence of these associations is the hypothesis that estrogens have a role in the pathophysiology of both disorders. This article offers an in-depth review of several studies linking psychiatric disorders and stress-related disorders with migraine. We also discuss the role of estrogen in the pathophysiologic overlap between these disorders. Finally, we briefly touch on where future research may be headed, in light of these data.
Collapse
Affiliation(s)
- B Lee Peterlin
- Drexel University College of Medicine, 245 North 15th Street, MS 423, Philadelphia, PA 19102, USA.
| | | | | |
Collapse
|
64
|
Paul M, Cholewa K, Mazurek U, Witek A, Wilczok T. Estrogen Receptor βΔ6 (ERβΔ6) Isoform in Human Endometrial Hyperplasia and Adenocarcinoma. Cancer Invest 2009; 22:211-8. [PMID: 15199603 DOI: 10.1081/cnv-120030209] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Estrogen receptor (ER)-beta may play a significant role in estrogen action in several human tissues. Estrogen receptor beta may act as a transdominant repressor of ER alpha transcriptional activity trough heterodimers form. Estrogen receptor messenger RNA (mRNA) variants also may be involved in various diseases, including endometrial cancer. The absence of estrogen receptors has often correlated with poor prognosis of endometrial tumors. The objective of the study was to determine the number of mRNA ER beta delta 6 and (wtER beta) in 1 microgram total RNA obtained from tissues of normal, hyperplastic endometrium, and endometrial adenocarcinoma. This study was designed to evaluate possible differences in the ER beta delta 6 and wtER beta messenger RNA (mRNA) level in the normal, hyperplastic endometrium, and endometrial endometrioid adenocarcinoma (G1, G2 morphological degree). Adenocarcinoma showed significantly lower ER beta delta 6 mRNA level than proliferative (p = 0.032) and secretory (p = 0.01) endometrium. A decrease of mRNA wtER beta in endometrial adenocarcinoma (p = 0.006) also was observed.
Collapse
Affiliation(s)
- Monika Paul
- Department of Molecular Biology, Biochemistry and Biopharmacy, Medical University of Silesia, Katowice, Poland.
| | | | | | | | | |
Collapse
|
65
|
Inhibitory effects of morinda officinalis extract on bone loss in ovariectomized rats. Molecules 2009; 14:2049-61. [PMID: 19513005 PMCID: PMC6254270 DOI: 10.3390/molecules14062049] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Revised: 06/02/2009] [Accepted: 06/05/2009] [Indexed: 11/17/2022] Open
Abstract
The present study was undertaken to investigate the protective effects of ethanol extract from the root of Morinda Officinalis (RMO) on ovariectomy-induced bone loss. Administration of RMO extract increased trabecular bone mineral content and bone mineral density of tibia, improved the levels of phosphorus (P), calcium (Ca) and OPG, decreased the levels of DPD/Cr, TRAP, ACTH and corticosterone, but did not reverse the levels of ALP, TNF-α and IL-6 in serum of ovariectomized rats. These findings demonstrated that RMO extract reduced bone loss in ovariectomized rats, probably via the inhibition of bone resorption, but was not involved with bone formation. Anthraquinones and polysaccharides from Morinda officinals could be responsible for their antiosteoporotic activity, and the action mechanism of these constituents needs to be further studied. Therefore, RMO has the potential to develop a clinically useful antiosteoporotic agent.
Collapse
|
66
|
Ben-Jonathan N, Hugo ER, Brandebourg TD. Effects of bisphenol A on adipokine release from human adipose tissue: Implications for the metabolic syndrome. Mol Cell Endocrinol 2009; 304:49-54. [PMID: 19433247 PMCID: PMC2775425 DOI: 10.1016/j.mce.2009.02.022] [Citation(s) in RCA: 180] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2009] [Accepted: 02/24/2009] [Indexed: 01/04/2023]
Abstract
Bisphenol A (BPA) is one of the most prevalent and best studied endocrine disruptors. After years of exposure to consumer products containing BPA, most individuals tested have circulating BPA at the low nanomolar levels. In addition to its well documented actions on the reproductive system, BPA exerts a wide variety of metabolic effects. This review summarizes recent findings on the ability of BPA, at environmentally relevant doses, to inhibit adiponectin and stimulate the release of inflammatory adipokines such as interleukin-6 (IL-6) and tumor necrosis factor alpha (TNFalpha) from human adipose tissue. Expression of several classical and non-classical estrogen receptors in human adipose tissue raises the possibility of their involvement as mediators of BPA actions. The implications of these observations to the obesity-related metabolic syndrome and its sequelae are discussed.
Collapse
Affiliation(s)
- Nira Ben-Jonathan
- Department of Cancer and Cell Biology, University of Cincinnati, OH 45267, USA.
| | | | | |
Collapse
|
67
|
Makimura H, Wei J, Dolan-Looby SE, Ricchiuti V, Grinspoon S. Retinol-binding protein levels are increased in association with gonadotropin levels in healthy women. Metabolism 2009; 58:479-87. [PMID: 19303967 PMCID: PMC2727279 DOI: 10.1016/j.metabol.2008.11.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2008] [Accepted: 11/13/2008] [Indexed: 10/21/2022]
Abstract
Recent studies have demonstrated an association between retinol-binding protein (RBP4) and insulin resistance. Retinol-binding protein is decreased in women and elevated in polycystic ovary syndrome. However, prior studies have not investigated the relationship between RBP4, gonadal steroids, and gonadotropins in healthy women. The aim of this study was to determine the RBP4 levels in a cohort of healthy women with a range of body mass indices and glucose tolerances to investigate the relationship between RBP4, gonadotropin levels, and menopausal status. Serum RBP4 levels were measured by enzyme-linked immunosorbent assay and quantitative Western blot in 88 healthy women (aged 24-59 years) from the general community in a cross-sectional study. Retinol-binding protein was higher in postmenopausal compared with premenopausal women (26.1 +/- 2.1 vs 19.3 +/- 0.5 mug/mL, P = .001). In univariate analysis, RBP4 was associated with follicle-stimulating hormone (r = 0.37, P = .0004), luteinizing hormone (r = 0.3, P = .005), and sex hormone-binding globulin (r = -0.24, P = .03) and trended to significance with estradiol (P = .09) but not with free testosterone or dehydroepiandrosterone sulfate. Retinol-binding protein was also associated with insulin at 2 hours during an oral glucose tolerance test (r = 0.24, P = .03) and the area under the curve for insulin during the oral glucose tolerance test (r = 0.26, P = .02). In multivariate regression modeling, both follicle-stimulating hormone (P = .03) and luteinizing hormone (P = .04) remained significantly associated with RBP4 after controlling for estradiol, sex hormone-binding globulin, insulin area under the curve, cholesterol, triglycerides, waist-to-hip ratio, and tumor necrosis factor alpha. Retinol-binding protein was not associated with inflammatory markers or with carotid intima-media thickness. Therefore, RBP4 is higher in postmenopausal women and is associated with gonadotropin concentrations in healthy women.
Collapse
Affiliation(s)
- Hideo Makimura
- Program in Nutritional Metabolism and Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | | | | | | | | |
Collapse
|
68
|
Bellemare V, Laberge P, Noël S, Tchernof A, Luu-The V. Differential estrogenic 17beta-hydroxysteroid dehydrogenase activity and type 12 17beta-hydroxysteroid dehydrogenase expression levels in preadipocytes and differentiated adipocytes. J Steroid Biochem Mol Biol 2009; 114:129-34. [PMID: 19429442 DOI: 10.1016/j.jsbmb.2009.01.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2008] [Revised: 12/31/2008] [Accepted: 01/12/2009] [Indexed: 12/26/2022]
Abstract
Estradiol (E2) is produced locally in adipose tissue and could play an important role in fat distribution and accumulation, especially in women. It is well recognized that aromatase is expressed in adipose tissue; however the identity of its estrogenic 17beta-hydroxysteroid dehydrogenase (17beta-HSD) partner is not identified. To gain a better knowledge about the enzyme responsible for the conversion of estrone into estradiol, we determined the activity and expression levels of known estrogenic 17beta-HSDs, namely types 1, 7 and 12 17beta-HSD in preadipocytes before and after differentiation into mature adipocytes using an adipogenic media. Estrogenic 17beta-HSD activity was assessed using [(14)C]-labelled estrone, while mRNA expression levels of types 1, 7 and 12 17beta-HSD were quantified using real-time PCR and protein expression levels of type 12 17beta-HSD was determined using immunoblot analysis. The data indicate that there is a low conversion of E1 into E2 in preadipocytes; however this activity is increased approximately 5-fold (p<0.0001) in differentiated adipocytes. The increased estrogenic 17beta-HSD activity is consistent with the increase in protein expression levels of 17beta-HSD12.
Collapse
Affiliation(s)
- Véronique Bellemare
- Molecular Endocrinology and Oncology Research Center, Laval University Medical Research Center and Laval University, Québec, Canada
| | | | | | | | | |
Collapse
|
69
|
Ohoka N, Kato S, Takahashi Y, Hayashi H, Sato R. The orphan nuclear receptor RORalpha restrains adipocyte differentiation through a reduction of C/EBPbeta activity and perilipin gene expression. Mol Endocrinol 2009; 23:759-71. [PMID: 19324970 DOI: 10.1210/me.2008-0277] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The nuclear receptor-type transcription factor retinoic acid receptor-related orphan receptor alpha (RORalpha) is a multifunctional molecule involved in tissue development and cellular function, such as inflammation, metabolism, and differentiation; however, the role of RORalpha during adipocyte differentiation has not yet been fully understood. Here we show that RORalpha inhibits the transcriptional activity of CCAAT/enhancer-binding protein beta (C/EBPbeta) without affecting its expression, thereby blocking the induction of both PPARgamma and C/EBPalpha, resulting in the suppression of C/EBPbeta-dependent adipogenesis. RORalpha interacted with C/EBPbeta so as to repress both the C/EBPbeta-p300 association and the C/EBPbeta-dependent recruitment of p300 to chromatin. In addition to the inhibitory effect on C/EBPbeta function, RORalpha also prevents the expression of the lipid droplet coating protein gene perilipin by peroxisome proliferators-activated receptor gamma (PPARgamma), acting through the specific mechanism of its promoter. We identified a suppressive ROR-responsive element overlapping the PPAR-responsive element in the perilipin promoter and verified that RORalpha competitively antagonizes the binding of PPARgamma. RORalpha inhibits PPARgamma-dependent adipogenesis along with the repression of perilipin induction. These findings suggest that RORalpha is a novel negative regulator of adipocyte differentiation that acts through dual mechanisms.
Collapse
Affiliation(s)
- Nobumichi Ohoka
- Department of Applied Biological Chemistry, The University of Tokyo, Japan
| | | | | | | | | |
Collapse
|
70
|
O'Sullivan AJ. Does oestrogen allow women to store fat more efficiently? A biological advantage for fertility and gestation. Obes Rev 2009; 10:168-77. [PMID: 19021869 DOI: 10.1111/j.1467-789x.2008.00539.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In normal healthy-weight humans, women have a higher percentage body fat than men, a difference that commences at puberty and continues throughout adult life, suggesting that the mechanism is related to sex steroids. The first half of pregnancy is also a stage of body fat gain in women. From an energy balance point, there is no explanation why women should be fatter than men, as the latter consume more calories proportionately. Moreover, women store fat in early pregnancy when caloric intake does not significantly change. The aim of this review is to focus on evidence supporting one mechanism that may account for these findings. That is, oestrogen reduces postprandial fatty acid oxidation leading to an increase in body fat which may account for the greater fat mass observed in women compared with men and the fat gain in early pregnancy. Therefore, female puberty and early pregnancy could be seen as states of efficient fat storage of energy in preparation for fertility, foetal development and lactation providing an obvious biological advantage. Further research into this mechanism of fat storage may provide further insights into the regulation of body fat.
Collapse
Affiliation(s)
- A J O'Sullivan
- Department of Medicine, St George Hospital, University of New South Wales, Sydney, Australia.
| |
Collapse
|
71
|
Park HJ, Della-Fera MA, Hausman DB, Rayalam S, Ambati S, Baile CA. Genistein inhibits differentiation of primary human adipocytes. J Nutr Biochem 2009; 20:140-8. [DOI: 10.1016/j.jnutbio.2008.01.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2007] [Revised: 12/14/2007] [Accepted: 01/03/2008] [Indexed: 11/29/2022]
|
72
|
Blouin K, Nadeau M, Mailloux J, Daris M, Lebel S, Luu-The V, Tchernof A. Pathways of adipose tissue androgen metabolism in women: depot differences and modulation by adipogenesis. Am J Physiol Endocrinol Metab 2009; 296:E244-55. [PMID: 18984855 DOI: 10.1152/ajpendo.00039.2008] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The objective was to examine pathways of androgen metabolism in abdominal adipose tissue in women. Abdominal subcutaneous (SC) and omental (OM) adipose tissue samples were surgically obtained in women. Total RNA was isolated from whole adipose tissue samples and from primary preadipocyte cultures before and after induction of differentiation. Expression levels of several steroid-converting enzyme transcripts were examined by real-time RT-PCR. Androgen conversion rates were also measured. We found higher expression levels in SC compared with OM adipose tissue for type 1 3beta-hydroxysteroid dehydrogenase (3beta-HSD-1; P < 0.05), for aldo-keto reductase 1C3 (AKR1C3; P < 0.0001), for AKR1C2 (P < 0.0001), and for the androgen receptor (P < 0.0001). 17beta-HSD-2 mRNA levels were lower in SC adipose tissue (P < 0.05). Induction of adipocyte differentiation led to significantly increased expression levels in SC cultures for AKR1C3 (4.7-fold, P < 0.01), 11-cis-retinol dehydrogenase (6.9-fold, P < 0.02), AKR1C2 (5.6-fold, P < 0.004), P-450 aromatase (5.7-fold, P < 0.02), steroid sulfatase (3.1-fold, P < 0.02), estrogen receptor-beta (11.8-fold, P < 0.01), and the androgen receptor (4.0-fold, P < 0.0005). Generally similar but nonsignificant trends were obtained in OM cultures. DHT inactivation rates increased with differentiation, this effect being mediated by dexamethasone alone, through a glucocorticoid receptor-dependent mechanism. In conclusion, higher mRNA levels of enzymes synthesizing and inactivating androgens are found in differentiated adipocytes, consistent with higher androgen-processing rates in these cells. Glucocorticoid-induced androgen inactivation may locally modulate the exposure of adipose cells to active androgens.
Collapse
Affiliation(s)
- Karine Blouin
- Molecular Endocrinology and Oncology Research Ctr., Laval University Medical Research Ctr., 2705 Laurier Blvd. (T3-67 Québec, QC, Canada G1V 4G2
| | | | | | | | | | | | | |
Collapse
|
73
|
Pedram A, Razandi M, Kim JK, O'Mahony F, Lee EY, Luderer U, Levin ER. Developmental phenotype of a membrane only estrogen receptor alpha (MOER) mouse. J Biol Chem 2008; 284:3488-95. [PMID: 19054762 DOI: 10.1074/jbc.m806249200] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Estrogen receptors (ERs) alpha and beta exist as nuclear, cytoplasmic, and membrane cellular pools in a wide variety of organs. The relative contributions of each ERalpha pool to in vivo phenotypes resulting from estrogen signaling have not been determined. To address this, we generated a transgenic mouse expressing only a functional E domain of ERalpha at the plasma membrane (MOER). Cells isolated from many organs showed membrane only localized E domain of ERalpha and no other receptor pools. Liver cells from MOER and wild type mice responded to 17-beta-estradiol (E2) with comparable activation of ERK and phosphatidylinositol 3-kinase, not seen in cells from ERalphaKO mice. Mating the MOER female mice with proven male wild type breeders produced no pregnancies because the uterus and vagina of the MOER female mice were extremely atrophic. Ovaries of MOER and homozygous Strasbourg ERalphaKO mice showed multiple hemorrhagic cysts and no corpus luteum, and the mammary gland development in both MOER and ERalphaKO mice was rudimentary. Despite elevated serum E2 levels, serum LH was not suppressed, and prolactin levels were low in MOER mice. MOER and Strasbourg female mice showed plentiful abdominal visceral and other depots of fat and increased body weight compared to wild type mice despite comparable food consumption. These results provide strong evidence that the normal development and adult functions of important organs in female mice requires nuclear ERalpha and is not rescued by membrane ERalpha domain expression alone.
Collapse
Affiliation(s)
- Ali Pedram
- Division of Endocrinology, Veterans Affairs Medical Center, Long Beach, California 90822, USA
| | | | | | | | | | | | | |
Collapse
|
74
|
Nott SL, Huang Y, Fluharty BR, Sokolov AM, Huang M, Cox C, Muyan M. Do Estrogen Receptor beta Polymorphisms Play A Role in the Pharmacogenetics of Estrogen Signaling? ACTA ACUST UNITED AC 2008; 6:239-259. [PMID: 19337586 DOI: 10.2174/187569208786733820] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Estrogen hormones play critical roles in the regulation of many tissue functions. The effects of estrogens are primarily mediated by the estrogen receptors (ER) alpha and beta. ERs are ligand-activated transcription factors that regulate a complex array of genomic events that orchestrate cellular growth, differentiation and death. Although many factors contribute to their etiology, estrogens are thought to be the primary agents for the development and/or progression of target tissue malignancies. Many of the current modalities for the treatment of estrogen target tissue malignancies are based on agents with diverse pharmacology that alter or prevent ER functions by acting as estrogen competitors. Although these compounds have been successfully used in clinical settings, the efficacy of treatment shows variability. An increasing body of evidence implicates ERalpha polymorphisms as one of the contributory factors for differential responses to estrogen competitors. This review aims to highlight the recent findings on polymorphisms of the lately identified ERbeta in order to provide a functional perspective with potential pharmacogenomic implications.
Collapse
Affiliation(s)
- Stephanie L Nott
- Department of Biochemistry & Biophysics, University of Rochester Medical School, Rochester, NY, 14642, USA
| | | | | | | | | | | | | |
Collapse
|
75
|
Hugo ER, Brandebourg TD, Woo JG, Loftus J, Alexander JW, Ben-Jonathan N. Bisphenol A at environmentally relevant doses inhibits adiponectin release from human adipose tissue explants and adipocytes. ENVIRONMENTAL HEALTH PERSPECTIVES 2008; 116:1642-7. [PMID: 19079714 PMCID: PMC2599757 DOI: 10.1289/ehp.11537] [Citation(s) in RCA: 329] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2008] [Accepted: 08/14/2008] [Indexed: 05/17/2023]
Abstract
BACKGROUND The incidence of obesity has risen dramatically over the last few decades. This epidemic may be affected by exposure to xenobiotic chemicals. Bisphenol A (BPA), an endocrine disruptor, is detectable at nanomolar levels in human serum worldwide. Adiponectin is an adipocyte-specific hormone that increases insulin sensitivity and reduces tissue inflammation. Thus, any factor that suppresses adiponectin release could lead to insulin resistance and increased susceptibility to obesity-associated diseases. OBJECTIVES In this study we aimed to compare a) the effects of low doses of BPA and estradiol (E(2)) on adiponectin secretion from human breast, subcutaneous, and visceral adipose explants and mature adipocytes, and b) expression of putative estrogen and estrogen-related receptors (ERRs) in these tissues. METHODS We determined adiponectin levels in conditioned media from adipose explants or adipocytes by enzyme-linked immunosorbant assay. We determined expression of estrogen receptors (ERs) alpha and beta, G-protein-coupled receptor 30 (GPR30), and ERRs alpha, beta, and gamma by quantitative real-time polymerase chain reaction. RESULTS BPA at 0.1 and 1 nM doses suppressed adiponectin release from all adipose depots examined. Despite substantial variability among patients, BPA was as effective, and often more effective, than equimolar concentrations of E(2). Adipose tissue expresses similar mRNA levels of ERalpha, ERbeta, and ERRgamma, and 20- to 30-fold lower levels of GPR30, ERRalpha, and ERRbeta. CONCLUSIONS BPA at environmentally relevant doses inhibits the release of a key adipokine that protects humans from metabolic syndrome. The mechanism by which BPA suppresses adiponectin and the receptors involved remains to be determined.
Collapse
Affiliation(s)
- Eric R. Hugo
- Department of Cell and Cancer Biology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Terry D. Brandebourg
- Department of Cell and Cancer Biology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jessica G. Woo
- Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | | | - J. Wesley Alexander
- Center for Surgical Weight Loss, University of Cincinnati, Cincinnati, Ohio, USA
| | - Nira Ben-Jonathan
- Department of Cell and Cancer Biology, University of Cincinnati, Cincinnati, Ohio, USA
- Address correspondence to N. Ben-Jonathan, Department of Cell and Cancer Biology, University of Cincinnati, 3125 Eden Ave., Cincinnati, OH 45267-0521 USA. Telephone: (513) 558-4821. Fax: (513) 558-4823. E-mail:
| |
Collapse
|
76
|
Yi KW, Shin JH, Seo HS, Lee JK, Oh MJ, Kim T, Saw HS, Kim SH, Hur JY. Role of estrogen receptor-alpha and -beta in regulating leptin expression in 3T3-L1 adipocytes. Obesity (Silver Spring) 2008; 16:2393-9. [PMID: 18719660 DOI: 10.1038/oby.2008.389] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We investigated the effects of the estrogen receptor-alpha (ERalpha) and -beta (ERbeta) in the regulation of leptin, resistin, and adiponectin expression in 3T3-L1 adipocytes. Mature adipocytes were exposed to estradiol (E2), ERalpha agonist (PPT (4,4',4''-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol)), ERbeta agonist (DPN (2,3-bis(4-Hydroxyphenyl)-propionitrile)), E2 with ERalpha antagonist (MPP (1,3-Bis(4-hydroxyphenyl)-4-methyl-5-[4-(2-piperidinylethoxy)phenol]-1H-pyrazole dihydrochloride)), and E2 with ERbeta antagonist (R,R-THC ((R,R)-5,11-diethyl-5,6,11,12-tetrahydro-2,8-chrysenediol)) at different concentrations. To clarify the expression and regulation of adipokines by ER subtypes, total RNA was extracted from cells and measured using quantitative PCR. Western blot analysis was performed to evaluate the protein expression of adipokines, ERalpha, and ERbeta. The leptin expression was significantly increased in the cells treated with high concentrations (10(-5) and 10(-6) mol/l) of the PPT (P < 0.01, P < 0.05). By contrast, the leptin expression decreased in a dose-dependent manner in the MPP-treated groups (P < 0.05). High concentrations (10(-5) mol/l) of R,R-THC with E2 (10(-7) mol/l) caused a significant increase of the leptin expression (P < 0.01). The leptin mRNA levels were positively correlated with the ERalpha mRNA levels (r = 0.584, P < 0.01) and negatively correlated with the ERbeta mRNA levels (r = -0.236, P = 0.03) in the adipocytes. The ratio of the ERalpha to ERbeta mRNA levels in the adipocytes was significantly associated with leptin mRNA levels (r = 0.454, P < 0.01). ERalpha induced leptin expression and ERbeta inhibited its expression in 3T3-L1 adipocytes. The ratio of the ERalpha-to-ERbeta expression in 3T3-L1 adipocytes may be an important potential regulatory factor in leptin expression.
Collapse
Affiliation(s)
- Kyong Wook Yi
- Department of Obstetrics and Gynecology, College of Medicine, Korea University, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Seidlová-Wuttke D, Prelle K, Fritzemeier KH, Wuttke W. Effects of estrogen receptor alpha- and beta-selective substances in the metaphysis of the tibia and on serum parameters of bone and fat tissue metabolism of ovariectomized rats. Bone 2008; 43:849-55. [PMID: 18708178 DOI: 10.1016/j.bone.2008.07.237] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Revised: 07/10/2008] [Accepted: 07/16/2008] [Indexed: 10/21/2022]
Abstract
The functions of estrogen receptors (ER) alpha and beta (ER-alpha and beta) in bone and fat tissue are not precisely described. Therefore we studied the effects of a specific ERalpha and ERbeta agonist in bone and fat of ovariectomized (ovx) rats and compared them with the effects of estradiol (E2). Animals were s.c. injected for 4-weeks with 3 doses of the ERalpha agonist 16alpha-LE2 or the ERbeta agonist 8beta-VE2 or with E2. The intermediate doses were antagonized by an additional daily treatment with ICI (1.53mg). Bone and fat parameters were evaluated by quantitative computer tomography (qCT). Estrogen regulated hormones were also measured. Uterine weights were stimulated; serum LH and leptin levels suppressed E2 and the ERalpha agonist. Density of the cancellous metaphyseal structures of the tibia was reduced in the controls which was prevented by E2 and the ERalpha agonist. Endosteal surface, endosteal, periosteal circumferences and fat depots were largest in the controls and the ERbeta treated animals and lowest in the E2 and the 16alpha-LE2 injected ovx rats. Osteocalcin and the CrossLaps were highest in the ovx controls and reduced by E2 and the ERalpha agonist. Serum osteocalcin was stimulated by the ERbeta agonist. The strain strength index (SSI) in relation to the bodyweight - an indicator of bone elasticity - was lowest in controls and increased dose dependently in the E2 and in the ERalpha treated animals. Most effects in the uterus, serum and bone were antagonized by ICI. Most effects in the bone and fat were exerted by mechanisms involving the ERalpha but the ERbeta agonist appears to stimulate osteoblasts.
Collapse
Affiliation(s)
- D Seidlová-Wuttke
- Department of Clinical and Experimental Endocrinology, University of Goettingen, Germany
| | | | | | | |
Collapse
|
78
|
Cederroth CR, Vinciguerra M, Gjinovci A, Kühne F, Klein M, Cederroth M, Caille D, Suter M, Neumann D, James RW, Doerge DR, Wallimann T, Meda P, Foti M, Rohner-Jeanrenaud F, Vassalli JD, Nef S. Dietary phytoestrogens activate AMP-activated protein kinase with improvement in lipid and glucose metabolism. Diabetes 2008; 57:1176-85. [PMID: 18420492 DOI: 10.2337/db07-0630] [Citation(s) in RCA: 142] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Emerging evidence suggests that dietary phytoestrogens can have beneficial effects on obesity and diabetes, although their mode of action is not known. Here, we investigate the mechanisms mediating the action of dietary phytoestrogens on lipid and glucose metabolism in rodents. RESEARCH DESIGN AND METHODS Male CD-1 mice were fed from conception to adulthood with either a high soy-containing diet or a soy-free diet. Serum levels of circulating isoflavones, ghrelin, leptin, free fatty acids, triglycerides, and cholesterol were quantified. Tissue samples were analyzed by quantitative RT-PCR and Western blotting to investigate changes of gene expression and phosphorylation state of key metabolic proteins. Glucose and insulin tolerance tests and euglycemic-hyperinsulinemic clamp were used to assess changes in insulin sensitivity and glucose uptake. In addition, insulin secretion was determined by in situ pancreas perfusion. RESULTS In peripheral tissues of soy-fed mice, especially in white adipose tissue, phosphorylation of AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase was increased, and expression of genes implicated in peroxisomal fatty acid oxidation and mitochondrial biogenesis was upregulated. Soy-fed mice also showed reduced serum insulin levels and pancreatic insulin content and improved insulin sensitivity due to increased glucose uptake into skeletal muscle. Thus, mice fed with a soy-rich diet have improved adipose and glucose metabolism. CONCLUSIONS Dietary soy could prove useful to prevent obesity and associated disorders. Activation of the AMPK pathway by dietary soy is likely involved and may mediate the beneficial effects of dietary soy in peripheral tissues.
Collapse
Affiliation(s)
- Christopher R Cederroth
- Department of Genetic Medicine and Development, National Centre of Competence in Research-Frontiers in Genetics, University of Geneva, 1211 Geneva 4, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Hernández-Morante JJ, Pérez-de-Heredia F, Luján JA, Zamora S, Garaulet M. Role of DHEA-S on body fat distribution: gender- and depot-specific stimulation of adipose tissue lipolysis. Steroids 2008; 73:209-15. [PMID: 18063002 DOI: 10.1016/j.steroids.2007.10.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2007] [Revised: 10/08/2007] [Accepted: 10/08/2007] [Indexed: 11/20/2022]
Abstract
The objective of this work was to study the possible impact of DHEA-S on body fat distribution and the specific action of the hormone on lipolysis from visceral and subcutaneous human adipose tissue. First, a clinical evaluation was performed in 84 obese patients (29 men, 55 women), measuring serum DHEA-S, computed tomography (CT) anthropometric parameters of abdominal fat distribution. In a second experiment, subcutaneous and visceral adipose tissue samples were obtained from 20 obese patients (10 men, 10 women) and cultured in vitro under stimulation with DHEA-S to further assess a possible effect of this hormone on adipose tissue lipolysis. Serum DHEA-S was inversely and specifically associated with visceral fat area (VA) as assessed by CT in men and with waist-to-hip ratio in women. In vitro, DHEA-S increased lipolysis in women's subcutaneous adipose tissue at 2 h, while in men, the effect was evident in visceral tissue and after 24 h of treatment. In conclusion, DHEA-S contributes to gender-related differences in body fat distribution probably by a differential lipolytic action. We have demonstrated for the first time in vitro that DHEA-S stimulates lipolysis preferably in subcutaneous fat in women and in visceral fat in men.
Collapse
Affiliation(s)
- Juan J Hernández-Morante
- Department of Physiology, University of Murcia, and Surgery Service, University Hospital Virgen de la Arrixaca, Spain
| | | | | | | | | |
Collapse
|
80
|
Alonso A, Moreno M, Ordóñez P, Fernández R, Pérez C, Díaz F, Navarro A, Tolivia J, González C. Chronic estradiol treatment improves brain homeostasis during aging in female rats. Endocrinology 2008; 149:57-72. [PMID: 17901235 DOI: 10.1210/en.2007-0627] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Aging is associated with a reduction in metabolic function, insulin resistance, increased incidence of neurodegenerative diseases, and memory or cognitive dysfunction. In aging females, loss of gonadal function determines the beginning of the period of reduced metabolic function. Estrogens have neuroprotective effects, but the mechanisms by which they exert these effects remain unclear. The effects of estradiol treatment on the activation of the insulin receptor substrate (IRS)-1 signaling pathway, the interactions between estrogen receptor (ER)-alpha and IRS-1 and the p85alpha subunit of phosphatidylinositol-3 kinase, together with the possible effects of estradiol treatment on glucose transporter-3 and -4 levels, were investigated in female rats. The level of expression of each glucose transporter was greater in control and estradiol-treated groups than in the ovariectomized group. Interactions of ERalpha46-IRS-1, ERalpha46-p85alpha, and p85alpha-IRS-1, as well as IRS-1 phosphorylation, appeared to increase with estradiol treatment. The results indicate that estradiol treatment improves some aspects of neuronal homeostasis that are affected by aging; this may indicate that estradiol has neuroprotective effects in female rats. Additional animal studies are required to clarify the neuroprotective role of estradiol in relation to other important molecules involved in the IRS-1-phosphatidylinositol-3 kinase signaling pathway.
Collapse
Affiliation(s)
- Ana Alonso
- Department of Functional Biology, University of Oviedo, Oviedo, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Rodriguez-Cuenca S, Monjo M, Frontera M, Gianotti M, Proenza AM, Roca P. Sex steroid receptor expression profile in brown adipose tissue. Effects of hormonal status. Cell Physiol Biochem 2007; 20:877-86. [PMID: 17982270 DOI: 10.1159/000110448] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2007] [Indexed: 11/19/2022] Open
Abstract
Recent investigations suggest that sex hormones play an important role in the brown adipose tissue (BAT) thermogenic program by acting on several steps of the lipolytic signal cascade and on the UCP1 transcription control. However, the number of studies focusing on steroid receptor status in brown adipose tissue is negligible. In the present study, we analyze steroid receptor mRNA levels in brown adipose tissue in male and female rats and in pregnant and lactating females, all of them models with a different hormonal background. The direct effect of sex hormones on the expression of their receptors was studied in vitro in primary culture of brown adipocytes. Oestrogen receptor (ERalpha) and androgen receptor (AR) densities were higher in male than in female BAT. PR A+B mRNA expression was downregulated in lactation, suggesting a role of progesterone signalling in thermogenesis impairment at this stage. In vitro studies showed that progesterone decreased PR A+B mRNA and that testosterone downregulated ERalpha mRNA. The results highlighted in this study demonstrate the presence of steroid receptor mRNA in BAT and in brown cell cultured adipocytes, supporting the idea that changes in steroid receptor expression would be important for the understanding of sex hormone effects on BAT physiology.
Collapse
Affiliation(s)
- Sergio Rodriguez-Cuenca
- Grup de Metabolisme Energetic i Nutrició, Departament de Biologia Fonamental i Ciències de la Salut, Institut Universitari d' Investigació en Ciencies de la Salut, Universitat de les Illes Balears, Palma de Mallorca, Spain
| | | | | | | | | | | |
Collapse
|
82
|
Nilsson M, Dahlman I, Jiao H, Gustafsson JA, Arner P, Dahlman-Wright K. Impact of estrogen receptor gene polymorphisms and mRNA levels on obesity and lipolysis--a cohort study. BMC MEDICAL GENETICS 2007; 8:73. [PMID: 18053221 PMCID: PMC2238734 DOI: 10.1186/1471-2350-8-73] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2007] [Accepted: 12/04/2007] [Indexed: 01/18/2023]
Abstract
Background The estrogen receptors α and β (ESR1, ESR2) have been implicated in adiposity, lipid metabolism and feeding behaviour. In this report we analyse ESR1 and ESR2 gene single nucleotide polymorphisms (SNPs) for association with obesity. We also relate adipose tissue ESR1 mRNA levels and ESR1 SNPs to adipocyte lipolysis and lipogenesis phenotypes. Methods 23 ESR1 and 11 ESR2 tag-SNPs, covering most of the common haplotype variation in each gene according to HAPMAP data, were analysed by Chi2 for association with obesity in a cohort comprising 705 adults with severe obesity and 402 lean individuals. Results were replicated in a cohort comprising 837 obese and 613 lean subjects. About 80% of both cohorts comprised women and 20% men. Adipose tissue ESR1 mRNA was quantified in 122 women and related to lipolysis and lipogenesis by multiple regression. ESR1 SNPs were analysed for association with adipocyte lipolysis and lipogenesis phenotypes in 204 obese women by simple regression. Results No ESR1 SNP was associated with obesity. Five ESR2 SNPs displayed nominal significant allelic association with obesity in women and one in men. The two ESR2 SNPs associated with obesity with nominal P value < 0.01 were genotyped in a second cohort where no association with obesity was observed. There was an inverse correlation between ESR1 mRNA levels in abdominal subcutaneous (sc) adipose tissue and basal lipolysis, as well as responsiveness to adrenoceptor agonists independent of age and BMI (P value 0.009–0.045). ESR1 rs532010 was associated with lipolytic sensitivity to noradrenaline (nominal P value 0.012), and ESR1 rs1884051 with responsiveness to the non-selective beta-adrenoceptor agonist isoprenaline (nominal P value 0.05). These associations became non-significant after Bonferroni correction. Conclusion ESR1 gene alleles are unlikely to be a major cause of obesity in women. A minor importance of ESR2 on severe obesity cannot be excluded. The inverse correlation between ESR1 mRNA levels and lipolytic responsiveness to adrenoceptor agonists implies that low adipose tissue ESR1 levels attenuate catecholamine resistance in sc fat cells of obese women hereby contributing to loss of sc and gain of visceral fat. There is no evidence for a genetic impact of ESR1 on lipolysis or lipogenesis.
Collapse
Affiliation(s)
- Maria Nilsson
- Departments of Biosciences and Nutrition, Karolinska Institutet, S-141 57 Huddinge, Sweden.
| | | | | | | | | | | |
Collapse
|
83
|
Badeau M, Vihma V, Mikkola TS, Tiitinen A, Tikkanen MJ. Estradiol fatty acid esters in adipose tissue and serum of pregnant and pre- and postmenopausal women. J Clin Endocrinol Metab 2007; 92:4327-31. [PMID: 17726068 DOI: 10.1210/jc.2007-1372] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
CONTEXT The 17beta-estradiol fatty acid esters are hormone derivatives with long-lasting estrogenic effect. They are transported in serum lipoproteins and thought to be sequestered in adipose tissue. OBJECTIVE Our objective was to determine the 17beta-estradiol fatty acid ester concentrations in serum and adipose tissue in women of various hormonal states. DESIGN After several chromatographic steps separating esterified from free estradiol, time-resolved fluoroimmunoassay was used as a quantifying tool. PARTICIPANTS Samples were obtained from pregnant women undergoing cesarean section (n = 13), or premenopausal (n = 8) and postmenopausal women (n = 6) during gynecological surgery. MAIN OUTCOME MEASURES 17beta-Estradiol and 17beta-estradiol fatty acid ester concentrations in serum, and visceral and sc adipose tissue were examined. RESULTS The ratio of esterified to free estradiol in plasma increased with decreasing estradiol level from 0.5% in pregnant, to 15% in premenopausal and 110% in postmenopausal women. Estradiol esters constituted about 10% of the free estradiol present in adipose tissue in pregnancy. In nonpregnant women, most of the adipose tissue estradiol was in esterified form, the median ester to free ratio being elevated to 150-490%. After menopause, the overwhelming majority of estradiol in both free and esterified form was present in adipose tissue. CONCLUSIONS The overall higher ester to free estradiol ratio in adipose tissue than in serum indicates active esterification capacity in adipose tissue. The predominance of esterified and free estradiol in postmenopausal adipose tissue compared with serum suggests in situ production and storage. Whether the estradiol esters have an independent physiological role in adipose tissue remains to be clarified.
Collapse
Affiliation(s)
- Maija Badeau
- Department of Medicine, Helsinki University Central Hospital, Haartmaninkatu 4, 00290 Helsinki, Finland
| | | | | | | | | |
Collapse
|
84
|
Cederroth CR, Vinciguerra M, Kühne F, Madani R, Doerge DR, Visser TJ, Foti M, Rohner-Jeanrenaud F, Vassalli JD, Nef S. A phytoestrogen-rich diet increases energy expenditure and decreases adiposity in mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2007; 115:1467-73. [PMID: 17938737 PMCID: PMC2022650 DOI: 10.1289/ehp.10413] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Accepted: 07/12/2007] [Indexed: 05/05/2023]
Abstract
BACKGROUND Obesity is an increasingly prevalent health problem, and natural effective therapeutic approaches are required to prevent its occurrence. Phytoestrogens are plant-derived compounds with estrogenic activities; they can bind to both estrogen receptors alpha and beta and mimic the action of estrogens on target organs. OBJECTIVES The purpose of this study was to examine the influence of soy-derived phytoestrogens on energy balance and metabolism. METHODS Male outbred mice (CD-1) were allowed ad libitum access to either a high soy-containing diet or a soy-free diet from conception to adulthood. We measured circulating serum isoflavone levels using reverse-phase solid-phase extraction for subsequent liquid chromatography electrospray tandem mass spectrometry analysis. Adult animals were analyzed for body composition by dual-energy X-ray absorptiometry, locomotor activity by running-wheel experiments, respiratory exchange rate by indirect calorimetry, and food intake using metabolic cages. Quantitative reverse transcriptase-polymerase chain reaction was performed to determine the expression of hypothalamic neuropeptide genes. RESULTS We found that adult mice fed a soy-rich diet had reduced body weight, adiposity, and resistance to cold. This lean phenotype was associated with an increase in lipid oxidation due to a preferential use of lipids as fuel source and an increase in locomotor activity. The modulation of energy balance was associated with a central effect of phytoestrogens on the expression of hypothalamic neuropeptides, including agouti-related protein. CONCLUSION The data suggest that dietary soy could have beneficial effects on obesity, but they also emphasize the importance of monitoring the phytoestrogen content of diets as a parameter of variability in animal experiments.
Collapse
Affiliation(s)
- Christopher R. Cederroth
- Department of Genetic Medicine and Development and National Center for Competence in Research - Frontiers in Genetics, University of Geneva, Geneva, Switzerland
| | - Manlio Vinciguerra
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Françoise Kühne
- Department of Genetic Medicine and Development and National Center for Competence in Research - Frontiers in Genetics, University of Geneva, Geneva, Switzerland
| | - Rime Madani
- Department of Genetic Medicine and Development and National Center for Competence in Research - Frontiers in Genetics, University of Geneva, Geneva, Switzerland
| | - Daniel R. Doerge
- National Center for Toxicological Research, Jefferson, Arkansas, USA
| | - Theo J. Visser
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Michelangelo Foti
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Françoise Rohner-Jeanrenaud
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jean-Dominique Vassalli
- Department of Genetic Medicine and Development and National Center for Competence in Research - Frontiers in Genetics, University of Geneva, Geneva, Switzerland
| | - Serge Nef
- Department of Genetic Medicine and Development and National Center for Competence in Research - Frontiers in Genetics, University of Geneva, Geneva, Switzerland
| |
Collapse
|
85
|
Andersen O, Pedersen SB, Svenstrup B, Hansen BR, Paulsen SK, Rathje GS, Richelsen B, Nielsen JO, Madsbad S, Iversen J, Haugaard SB. Circulating sex hormones and gene expression of subcutaneous adipose tissue oestrogen and alpha-adrenergic receptors in HIV-lipodystrophy: implications for fat distribution. Clin Endocrinol (Oxf) 2007; 67:250-8. [PMID: 17524033 DOI: 10.1111/j.1365-2265.2007.02872.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Circulating oestradiol and testosterone, which have been shown to increase in human immunodeficiency virus (HIV)-infected patients following highly active antiretroviral therapy (HAART), may influence fat distribution and insulin sensitivity. Oestradiol increases subcutaneous adipose tissue in humans possibly through binding to oestrogen-receptor-alpha, which in turn activates anti-lipolytic alpha2A-adrenergic-receptor. DESIGN AND METHODS To address these issues circulating pituitary-gonadal-axis hormones and gene expression of receptors in subcutaneous adipose tissue were determined in 31 nondiabetic HIV-infected male patients receiving HAART (16 with lipodystrophy), in whom measures of fat distribution (CT and DEXA-scans) and insulin sensitivity (hyperinsulinaemic euglycaemic clamp) were available. RESULTS Total and free oestradiol and testosterone were decreased in lipodystrophic patients compared to nonlipodystrophic patients, whereas luteinizing hormone, follicle-stimulating hormone and prolactin were similar and normal in both study groups. Ratio of subcutaneous to total abdominal fat mass, limb fat, and insulin sensitivity, which were all decreased in lipodystrophic patients, correlated positively with both plasma oestradiol and testosterone (n = 31). Glycerol concentration during clamp (a marker of lipolysis) correlated inversely with expression of alpha2A-adrenergic-receptor, ratio of subcutaneous to total abdominal fat mass, and limb fat, respectively. Expression of alpha2A-adrenergic-receptor correlated positively with expression of oestrogen-receptor-alpha. CONCLUSIONS The results fit the hypothesis that sex hormones play a role in altered fat distribution and insulin sensitivity of male patients with HIV-lipodystrophy. The effect of oestradiol on the subcutaneous fat depot and lipolysis may be mediated in part through binding to the oestrogen-receptor-alpha, in turn activating anti-lipolytic alpha2A-adrenergic-receptor.
Collapse
Affiliation(s)
- Ove Andersen
- Department of Infectious Diseases, Hvidovre University Hospital, DK 2650 Hvidovre, Denmark.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Shin JH, Hur JY, Seo HS, Jeong YA, Lee JK, Oh MJ, Kim T, Saw HS, Kim SH. The ratio of estrogen receptor alpha to estrogen receptor beta in adipose tissue is associated with leptin production and obesity. Steroids 2007; 72:592-9. [PMID: 17509633 DOI: 10.1016/j.steroids.2007.03.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2007] [Revised: 03/22/2007] [Accepted: 03/26/2007] [Indexed: 11/16/2022]
Abstract
The loss of estrogen associated with menopause is suspected to play an important regulatory role in changes of fat metabolism and obesity. To evaluate the relationship between obesity and the ratio of estrogen receptor subtypes (ERalpha/ERbeta) in adipose tissues in pre- and postmenopausal women, we measured the anthropometric indices of 31 premenopausal women and 12 postmenopausal women. Serum samples, subcutaneous and omental adipose tissues were also obtained from study participants. Serum leptin, adiponectin, IL-6, and TNF-alpha levels were measured using ELISA methods. Real-time RT-PCR analysis was performed to detect and to compare mRNA levels of leptin and estrogen receptor subtypes (ERalpha and ERbeta) from adipose tissues. The ratio of abdominal subcutaneous to omental adipose tissue for the ER subtypes (Sc-Om ratio of the ER subtypes), i.e., subcutaneous ERalpha/ERbeta over omental ERalpha/ERbeta, showed significant correlations with anthropometric indices including BMI (r=0.801, p<0.05) and waist circumference (r=0.696, p<0.05) in both pre- and postmenopausal women. The Sc-Om ratio of the ER subtypes also had a significant correlation with the serum leptin level (r=0.735, p<0.05) as well as the mRNA level of leptin in omental adipose tissue (r=0.753, p<0.05). However, there were no significant differences between the pre- and postmenopausal groups with regard to the expressed level of ER subtypes. In conclusion, our study results showed that the ratio of ERalpha to ERbeta in adipose tissue was associated with obesity as well as the serum level and production of leptin in omental adipose tissue.
Collapse
Affiliation(s)
- Jung-Ho Shin
- Department of Obstetrics and Gynecology, College of Medicine, Korea University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Niemelä SM, Miettinen S, Konttinen Y, Waris T, Kellomäki M, Ashammakhi NA, Ylikomi T. Fat tissue: views on reconstruction and exploitation. J Craniofac Surg 2007; 18:325-35. [PMID: 17414282 DOI: 10.1097/scs.0b013e3180333b6a] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Transplantation of autologous fat as pedicle or transposition flaps has been a classical method in plastic surgery for tissue reconstruction. The injection of fat for soft tissue reconstruction is also an old innovation. This approach has some significant drawbacks such as resorption of the fat transplant. To regenerate additional and self-regenerating adipose tissue for reconstructive purposes, a thorough understanding of adipose tissue (mesodermal stem cells, adipoblasts, pre-adipocytes, mature, lipid-synthesizing, and lipid-storing white or brown adipocytes) on cellular and molecular levels is required. Several transcription factors that play a central role in the control of adipogenesis have been identified. Among these are the CCAAT/enhancer binding protein gene family and peroxisome proliferator-activated receptor-gamma. Hormones and growth factors, such as insulin and insulin-like growth factor (IGF), transfer external signals to differentiating adipocytes. In an attempt to improve the quality of tissue-engineered fat by culture-expanded adipocytes, various pre-adipocyte and stem cell culture conditions and expansion methods have been developed. In the presence of fetal calf serum, spontaneous differentiation of pre-adipocytes into fat cell clusters occurs to some degree. This in vitro differentiation can be enhanced by addition of inducing agents such as dexamethasone, isobutylmethylxantine, and insulin into the culture medium. Recent work has shown the multipotency of pre-adipocytes, which are fibroblast-like precursors of adipocytes. With use of specific culture conditions, human adipose tissue-derived stem cells can be induced to express markers of adipocyte, osteoblast, and myocyte cell lineages. The multipotent characteristics of adipose tissue-derived stem cells, as well as their abundance and accessibility in the human body, make them a potential cell source for tissue engineering applications.
Collapse
Affiliation(s)
- Sanna-Mari Niemelä
- Department of Cell Biology, Medical School, University of Tampere, Tampere, and Department of Surgery, Oulu University Hospital, Oulu, Finland
| | | | | | | | | | | | | |
Collapse
|
88
|
Kadowaki K, Fukino K, Negishi E, Ueno K. Sex differences in PPARgamma expressions in rat adipose tissues. Biol Pharm Bull 2007; 30:818-20. [PMID: 17409529 DOI: 10.1248/bpb.30.818] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Clinical studies had suggested that there were sex differences in pharmacological and side effects of pioglitazone. However, there are few studies on the sex differences of peroxisome proliferator-activated receptor (PPAR) gamma expressions in rat adipose tissues. We investigated the sex differences in peroxisome proliferator-activated receptor (PPAR) gamma expressions in rat adipose tissues. Subcutaneous abdominal adipose tissues and perigonadal adipose tissues were obtained from male and female Wistar rats (12 weeks of age). Expressions of PPARgamma protein were determined by Western blot analysis with anti-PPARgamma antibody. Vaginal smear check was performed in female rats. We obtained adipose tissues from females, according to the different phases of the estrous cycle. Both PPARgamma1 and gamma2 subtypes were expressed in subcutaneous adipose tissues. Expressions of PPARgamma2 in subcutaneous adipose tissues were significantly higher in males than in females. On the other hand, expressions of PPARgamma2 in perigonadal adipose tissues were significantly higher in females than in males. Expressions of PPARgamma2 in perigonadal adipose tissues in females significantly decreased during diestrus. It can be suggested that the sex hormones might affect the expressions of PPARgamma2 in adipose tissues.
Collapse
Affiliation(s)
- Kyoko Kadowaki
- Department of Geriatric Pharmacology & Therapeutics, Graduate School of Pharmaceutical Sciences, Chiba University, Japan
| | | | | | | |
Collapse
|
89
|
Jaubert AM, Mehebik-Mojaat N, Lacasa D, Sabourault D, Giudicelli Y, Ribière C. Nongenomic estrogen effects on nitric oxide synthase activity in rat adipocytes. Endocrinology 2007; 148:2444-52. [PMID: 17303666 DOI: 10.1210/en.2006-1329] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Estrogens exert multiple genomic effects on adipose tissue through binding to nuclear estrogen receptors. However, there is evidence for additional nongenomic mechanisms whereby estrogens may exert their control on adipose tissue metabolism through rapid activation of various membrane-initiated kinase cascades. Here, we tested rapid effects of estrogens on nitric oxide production in white adipose tissue using 17-beta estradiol (E2) and its membrane impermeant albumin conjugated form (17-beta estradiol hemisuccinate BSA, E2-BSA). We found that both E2 and E2-BSA stimulate nitric oxide synthase (NOS) activity in adipocytes. These effects were abolished by 1) ICI 182-780, a selective estrogen receptor antagonist; 2) wortmannin, an inhibitor of phosphatidylinositol 3-kinase; and 3) N-[2-(p-bromocinnamylamino) ethyl]-5-isoquinolinesulfonamide (H-89) an inhibitor of protein kinase A. In contrast to NOS activation by E2, E2-BSA-induced NOS activity was abolished by UO126, an inhibitor of MAPK kinase/ERK (p42/p44 MAPKs). Immunoblotting studies have shown that both estrogens phosphorylate endothelial NOS (NOS III) on Ser(1179), an effect that is prevented by wortmannin and H89, suggesting that NOS III is the target for estrogen-induced NOS activity. Furthermore, only the E2-BSA-induced NOS III phosphorylation on Ser(1179) was totally abolished by UO126. These results indicate that the signaling cascades involved in adipocyte NOS stimulation by estrogens are different depending on whether estrogens are free or conjugated to albumin and therefore underline the importance of estrogen receptor locations in the nongenomic actions of estrogens in these cells.
Collapse
Affiliation(s)
- Anne-Marie Jaubert
- Départment de Biochimie et de Biologie Moléculaire, Université de Versailles Saint-Quentine en Yuelines, Versailles, France
| | | | | | | | | | | |
Collapse
|
90
|
Abstract
1. The aim of the present review is to outline: (i) the association between sleep and metabolism; (ii) how sleep duration influences the development of disease; and (iii) how sex differences, ageing and obesity may potentially influence the relationship between sleep, metabolic control and subsequent disease. 2. Sleep is associated with a number of endocrine changes, including a change in insulin action in healthy young individuals. Sleep duration shows a prospective U-shaped relationship with all-cause mortality, cardiovascular disease and Type 2 diabetes. 3. Chronic sleep restriction is becoming more common. Experimental sleep restriction impedes daytime glucose control and increases appetite. 4. The sex hormones oestrogen and testosterone influence sleep duration and quality and may account for sex differences in the prevalence of sleep-related disorders. 5. Ageing is associated with a decreased sleep duration, decreased muscle mass and impaired insulin action. 6. Obesity impairs insulin action and is associated with the incidence and severity of obstructive sleep apnoea. 7. Sleep plays an integral role in metabolic control. Consequently, insufficient sleep may represent a modifiable risk factor for the development of Type 2 diabetes. The challenge ahead is to identify how sex differences, ageing and obesity could potentially influence the relationship between sleep and metabolism.
Collapse
Affiliation(s)
- Michael I Trenell
- Sleep and Circadian Research Group, Woolcock Institute of Medical Research, Sydney, NSW, Australia
| | | | | |
Collapse
|
91
|
Wake DJ, Strand M, Rask E, Westerbacka J, Livingstone DEW, Soderberg S, Andrew R, Yki-Jarvinen H, Olsson T, Walker BR. Intra-adipose sex steroid metabolism and body fat distribution in idiopathic human obesity. Clin Endocrinol (Oxf) 2007; 66:440-6. [PMID: 17302881 DOI: 10.1111/j.1365-2265.2007.02755.x] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Causes of visceral fat accumulation include glucocorticoid excess or decreased oestrogen/androgen ratio either in plasma or within adipose tissue. In obese subjects, the intra-adipose cortisol-generating enzyme 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) is increased, but information on sex steroid signalling is sparse. We aimed to test associations between body fat or fat distribution and mRNA transcript levels for androgen and oestrogen receptors and for enzymes metabolizing sex steroids in adipose tissue. DESIGN A cross-sectional study. PATIENTS Forty-five healthy men and women with body mass index (BMI) 21-36 kg/m(2). MEASUREMENTS In subcutaneous adipose biopsies we measured mRNAs for enzymes metabolizing local oestrogens (aromatase) and androgens [5alpha-reductase type 1; AKR1C2 (3alpha-HSD3); AKR1C3 (17beta-HSD5, 3alpha-HSD2)] and for sex steroid receptors [oestrogen receptor (ER)-alpha and androgen receptor (AR)]. We related these to body fat mass and distribution. RESULTS Generalized obesity (BMI) was associated with increased aromatase mRNA (r = 0.35, P < 0.05). Central obesity (waist : hip ratio) was associated with mRNA for AKR1C2 (r = 0.28, P < 0.05) and AKR1C3 (r = 0.38, P < 0.01) but not aromatase (r = 0.06). 5alpha-Reductase type 1, ER and AR mRNA levels did not predict fat amount or its distribution. CONCLUSION These data on transcript levels suggest that, in idiopathic obesity, increased intra-adipose oestrogen generation by aromatase predicts peripheral fat distribution, while androgen metabolism by AKR1C isoforms predicts central fat distribution, supporting the hypothesis that intra-adipose sex steroid metabolism is a determinant of gynoid vs. android patterns of body fat.
Collapse
Affiliation(s)
- Deborah J Wake
- Endocrinology Unit, Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Scotland, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Smidt K, Pedersen SB, Brock B, Schmitz O, Fisker S, Bendix J, Wogensen L, Rungby J. Zinc-transporter genes in human visceral and subcutaneous adipocytes: lean versus obese. Mol Cell Endocrinol 2007; 264:68-73. [PMID: 17118530 DOI: 10.1016/j.mce.2006.10.010] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2006] [Revised: 09/26/2006] [Accepted: 10/08/2006] [Indexed: 11/26/2022]
Abstract
Zinc ions influence adipose tissue metabolism by regulating leptin secretion and by promoting free fatty acid release and glucose uptake. The mechanisms controlling zinc metabolism in adipose tissue are unknown. We therefore examined the gene-expression levels of a number of zinc-transporting proteins in adipose tissue, comparing subcutaneous fat with visceral fat from lean and obese humans. Both ZnT-proteins responsible for zinc transport from cytosol to extracellular compartments and intracellular vesicles and Zip-proteins responsible for zinc transport to the cytoplasm were expressed in all samples. This suggests that zinc metabolism in adipocytes is actively controlled by zinc-transporters. The expression levels were different in lean and obese subjects suggesting a role for these proteins in obesity. Furthermore, the expression levels were different from subcutaneous fat to intra-abdominal fat suggesting that the metabolic activity in adipocytes is to some extent dependent upon zinc and the activity of zinc-transporting proteins or vice versa.
Collapse
Affiliation(s)
- Kamille Smidt
- Department of Pharmacology, University of Aarhus, DK-8000 Aarhus C, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
93
|
Alonso A, Fernández R, Ordóñez P, Moreno M, Patterson AM, González C. Regulation of estrogen receptor alpha by estradiol in pregnant and estradiol treated rats. Steroids 2006; 71:1052-61. [PMID: 17030051 DOI: 10.1016/j.steroids.2006.09.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Revised: 07/07/2006] [Accepted: 09/05/2006] [Indexed: 10/24/2022]
Abstract
Estrogens play an important role in tissue metabolism through specific regulation of several intracellular pathways. We studied ERalpha regulation in muscle and adipose tissue from pregnant and estradiol treated rats. In both groups, we identified three different ERalpha inmunoreactive proteins (80, 67 and 46 kDa) using total protein extracts. Because it has been showed that estrogens are able to promote rapid effects in several cellular models, we looked for three ERalpha-related proteins at plasma membrane. In skeletal muscle of both groups, we positively identified the three ERalpha-related isoforms in plasma membrane, but in adipose tissue from pregnant we were not able to identify ERalpha67, and in estradiol treated animals ERalpha80 was absent. Taking together, our results showed a tissue-specific regulation of whole-cell ERalpha-related proteins and ERalpha located at plasma membrane, which should be involved in non-genomic actions of 17beta-estradiol. The role of the three ERalpha inmunoreactive proteins is unknown, however, seems probably related to rapid activation of signalling pathways.
Collapse
Affiliation(s)
- Ana Alonso
- Department of Functional Biology. Physiology Area, University of Oviedo, C/ Julián Clavería s/n, 33006 Oviedo, Spain
| | | | | | | | | | | |
Collapse
|
94
|
Penza M, Montani C, Romani A, Vignolini P, Pampaloni B, Tanini A, Brandi ML, Alonso-Magdalena P, Nadal A, Ottobrini L, Parolini O, Bignotti E, Calza S, Maggi A, Grigolato PG, Di Lorenzo D. Genistein affects adipose tissue deposition in a dose-dependent and gender-specific manner. Endocrinology 2006; 147:5740-51. [PMID: 16959845 DOI: 10.1210/en.2006-0365] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The soy isoflavone genistein targets adipose tissue and elicits physiological effects that may vary based on dietary intake. We hypothesized that the adipose effects of genistein are dose and gender dependent. Four-week-old C57BL/6 male and female mice received daily oral doses of genistein (50-200,000 microg/kg.d) or 17beta-estradiol (E2) (5 microg/kg.d) for 15 d or a diet containing 800 ppm genistein. Genistein increased epididymal and renal fat pad and adipocyte size at doses up to 50,000 microg/kg.d or at 800 ppm in the diet in males but not in females. The alteration in adipocity correlated with changes in peripheral insulin resistance. These treatments increased genistein serum concentrations from 35+/-6 to 103+/-26 nM 12 h after treatment and lowered plasma triglycerides and cholesterol levels. The 200,000 microg/kg.d genistein dose decreased adipose tissue weight similarly to E2. This genistein dose down-regulated estrogen receptor (beta more than alpha) and progesterone receptor expression and induced estrogen-dependent adipose differentiation factors; it did not change expression of the minimal consensus estrogen-responsive element in ERE-tK-LUC mice, which was positively modulated in other tissues (e.g. the lung). E2 down-regulated almost all examined adipogenic factors. Gene microarray analysis identified factors in fat metabolism and obesity-related phenotypes differentially regulated by low and high doses of genistein, uncovering its adipogenic and antiadipogenic actions. The lower dose induced the phospholipase A2 group 7 and the phospholipid transfer protein genes; the 200,000 microg/kg.d dose inhibited them. The antiadipogenic action of genistein and down-regulation of adipogenic genes required the expression of ERbeta. In conclusion, nutritional doses of genistein are adipogenic in a gender-specific manner, whereas pharmacological doses inhibited adipose deposition.
Collapse
Affiliation(s)
- M Penza
- 3rd Laboratory/Biotechnology, and Department of Diagnostics, Civic Hospital of Brescia, 25123 and Department of Pathology, University of Brescia, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Dieudonné MN, Sammari A, Dos Santos E, Leneveu MC, Giudicelli Y, Pecquery R. Sex steroids and leptin regulate 11beta-hydroxysteroid dehydrogenase I and P450 aromatase expressions in human preadipocytes: Sex specificities. J Steroid Biochem Mol Biol 2006; 99:189-96. [PMID: 16621515 DOI: 10.1016/j.jsbmb.2006.01.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2005] [Accepted: 01/25/2006] [Indexed: 12/14/2022]
Abstract
Adipose tissue is an important site of steroid hormone biosynthesis, as type I 11beta-hydroxysteroid dehydrogenase (HSD1), the enzyme responsible for the conversion of cortisone into cortisol and the P450 aromatase, the enzyme catalysing androgens aromatization into estrogens, are both expressed in human adipose tissue. In the present report, we have investigated the possibility that sex steroids and leptin could regulate these two enzymes in cultured preadipocytes from men and women intra-abdominal fat depots. In women preadipocytes, human recombinant leptin down-regulates HSD1 mRNA expression (-58%) and P450 aromatase activity (-26%). Conversely, leptin up-regulates the HSD1 (2.4-fold) and the P450 aromatase (1.6-fold) mRNA expression in men preadipocytes. In women preadipocytes, 17beta-estradiol strongly stimulates HSD1 mRNA expression (10-fold) and, in contrast, decreases by half the P450 aromatase expression. In men, 17beta-estradiol has no influence on HSD1 expression but up-regulates P450 aromatase mRNA expression (2.4-fold). Finally, androgens increase by a factor of 2.5-5 the mRNA expression of both enzymes in men. These findings suggest that sex steroids and leptin either increase or decrease local cortisol and estrogens productions in men or in women preadipocytes, respectively. They also indicate that steroid metabolism in adipose tissue is controlled by a coordinated regulation of P450 aromatase and HSD1 expressions. Finally, the important sex-specific differences described herein may also contribute to explain the sexual dimorphism of body fat distribution in humans.
Collapse
Affiliation(s)
- Marie-Noëlle Dieudonné
- Service de Biochimie et de Biologie Moléculaire, UPRES EA 2493, Faculté de Médecine Paris-Ile de France-Ouest, Université Versailles St Quentin, Centre Hospitalier de Poissy, 78303 Poissy Cedex, France
| | | | | | | | | | | |
Collapse
|
96
|
Abstract
Acquired fat redistribution, that is, peripheral fat loss often accompanied by central fat accumulation in patients with HIV infection is the most common form of lipodystrophy in man. Approximately 30 - 50% of HIV-infected individuals after > or = 12 months on highly active antiretroviral therapy (HAART) may encounter the HIV-associated lipodystrophy syndrome (HALS), which attenuates patient compliance to this treatment. HALS is characterised by impaired glucose and lipid metabolism and other risk factors for cardiovascular disease. This review depicts the metabolic abnormalities associated with HAART by describing the key cell and organ systems that are involved, emphasising the role of insulin resistance. An opinion on the remedies available to treat the metabolic abnormalities and phenotype of HALS is provided.
Collapse
Affiliation(s)
- Steen B Haugaard
- Clinical Research Unit, Department of Endocrinology and Internal Medicine, Hvidovre University Hospital, DK 2650 Hvidovre, Copenhagen, Denmark.
| |
Collapse
|
97
|
Kuhl H. Pharmacology of estrogens and progestogens: influence of different routes of administration. Climacteric 2005; 8 Suppl 1:3-63. [PMID: 16112947 DOI: 10.1080/13697130500148875] [Citation(s) in RCA: 492] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
This review comprises the pharmacokinetics and pharmacodynamics of natural and synthetic estrogens and progestogens used in contraception and therapy, with special consideration of hormone replacement therapy. The paper describes the mechanisms of action, the relation between structure and hormonal activity, differences in hormonal pattern and potency, peculiarities in the properties of certain steroids, tissue-specific effects, and the metabolism of the available estrogens and progestogens. The influence of the route of administration on pharmacokinetics, hormonal activity and metabolism is presented, and the effects of oral and transdermal treatment with estrogens on tissues, clinical and serum parameters are compared. The effects of oral, transdermal (patch and gel), intranasal, sublingual, buccal, vaginal, subcutaneous and intramuscular administration of estrogens, as well as of oral, vaginal, transdermal, intranasal, buccal, intramuscular and intrauterine application of progestogens are discussed. The various types of progestogens, their receptor interaction, hormonal pattern and the hormonal activity of certain metabolites are described in detail. The structural formulae, serum concentrations, binding affinities to steroid receptors and serum binding globulins, and the relative potencies of the available estrogens and progestins are presented. Differences in the tissue-specific effects of the various compounds and regimens and their potential implications with the risks and benefits of hormone replacement therapy are discussed.
Collapse
Affiliation(s)
- H Kuhl
- Department of Obstetrics and Gynecology, J. W. Goethe University of Frankfurt, Germany
| |
Collapse
|
98
|
Misso ML, Jang C, Adams J, Tran J, Murata Y, Bell R, Boon WC, Simpson ER, Davis SR. Adipose aromatase gene expression is greater in older women and is unaffected by postmenopausal estrogen therapy. Menopause 2005; 12:210-5. [PMID: 15772569 DOI: 10.1097/00042192-200512020-00016] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Although natural menopause is associated with loss of ovarian estrogen production, this life phase is followed by a significant increase in estrogen-related cancers, namely breast and endometrial cancer. These tissues, as well as adipose, skeletal, and vascular tissues and the brain are important sites of postmenopausal estrogen production. Circulating C19 steroid precursors are essential substrates for extragonadal estrogen synthesis; however, the levels of these androgenic precursors decline markedly with advancing age. This implies an increase in capacity for extragonadal tissues to produce estrogen with age. DESIGN To explore this, and the effects of the menopause transition and postmenopausal estrogen therapy on extragonadal estrogen biosynthesis, we have compared the expression of the aromatase gene and estrogen (ER) and androgen receptors (AR) in subcutaneous abdominal and gluteal fat taken from premenopausal (group 1: n = 11), postmenopausal (group 2: n = 10), and postmenopausal women taking estrogen therapy (group 3: n = 10). All subjects were of normal body mass index, euglycemic, and normolipemic. RESULTS The postmenopausal women were older (group 1, 43.1 +/- 5.0 vs groups 2 and 3, 57.9 +/- 7.4 years, P < 0.001 and 56.1 +/- 4.5 years, P < 0.001, respectively) and had lower serum estradiol levels (group 2, 22.2 +/- 3.2 vs group 1, 442.5 +/- 248.2 pmol/L, P < 0.05), which were restored to premenopausal levels with estrogen therapy. Expression analysis revealed that levels of transcripts encoding aromatase were greater in gluteal than abdominal depots in each group in postmenopausal versus premenopausal women (P < 0.05). Use of hormone therapy did not influence aromatase gene expression in either depot. No differences were detected in the expression of ER or AR between groups of between tissue depots. CONCLUSION Thus, the capacity of adipose tissue to produce estrogen seems to increase significantly with age at the time of menopause and to be unaltered by exogenous estrogen therapy. This difference in extragonadal estrogen production with age may play a pivotal role in the increase in estrogen-dependent malignancies in the postmenopausal years.
Collapse
Affiliation(s)
- Marie L Misso
- Prince Henry's Institute of Medical Research, Clayton, Victoria, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Barzon L, Zamboni M, Pacenti M, Milan G, Bosello O, Federspil G, Palù G, Vettor R. Do oestrogen receptors play a role in the pathogenesis of HIV-associated lipodystrophy? AIDS 2005; 19:531-3. [PMID: 15764861 DOI: 10.1097/01.aids.0000162344.73804.a1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Epidemiological data show an increased risk of HIV-associated lipodystrophy in women, and sex hormone abnormalities have been reported with highly active antiretroviral therapy (HAART). This study, which demonstrates that oestrogen receptor beta expression is significantly reduced in the subcutaneous adipose tissue of HIV-infected lipodystrophic patients, downregulated by HAART regimens including protease inhibitors (PI), and restored after switching from PI, opens perspectives for the investigation of selective oestrogen receptor modifiers for the management of this syndrome.
Collapse
Affiliation(s)
- Luisa Barzon
- Departments of Histology, Microbiology and Medical Biotechnologies, University of Padova, Padova, Italy
| | | | | | | | | | | | | | | |
Collapse
|
100
|
van der Schouw YT, Sampson L, Willett WC, Rimm EB. The usual intake of lignans but not that of isoflavones may be related to cardiovascular risk factors in U.S. men. J Nutr 2005; 135:260-6. [PMID: 15671223 DOI: 10.1093/jn/135.2.260] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Isoflavone supplementation in the form of soy protein-containing isoflavones is associated with beneficial lipid changes. Information on usual isoflavone and lignan intakes in the diet of Western men and their associations with cardiovascular risk factors is not available. From the Health Professionals Follow-up Study, we selected 468 men, aged 47-83 y, who were free of cardiovascular disease, diabetes, and cancer, and who had provided a blood sample in 1994. We measured circulating total, LDL, and HDL cholesterol, triacylglycerol, lipoprotein(a), apolipoprotein (apo) A1, apoB, hemoglobin (Hb)A1c, insulin, C-peptide, and leptin concentrations. Isoflavone and lignan intakes were calculated from a FFQ. We used multivariate linear regression. None of the cardiovascular risk factors was strongly associated with isoflavone intake. Blood levels of LDL cholesterol and apoB tended to increase with increasing lignan intake [for LDL cholesterol, quartile 4 -quartile 1 = 9% (95% CI 1%; 16%), P for trend = 0.01, and for apo B, quartile 4 -quartile 1 = 9% (95% CI 1%; 16%), P for trend = 0.02]. Fasting insulin and C-peptide tended to decrease with increasing lignan intake [for insulin, quartile 4 -quartile 1 = -11% (95% CI -55%; -8%), P for trend = 0.02, and for C-peptide, quartile 4 -quartile 1 = -25% (95% CI -44; -6%), P for trend = 0.01]. Our results suggest that intake of isoflavones within the range of Western diets is not associated with a cardiovascular risk profile among men. Diets high in lignan intake may increase apoB-containing lipoproteins and decrease fasting insulin secretion, but these findings require confirmation.
Collapse
|