1
|
Kancheva R, Hill M, Velíková M, Kancheva L, Včelák J, Ampapa R, Židó M, Štětkářová I, Libertínová J, Vosátková M, Kubala Havrdová E. Altered Steroidome in Women with Multiple Sclerosis. Int J Mol Sci 2024; 25:12033. [PMID: 39596101 PMCID: PMC11593676 DOI: 10.3390/ijms252212033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) mainly afflicting young women. Various steroids can influence the onset and development of the disease or, on the contrary, mitigate its course; however, a systematic review of steroidomic changes in MS patients is lacking. Based on the gas chromatography tandem mass spectrometry (GC-MS/MS) platform and, in the case of estradiol, also using immunoassay, this study performed a comprehensive steroidomic analysis in 25 female MS patients aged 39(32, 49) years compared to 15 female age-matched controls aged 38(31, 46) years. A significant trend towards higher ratios of conjugated steroids to their unconjugated counterparts was found in patients, which is of particular interest in terms of the balance between excitatory and inhibitory steroid modulators of ionotropic receptors. Patients showed altered metabolic pathway to cortisol with decreased conversion of pregnenolone to 17-hydroxypregnenolone and 17-hydroxypregnenolone to 17-hydroxyprogesterone and increased conversion of 17-hydroxypregnenolone to dehydroepiandrosterone (DHEA), resulting in lower levels of 17-hydroxyprogesterone, as well as indications of impaired conversion of 11-deoxy-steroids to 11β-hydroxy-steroids but reduced conversion of cortisol to cortisone. Due to over-activation of hypothalamic-pituitary-adrenal axis (HPAA), however, cortisol and cortisone levels were higher in patients with indications of depleted cortisol synthesizing enzymes. Patients showed lower conversion of DHEA to androstenedione, androstenedione to testosterone, androstenedione to estradiol in the major pathway, and testosterone to estradiol in the minor pathway for estradiol synthesis at increased conversion of androstenedione to testosterone. They also showed lower conversion of immunoprotective Δ5 androstanes to their more potent 7α/β-hydroxy metabolites and had lower circulating allopregnanolone and higher ratio 3β-hydroxy-steroids to their neuroprotective 3α-hydroxy-counterparts.
Collapse
Affiliation(s)
- Radmila Kancheva
- Institute of Endocrinology, 11000 Prague, Czech Republic; (M.V.); (L.K.); (J.V.); (M.V.)
| | - Martin Hill
- Institute of Endocrinology, 11000 Prague, Czech Republic; (M.V.); (L.K.); (J.V.); (M.V.)
| | - Marta Velíková
- Institute of Endocrinology, 11000 Prague, Czech Republic; (M.V.); (L.K.); (J.V.); (M.V.)
| | - Ludmila Kancheva
- Institute of Endocrinology, 11000 Prague, Czech Republic; (M.V.); (L.K.); (J.V.); (M.V.)
| | - Josef Včelák
- Institute of Endocrinology, 11000 Prague, Czech Republic; (M.V.); (L.K.); (J.V.); (M.V.)
| | - Radek Ampapa
- MS Center, Jihlava Hospital, 58633 Jihlava, Czech Republic;
| | - Michal Židó
- Department of Neurology 3FM CU and UHKV, Third Faculty of Medicine, Charles University, 10000 Prague, Czech Republic; (M.Ž.); (I.Š.)
| | - Ivana Štětkářová
- Department of Neurology 3FM CU and UHKV, Third Faculty of Medicine, Charles University, 10000 Prague, Czech Republic; (M.Ž.); (I.Š.)
| | - Jana Libertínová
- MS Center, Second Faculty of Medicine, Charles University, 15006 Prague, Czech Republic;
| | - Michala Vosátková
- Institute of Endocrinology, 11000 Prague, Czech Republic; (M.V.); (L.K.); (J.V.); (M.V.)
| | - Eva Kubala Havrdová
- Department of Neurology, First Faculty of Medicine, Charles University, 12008 Prague, Czech Republic;
| |
Collapse
|
2
|
Galey L, Olanrewaju A, Nabi H, Paquette JS, Pouliot F, Audet-Walsh É. PSA, an outdated biomarker for prostate cancer: In search of a more specific biomarker, citrate takes the spotlight. J Steroid Biochem Mol Biol 2024; 243:106588. [PMID: 39025336 DOI: 10.1016/j.jsbmb.2024.106588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/12/2024] [Accepted: 07/14/2024] [Indexed: 07/20/2024]
Abstract
The prevailing biomarker employed for prostate cancer (PCa) screening and diagnosis is the prostate-specific antigen (PSA). Despite excellent sensitivity, PSA lacks specificity, leading to false positives, unnecessary biopsies and overdiagnosis. Consequently, PSA is increasingly less used by clinicians, thus underscoring the imperative for the identification of new biomarkers. An emerging biomarker in this context is citrate, a molecule secreted by the normal prostate, which has been shown to be inversely correlated with PCa. Here, we discuss about PSA and its usage for PCa diagnosis, its lack of specificity, and the various conditions that can affect its levels. We then provide our vision about what we think would be a valuable addition to our PCa diagnosis toolkit, citrate. We describe the unique citrate metabolic program in the prostate and how this profile is reprogrammed during carcinogenesis. Finally, we summarize the evidence that supports the usage of citrate as a biomarker for PCa diagnosis, as it can be measured in various patient samples and be analyzed by several methods. The unique relationship between citrate and PCa, combined with the stability of citrate levels in other prostate-related conditions and the simplicity of its detection, further accentuates its potential as a biomarker.
Collapse
Affiliation(s)
- Lucas Galey
- Endocrinology - Nephrology Research Axis, Centre de recherche du CHU de Québec - Université Laval, Québec City, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec City, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec City, Canada
| | - Ayokunle Olanrewaju
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Hermann Nabi
- Centre de recherche sur le cancer de l'Université Laval, Québec City, Canada
| | - Jean-Sébastien Paquette
- Laboratoire de recherche et d'innovation en médecine de première ligne (ARIMED), Groupe de médecine de famille universitaire de Saint-Charles-Borromée, CISSS Lanaudière, Saint-Charles-Borromée, QC, Canada; VITAM Research Centre for Sustainable Health, Québec, QC, Canada; Department of Family Medicine and Emergency Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Frédéric Pouliot
- Centre de recherche sur le cancer de l'Université Laval, Québec City, Canada; Department of Family Medicine and Emergency Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada; Department of surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Étienne Audet-Walsh
- Endocrinology - Nephrology Research Axis, Centre de recherche du CHU de Québec - Université Laval, Québec City, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec City, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec City, Canada.
| |
Collapse
|
3
|
Jugan JA, Jackson KB, Elmore SE, La Merrill MA. Impaired energy expenditure following exposure to either DDT or DDE in mice may be mediated by DNA methylation changes in brown adipose. ENVIRONMENTAL EPIGENETICS 2024; 10:dvae011. [PMID: 39403345 PMCID: PMC11472829 DOI: 10.1093/eep/dvae011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/09/2024] [Accepted: 08/16/2024] [Indexed: 10/19/2024]
Abstract
The insecticide dichlorodiphenyltrichloroethane (DDT) and its persistent metabolite, dichlorodiphenyldichloroethylene (DDE), have been associated with increased adiposity and obesity in multiple generations of rodents and humans. These lipophilic pollutants accumulate in adipose tissue and appear to decrease energy expenditure through the impairment of thermogenesis in brown adipose tissue (BAT). We hypothesized that impaired thermogenesis is due to persistent epigenetic modifications of BAT. To address this, we exposed C57BL/6 J mice to DDT or DDE from gestational day (GD) 11.5 to postnatal day (PND) 5, evaluated longitudinal body temperature, and performed reduced representation bisulfite sequencing and RNA sequencing of BAT from infant and adult offspring. Exposure to DDT or DDE reduced core body temperature in adult mice, and differential methylation at the pathway and gene level was persistent from infancy to adulthood. Furthermore, thermogenesis and biological pathways essential for thermogenic function, such as oxidative phosphorylation and mechanistic target of rapamycin kinase (mTOR) signaling, were enriched with differential methylation and RNA transcription in adult mice exposed to DDT or DDE. PAZ6 human brown preadipocytes were differentiated in the presence of DDT or DDE to understand the brown adipocyte-autonomous effect of these pollutants. In vitro exposure led to limited changes in RNA expression; however, mitochondrial membrane potential was decreased in vitro with 0.1 µM and 1 µM doses of DDT or DDE. These results demonstrate that concentrations of DDT and DDE relevant to human exposure have a significant effect on thermogenesis, the transcriptome, and DNA methylome of mouse BAT and the mitochondrial function of human brown adipocytes.
Collapse
Affiliation(s)
- Juliann A Jugan
- Department of Environmental Toxicology, University of California, Davis, Davis, CA 95616, United States
| | - Kyle B Jackson
- Department of Environmental Toxicology, University of California, Davis, Davis, CA 95616, United States
| | - Sarah E Elmore
- Department of Environmental Toxicology, University of California, Davis, Davis, CA 95616, United States
| | - Michele A La Merrill
- Department of Environmental Toxicology, University of California, Davis, Davis, CA 95616, United States
| |
Collapse
|
4
|
Hill M, Velíková M, Hovorková T, Bulant J, Janšáková K, Valeš K. Steroidomics in Men with Schizophrenia. Int J Mol Sci 2024; 25:8729. [PMID: 39201417 PMCID: PMC11354902 DOI: 10.3390/ijms25168729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 09/02/2024] Open
Abstract
Schizophrenia is associated with numerous abnormalities, including imbalances in all hormonal axes, among which steroids play a major role. Steroidomic studies therefore represent a promising tool for early diagnosis and appropriate treatment of schizophrenia. A total of 51 adult male schizophrenics aged 27 (22, 34) years (shown as median with quartiles) and 16 healthy controls (HCs) aged 28 (25, 32) years were enrolled into this study. Our results showed the effective differentiation of men with schizophrenia from controls based on steroidomic profiles. We also found an altered metabolic pathway from pregnenolone and its sulfate (PREG/S) to cortisol in schizophrenics with several metabolic bottlenecks such as lower PREG levels due to increased PREG sulfation and/or suppressed PREGS desulfation and attenuated conversion of 17-hydroxy-PREG to 17-hydroxy-progesterone, as well as the results suggestive of suppressed CYP11B1 activity. In contrast, steroid molar ratios suggested two counterregulatory steps involving increased conversion of PREG/S to 17-hydroxy-PREG/S and decreased conversion of cortisol to cortisone, which may maintain unchanged basal cortisol levels but may not ensure a sufficient cortisol response to stress. Our data also indicated a trend to higher 7α-, 7β-, and 16α-hydroxylation that may counteract the autoimmune complications and proinflammatory processes accompanying schizophrenia. Finally, a possible suppression of HSD17B3 activity was suggested, resulting in decreased circulating testosterone levels with increased androstenedione levels.
Collapse
Affiliation(s)
- Martin Hill
- Department of Steroids and Proteofactors, Institute of Endocrinology, Narodni 139/8, 110 00 Prague, Czech Republic; (M.V.); (T.H.); (J.B.)
| | - Marta Velíková
- Department of Steroids and Proteofactors, Institute of Endocrinology, Narodni 139/8, 110 00 Prague, Czech Republic; (M.V.); (T.H.); (J.B.)
| | - Tereza Hovorková
- Department of Steroids and Proteofactors, Institute of Endocrinology, Narodni 139/8, 110 00 Prague, Czech Republic; (M.V.); (T.H.); (J.B.)
| | - Josef Bulant
- Department of Steroids and Proteofactors, Institute of Endocrinology, Narodni 139/8, 110 00 Prague, Czech Republic; (M.V.); (T.H.); (J.B.)
| | - Katarína Janšáková
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, 811 08 Bratislava, Slovakia;
| | - Karel Valeš
- National Institute of Mental Health, Topolova 748, 250 67 Klecany, Czech Republic;
- Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, 100 00 Prague, Czech Republic
| |
Collapse
|
5
|
Podraza J, Gutowska K, Lenartowicz A, Wąsowski M, Jonas MI, Bartoszewicz Z, Lisik W, Jonas M, Binda A, Jaworski P, Tarnowski W, Noszczyk B, Puzianowska-Kuźnicka M, Kuryłowicz A. The Role of microRNA in the Regulation of Cortisol Metabolism in the Adipose Tissue in the Course of Obesity. Int J Mol Sci 2024; 25:5058. [PMID: 38791098 PMCID: PMC11120731 DOI: 10.3390/ijms25105058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/30/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024] Open
Abstract
The similarity of the clinical picture of metabolic syndrome and hypercortisolemia supports the hypothesis that obesity may be associated with impaired expression of genes related to cortisol action and metabolism in adipose tissue. The expression of genes encoding the glucocorticoid receptor alpha (GR), cortisol metabolizing enzymes (HSD11B1, HSD11B2, H6PDH), and adipokines, as well as selected microRNAs, was measured by real-time PCR in adipose tissue from 75 patients with obesity, 19 patients following metabolic surgery, and 25 normal-weight subjects. Cortisol levels were analyzed by LC-MS/MS in 30 pairs of tissues. The mRNA levels of all genes studied were significantly (p < 0.05) decreased in the visceral adipose tissue (VAT) of patients with obesity and normalized by weight loss. In the subcutaneous adipose tissue (SAT), GR and HSD11B2 were affected by this phenomenon. Negative correlations were observed between the mRNA levels of the investigated genes and selected miRNAs (hsa-miR-142-3p, hsa-miR-561, and hsa-miR-579). However, the observed changes did not translate into differences in tissue cortisol concentrations, although levels of this hormone in the SAT of patients with obesity correlated negatively with mRNA levels for adiponectin. In conclusion, although the expression of genes related to cortisol action and metabolism in adipose tissue is altered in obesity and miRNAs may be involved in this process, these changes do not affect tissue cortisol concentrations.
Collapse
Affiliation(s)
- Jakub Podraza
- The Faculty of Biology and Biotechnology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland;
| | - Klaudia Gutowska
- II Department of Obstetrics and Gynecology, Warsaw Medical University, 00-315 Warsaw, Poland;
| | | | - Michał Wąsowski
- Department of General Medicine and Geriatric Cardiology, Medical Centre of Postgraduate Education, 00-401 Warsaw, Poland;
| | - Marta Izabela Jonas
- Department of Human Epigenetics, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (M.I.J.); (M.P.-K.)
| | - Zbigniew Bartoszewicz
- Department of Internal Medicine and Endocrinology, The Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Wojciech Lisik
- Department of General and Transplantation Surgery, The Medical University of Warsaw, 00-694 Warsaw, Poland
| | - Maurycy Jonas
- Department of General Surgery, Barska Hospital, 02-315 Warsaw, Poland;
| | - Artur Binda
- Department of General, Oncological and Bariatric Surgery, Medical Centre of Postgraduate Education, 00-401 Warsaw, Poland (W.T.)
| | - Paweł Jaworski
- Department of General, Oncological and Bariatric Surgery, Medical Centre of Postgraduate Education, 00-401 Warsaw, Poland (W.T.)
| | - Wiesław Tarnowski
- Department of General, Oncological and Bariatric Surgery, Medical Centre of Postgraduate Education, 00-401 Warsaw, Poland (W.T.)
| | - Bartłomiej Noszczyk
- Department of Plastic Surgery, Medical Centre of Postgraduate Education, 00-401 Warsaw, Poland;
| | - Monika Puzianowska-Kuźnicka
- Department of Human Epigenetics, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (M.I.J.); (M.P.-K.)
- Department of Geriatrics and Gerontology, Medical Centre of Postgraduate Education, 01-826 Warsaw, Poland
| | - Alina Kuryłowicz
- Department of General Medicine and Geriatric Cardiology, Medical Centre of Postgraduate Education, 00-401 Warsaw, Poland;
- Department of Human Epigenetics, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (M.I.J.); (M.P.-K.)
| |
Collapse
|
6
|
Alemany M. The Metabolic Syndrome, a Human Disease. Int J Mol Sci 2024; 25:2251. [PMID: 38396928 PMCID: PMC10888680 DOI: 10.3390/ijms25042251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
This review focuses on the question of metabolic syndrome (MS) being a complex, but essentially monophyletic, galaxy of associated diseases/disorders, or just a syndrome of related but rather independent pathologies. The human nature of MS (its exceptionality in Nature and its close interdependence with human action and evolution) is presented and discussed. The text also describes the close interdependence of its components, with special emphasis on the description of their interrelations (including their syndromic development and recruitment), as well as their consequences upon energy handling and partition. The main theories on MS's origin and development are presented in relation to hepatic steatosis, type 2 diabetes, and obesity, but encompass most of the MS components described so far. The differential effects of sex and its biological consequences are considered under the light of human social needs and evolution, which are also directly related to MS epidemiology, severity, and relations with senescence. The triggering and maintenance factors of MS are discussed, with especial emphasis on inflammation, a complex process affecting different levels of organization and which is a critical element for MS development. Inflammation is also related to the operation of connective tissue (including the adipose organ) and the widely studied and acknowledged influence of diet. The role of diet composition, including the transcendence of the anaplerotic maintenance of the Krebs cycle from dietary amino acid supply (and its timing), is developed in the context of testosterone and β-estradiol control of the insulin-glycaemia hepatic core system of carbohydrate-triacylglycerol energy handling. The high probability of MS acting as a unique complex biological control system (essentially monophyletic) is presented, together with additional perspectives/considerations on the treatment of this 'very' human disease.
Collapse
Affiliation(s)
- Marià Alemany
- Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Catalonia, Spain
| |
Collapse
|
7
|
Vallée M. Advances in steroid research from the pioneering neurosteroid concept to metabolomics: New insights into pregnenolone function. Front Neuroendocrinol 2024; 72:101113. [PMID: 37993022 DOI: 10.1016/j.yfrne.2023.101113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/13/2023] [Accepted: 11/19/2023] [Indexed: 11/24/2023]
Abstract
Advances in neuroendocrinology have led to major discoveries since the 19th century, identifying adaptive loops for maintaining homeostasis. One of the most remarkable discoveries was the concept of neurosteroids, according to which the brain is not only a target but also a source of steroid production. The identification of new membrane steroid targets now underpins the neuromodulatory effects of neurosteroids such as pregnenolone, which is involved in functions mediated by the GPCR CB1 receptor. Structural analysis of steroids is a key feature of their interactions with the phospholipid membrane, receptors and resulting activity. Therefore, mass spectrometry-based methods have been developed to elucidate the metabolic pathways of steroids, the ultimate approach being metabolomics, which allows the identification of a large number of metabolites in a single sample. This approach should enable us to make progress in understanding the role of neurosteroids in the functioning of physiological and pathological processes.
Collapse
Affiliation(s)
- Monique Vallée
- University Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000 Bordeaux, France.
| |
Collapse
|
8
|
Engin AB, Engin A. The Checkpoints of Intestinal Fat Absorption in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:73-95. [PMID: 39287849 DOI: 10.1007/978-3-031-63657-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
In this chapter, intestinal lipid transport, which plays a central role in fat homeostasis and the development of obesity in addition to the mechanisms of fatty acids and monoacylglycerol absorption in the intestinal lumen and reassembly of these within the enterocyte was described. A part of the resynthesized triglycerides (triacylglycerols; TAG) is repackaged in the intestine to form the hydrophobic core of chylomicrons (CMs). These are delivered as metabolic fuels, essential fatty acids, and other lipid-soluble nutrients, from enterocytes to the peripheral tissues following detachment from the endoplasmic reticulum membrane. Moreover, the attitudes of multiple receptor functions in dietary lipid uptake, synthesis, and transport are highlighted. Additionally, intestinal fatty acid binding proteins (FABPs), which increase the cytosolic flux of fatty acids via intermembrane transfer in enterocytes, and the functions of checkpoints for receptor-mediated fatty acid signaling are debated. The importance of the balance between storage and secretion of dietary fat by enterocytes in determining the physiological fate of dietary fat, including regulation of blood lipid concentrations and energy balance, is mentioned. Consequently, promising checkpoints regarding how intestinal fat processing affects lipid homeostatic mechanisms and lipid stores in the body and the prevention of obesity-lipotoxicity due to excessive intestinal lipid absorption are evaluated. In this context, dietary TAG digestion, pharmacological inhibition of TAG hydrolysis, the regulation of long-chain fatty acid uptake traffic into adipocytes, intracellular TAG resynthesis, the enlargement of cytoplasmic lipid droplets in enterocytes and constitutional alteration of their proteome, CD36-mediated conversion of diet-derived fatty acid into cellular lipid messengers and their functions are discussed.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey
| |
Collapse
|
9
|
Sumińska M, Podgórski R, Fichna P, Mazur A, Fichna M. Assessment of steroid enzymes action in children and adolescents with obesity. Steroids 2023; 200:109325. [PMID: 37806604 DOI: 10.1016/j.steroids.2023.109325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 10/10/2023]
Abstract
Rising prevalence of obesity has become an important impulse to investigate basic mechanisms involved in regulating the energy balance. It is widely accepted that steroids are potent factors affecting glucose, fat, and protein metabolism. Our study was aimed to analyze differences in the total amount of selected enzymes implicated in steroid metabolism in a group of children suffering from obesity and those with normal weight, further subdivided according to sex and pubertal stage. Data were obtained from 187 Caucasian children and adolescents, including 113 patients (63 girls, 50 boys) with obesity and 74 (34 girls, 40 boys) normal weight volunteers. Standard clinical examinations were performed in both groups. To evaluate the impact of puberty, preadolescent children and those with advanced puberty were assessed separately. Urine steroid excretion profiles were analyzed using gas chromatography/mass spectrometry method. Children with obesity revealed several changes in in the total amount of steroid enzymes as assessed by the relevant metabolite proportions, compared to their norm weight peers. Girls showed a significant increase in the activity of 11βHSD1, while boys demonstrated a relevant elevation in 20αHSD action. Regardless of sex, children with obesity showed an increase in the activity of 5β-reductase + 3αHSD complex and a decrease in the involvement of 11βOH-lase. The effect is attenuated when consider pre- and pubertal subgroups. We hypothesize that changes in the activity levels of selected enzymes may be a compensatory mechanism to limit the glucocorticoid exposure of key target tissues as well as to improve metabolic control and reduce long-term complications of obesity.
Collapse
Affiliation(s)
- Marta Sumińska
- Department of Pediatric Diabetes, Auxology and Obesity, Institute of Pediatrics, Poznan University of Medical Sciences, Poznan, Poland; Doctoral School, Poznan University of Medical Sciences, Poznan, Poland.
| | - Rafał Podgórski
- Department of Biochemistry, Institute of Medical Sciences, Collegium of Medical Sciences, University of Rzeszow, Rzeszow, Poland
| | - Piotr Fichna
- Department of Pediatric Diabetes, Auxology and Obesity, Institute of Pediatrics, Poznan University of Medical Sciences, Poznan, Poland
| | - Artur Mazur
- Department of Pediatrics, Childhood Endocrinology and Diabetes, Collegium of Medical Sciences, University of Rzeszow, Rzeszow, Poland
| | - Marta Fichna
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
10
|
Poutanen M, Hagberg Thulin M, Härkönen P. Targeting sex steroid biosynthesis for breast and prostate cancer therapy. Nat Rev Cancer 2023:10.1038/s41568-023-00609-y. [PMID: 37684402 DOI: 10.1038/s41568-023-00609-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/20/2023] [Indexed: 09/10/2023]
Affiliation(s)
- Matti Poutanen
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.
- Turku Center for Disease Modelling, University of Turku, Turku, Finland.
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- FICAN West Cancer Center, University of Turku and Turku University Hospital, Turku, Finland.
| | - Malin Hagberg Thulin
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Pirkko Härkönen
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- FICAN West Cancer Center, University of Turku and Turku University Hospital, Turku, Finland
| |
Collapse
|
11
|
Vaňková M, Velíková M, Vejražková D, Včelák J, Lukášová P, Rusina R, Vaňková H, Jarolímová E, Kancheva R, Bulant J, Horáčková L, Bendlová B, Hill M. The Role of Steroidomics in the Diagnosis of Alzheimer's Disease and Type 2 Diabetes Mellitus. Int J Mol Sci 2023; 24:ijms24108575. [PMID: 37239922 DOI: 10.3390/ijms24108575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/04/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Epidemiological studies suggest an association between Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM). This study aimed to investigate the pathophysiological markers of AD vs. T2DM for each sex separately and propose models that would distinguish control, AD, T2DM, and AD-T2DM comorbidity groups. AD and T2DM differed in levels of some circulating steroids (measured mostly by GC-MS) and in other observed characteristics, such as markers of obesity, glucose metabolism, and liver function tests. Regarding steroid metabolism, AD patients (both sexes) had significantly higher sex hormone binding globulin (SHBG), cortisol, and 17-hydroxy progesterone, and lower estradiol and 5α-androstane-3α,17β-diol, compared to T2DM patients. However, compared to healthy controls, changes in the steroid spectrum (especially increases in levels of steroids from the C21 group, including their 5α/β-reduced forms, androstenedione, etc.) were similar in patients with AD and patients with T2DM, though more expressed in diabetics. It can be assumed that many of these steroids are involved in counter-regulatory protective mechanisms that mitigate the development and progression of AD and T2DM. In conclusion, our results demonstrated the ability to effectively differentiate AD, T2DM, and controls in both men and women, distinguish the two pathologies from each other, and differentiate patients with AD and T2DM comorbidities.
Collapse
Affiliation(s)
- Markéta Vaňková
- Institute of Endocrinology, Národní 8, 110 00 Prague, Czech Republic
| | - Marta Velíková
- Institute of Endocrinology, Národní 8, 110 00 Prague, Czech Republic
| | | | - Josef Včelák
- Institute of Endocrinology, Národní 8, 110 00 Prague, Czech Republic
| | - Petra Lukášová
- Institute of Endocrinology, Národní 8, 110 00 Prague, Czech Republic
| | - Robert Rusina
- Department of Neurology, Third Faculty of Medicine, Charles University and Thomayer University Hospital, Ruská 2411, 100 00 Prague, Czech Republic
| | - Hana Vaňková
- Third Faculty of Medicine, Charles University, Ruská 2411, 100 00 Prague, Czech Republic
| | - Eva Jarolímová
- Third Faculty of Medicine, Charles University, Ruská 2411, 100 00 Prague, Czech Republic
| | - Radmila Kancheva
- Institute of Endocrinology, Národní 8, 110 00 Prague, Czech Republic
| | - Josef Bulant
- Institute of Endocrinology, Národní 8, 110 00 Prague, Czech Republic
| | - Lenka Horáčková
- Institute of Endocrinology, Národní 8, 110 00 Prague, Czech Republic
| | - Běla Bendlová
- Institute of Endocrinology, Národní 8, 110 00 Prague, Czech Republic
| | - Martin Hill
- Institute of Endocrinology, Národní 8, 110 00 Prague, Czech Republic
| |
Collapse
|
12
|
Kvernebo Sunnergren K, Dahlgren J, Karlsson AK, Nilsson S, Allvin K, Ankarberg-Lindgren C. Pre- and peripubertal sex steroids are inversely associated with birth weight in preterm boys. Clin Endocrinol (Oxf) 2023; 98:342-350. [PMID: 36071648 DOI: 10.1111/cen.14821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 08/09/2022] [Accepted: 08/15/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE The relationship between sex hormone concentrations during childhood and birth weight (BW) is poorly understood. We aimed to investigate this relationship and the associations with anthropometric data at 5, 6, 7, 8, and 10 years of age in preterm boys. DESIGN A prospective longitudinal single-centre study, including 58 boys with a BW of 1325-3320 g and gestational age (GA) of 32 + 2 to 36 + 6 weeks. PATIENTS AND MEASUREMENTS Data on GA, BW and anthropometric data between 5 and 10 years of age were recorded. Testicular development was assessed at 8 and 10 years of age. Serum concentrations of sex steroids were analysed with gas chromatography-tandem mass spectrometry at 5-10 years and luteinizing hormone (LH) and follicle-stimulating hormone (FSH) with immunoassays at 10 years of age. RESULTS At 8 years of age, testosterone and estrone correlated negatively with BW, (ρ = -0.35, p = .021) and (ρ = -0.34, p = .024), respectively. At 10 years of age, testosterone, dihydrotestosterone, estrone and estradiol correlated negatively with BW (ρ = -0.39, p = .010), (ρ = -0.38, p = .013), (ρ = -0.44, p = .003) and (ρ = -0.36, p = .019), respectively. Weight gain from birth correlated with testosterone at 5 years (ρ = 0.40, p = .002), 7 years (ρ = 0.30, p = .040), 8 years (ρ = 0.44, p = .003) and 10 years (ρ = 0.40, p = .008) of age. At 10 years of age, testosterone correlated with LH (ρ = 0.42, p = .006) and FSH (ρ = 0.33, p = .033) but not with testicular volume. CONCLUSIONS Lower BW was associated with increased sex steroid concentrations from 8 years of age, independently of clinical signs of puberty.
Collapse
Affiliation(s)
- Kjersti Kvernebo Sunnergren
- Department of Pediatrics, Göteborg Pediatric Growth Research Center (GP-GRC), Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Pediatrics, Ryhov County Hospital, Jönköping, Sweden
| | - Jovanna Dahlgren
- Department of Pediatrics, Göteborg Pediatric Growth Research Center (GP-GRC), Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Pediatrics, Region Västra Götaland, Sahlgrenska University Hospital, Queen Silvia Children's Hospital, Gothenburg, Sweden
| | - Ann-Katrine Karlsson
- Department of Pediatrics, Göteborg Pediatric Growth Research Center (GP-GRC), Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Staffan Nilsson
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kerstin Allvin
- Department of Pediatrics, Göteborg Pediatric Growth Research Center (GP-GRC), Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Pediatrics, Region Västra Götaland, Sahlgrenska University Hospital, Queen Silvia Children's Hospital, Gothenburg, Sweden
| | - Carina Ankarberg-Lindgren
- Department of Pediatrics, Göteborg Pediatric Growth Research Center (GP-GRC), Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
13
|
Kuryłowicz A. Estrogens in Adipose Tissue Physiology and Obesity-Related Dysfunction. Biomedicines 2023; 11:biomedicines11030690. [PMID: 36979669 PMCID: PMC10045924 DOI: 10.3390/biomedicines11030690] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/30/2023] Open
Abstract
Menopause-related decline in estrogen levels is accompanied by a change in adipose tissue distribution from a gynoid to an android and an increased prevalence of obesity in women. These unfavorable phenomena can be partially restored by hormone replacement therapy, suggesting a significant role for estrogen in the regulation of adipocytes' function. Indeed, preclinical studies proved the involvement of these hormones in adipose tissue development, metabolism, and inflammatory activity. However, the relationship between estrogen and obesity is bidirectional. On the one hand-their deficiency leads to excessive fat accumulation and impairs adipocyte function, on the other-adipose tissue of obese individuals is characterized by altered expression of estrogen receptors and key enzymes involved in their synthesis. This narrative review aims to summarize the role of estrogen in adipose tissue development, physiology, and in obesity-related dysfunction. Firstly, the estrogen classification, synthesis, and modes of action are presented. Next, their role in regulating adipogenesis and adipose tissue activity in health and the course of obesity is described. Finally, the potential therapeutic applications of estrogen and its derivates in obesity treatment are discussed.
Collapse
Affiliation(s)
- Alina Kuryłowicz
- Department of Human Epigenetics, Mossakowski Medical Research Centre PAS, 02-106 Warsaw, Poland
- Department of General Medicine and Geriatric Cardiology, Medical Centre of Postgraduate Education, 00-401 Warsaw, Poland
| |
Collapse
|
14
|
Söderqvist G, Naessén S. Androgens impact on psychopathological variables according to CPRS, and EDI 2 scores: In women with bulimia nervosa, and eating disorder not otherwise specified. J Steroid Biochem Mol Biol 2023; 226:106217. [PMID: 36368624 DOI: 10.1016/j.jsbmb.2022.106217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/28/2022] [Accepted: 11/06/2022] [Indexed: 11/09/2022]
Abstract
Bulimia nervosa (BN) is characterized by binge eating, compensatory behavior, over-evaluation of weight and shape, which often co-occur with symptoms of anxiety and depression. Depression is the most common comorbid diagnosis in women with eating disorders. The role of androgens in the pathophysiology of depression has been recognized in recent years. However, the research on psychopathological comorbidity and androgen levels in bulimic disease is sparse. This study aimed to investigate, if there were any correlations between the androgens, testosterone (T), dehydroepiandrosterone sulphate (DHEAS), androstenedione (A4), 5α-dihydrotestosterone, (5α-DHT), and test scores of psychopathological variables, in women with bulimia nervosa (BN), eating disorder not otherwise specified of purging subtype (EDNOS-P) assessed by CPRS, and EDI 2. Women with DSM-IV diagnosis of BN (n = 36), EDNOS-P (n = 27), and healthy control subjects (n = 58) evaluated for fifteen psychopathological variables, i.a. depressive symptoms, impulsivity, personal traits, as well as serum androgen levels. All women were euthyroid, and polycystic ovarian syndrome (PCOS) diagnosis was excluded. Although androgen levels were almost equal for all three groups, significant correlations between core psychopathological symptoms (9/15) of bulimia nervosa and the most potent endogenous androgen, 5α-DHT, was found only in the EDNOS-P group. The role of 5α-DHT in women is not fully elucidated. Both animal and human studies have shown that the brain is able to locally synthesize steroids de novo and is a target of steroid hormones. Maybe these results can be interpreted in the light of differences in androgen receptor variability, metabolism and origin of T and 5α-DHT.
Collapse
Affiliation(s)
- Gunnar Söderqvist
- Department of Women's, and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Sabine Naessén
- Department of Women's, and Children's Health, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
15
|
Fouad Mansour M, Blanchette S, Pelletier M, Poirier D, Tchernof A. 17β-hydroxysteroid dehydrogenase type 2 activity, expression and cellular localization in abdominal adipose tissues from women. Clin Endocrinol (Oxf) 2023; 98:229-237. [PMID: 36267004 DOI: 10.1111/cen.14838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/26/2022] [Accepted: 10/08/2022] [Indexed: 01/04/2023]
Abstract
CONTEXT 17β-hydroxysteroid dehydrogenase type 2 (17β-HSD2) may be involved in the local modulation of estradiol (E2) availability in adipose tissues. OBJECTIVE To assess the conversion of E2 into estrone (E1) as well as the expression of 17β-HSD2 and its localization in omental (OM) and subcutaneous (SC) adipose tissues obtained from women. METHODS Rates of 14 C-E1 formation from 14 C-E2 were measured in OM and SC adipose tissue homogenates from 29 women. Specific 17β-HSD2 inhibitor EM-919 was tested in OM and SC adipose tissue homogenates (n = 6). 17β-HSD2 mRNA expression was measured in whole OM and SC adipose tissues (n = 14). Cellular localization of the enzyme was examined using immunohistochemistry. Anthropometric measurements were obtained and body composition as well as body fat distribution were measured. RESULTS Significant 14 C-E1 formation from 14 C-E2 in OM and SC tissue homogenates was detected. The rate of 14 C-E1 formation was significantly higher in OM than SC adipose tissue (p < .0001). The conversion of 14 C-E2 to 14 C-E1 was significantly inhibited by EM-919 in OM (p < .05) and SC (p < .05) adipose tissues. Significantly higher expression of 17β-HSD2 mRNA in OM versus SC fat was found (p = .03). 17β-HSD2 was localized in the vasculature of OM and SC tissues. Significant negative associations were detected between OM 17β-HSD2 activity and body mass index, WC, lean body mass as well as SC adipose tissue areas. CONCLUSION 17β-HSD2 converts E2 to E1 in OM and SC adipose tissues of women. The activity of this enzyme decreases with increasing adiposity.
Collapse
Affiliation(s)
- Mohamed Fouad Mansour
- Québec Heart and Lung Institute Research Center, Université Laval, Québec, Québec, Canada
- Department of Biochemistry, Faculty of Veterinary Medicine, Endocrinology and Nephrology, Zagazig University, Zagazig, Egypt
| | - Sophie Blanchette
- Québec Heart and Lung Institute Research Center, Université Laval, Québec, Québec, Canada
| | - Mélissa Pelletier
- Québec Heart and Lung Institute Research Center, Université Laval, Québec, Québec, Canada
| | | | - André Tchernof
- Québec Heart and Lung Institute Research Center, Université Laval, Québec, Québec, Canada
- Department of Biochemistry, Faculty of Veterinary Medicine, Endocrinology and Nephrology, Zagazig University, Zagazig, Egypt
- School of nutrition, Université Laval, Québec, Québec, Canada
| |
Collapse
|
16
|
Li S, Lee W, Heo W, Son HY, Her Y, Kim JI, Moon HG. AKR1C2 Promotes Metastasis and Regulates the Molecular Features of Luminal Androgen Receptor Subtype in Triple Negative Breast Cancer Cells. J Breast Cancer 2022; 26:60-76. [PMID: 36762781 PMCID: PMC9981988 DOI: 10.4048/jbc.2023.26.e1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/12/2022] [Accepted: 11/18/2022] [Indexed: 01/07/2023] Open
Abstract
PURPOSE Patients with triple-negative breast cancer (TNBC) have an increased risk of distant metastasis compared to those with other subtypes. In this study, we aimed to identify the genes associated with distant metastasis in TNBC and their underlying mechanisms. METHODS We established patient-derived xenograft (PDX) models using surgically resected breast cancer tissues from 31 patients with TNBC. Among these, 15 patients subsequently developed distant metastases. Candidate metastasis-associated genes were identified using RNA sequencing. In vitro wound healing, proliferation, migration, and invasion assays and in vivo tumor xenograft and metastasis assays were performed to determine the functional importance of aldo-keto reductase family 1 member C2 (AKR1C2). Additionally, we used the METABRIC dataset to investigate the potential role of AKR1C2 in regulating TNBC subtypes and their downstream signaling activities. RESULTS RNA sequencing of primary and PDX tumors showed that genes involved in steroid hormone biosynthesis, including AKR1C2, were significantly upregulated in patients who subsequently developed metastasis. In vitro and in vivo assays showed that silencing of AKR1C2 resulted in reduced cell proliferation, migration, invasion, tumor growth, and incidence of lung metastasis. AKR1C2 was upregulated in the luminal androgen receptor (LAR) subtype of TNBC in the METABRIC dataset, and AKR1C2 silencing resulted in the downregulation of LAR classifier genes in TNBC cell lines. The androgen receptor (AR) gene was a downstream mediator of AKR1C2-associated phenotypes in TNBC cells. AKR1C2 expression was associated with gene expression pathways that regulate AR expression, including JAK-STAT signaling or interleukin 6 (IL-6). The levels of phospho-signal transducer and activator of transcription and IL-6, along with secreted IL-6, were significantly downregulated in AKR1C2-silenced TNBC cells. CONCLUSION Our data indicate that AKR1C2 is an important regulator of cancer growth and metastasis in TNBC and may be a critical determinant of LAR subtype features.
Collapse
Affiliation(s)
- Songbin Li
- Interdisciplinary Graduate Program in Cancer Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Woochan Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.,Genomic Medicine Institute, Medical Research Center, Seoul, Korea
| | - Woohang Heo
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Korea
| | - Hye-Youn Son
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Korea
| | - Yujeong Her
- Interdisciplinary Graduate Program in Cancer Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Jong-Il Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.,Genomic Medicine Institute, Medical Research Center, Seoul, Korea
| | - Hyeong-Gon Moon
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Department of Surgery, Seoul National University Hospital, Seoul, Korea.,Department of Surgery, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
17
|
Ostinelli G, Laforest S, Denham SG, Gauthier MF, Drolet-Labelle V, Scott E, Hould FS, Marceau S, Homer NZM, Bégin C, Andrew R, Tchernof A. Increased Adipose Tissue Indices of Androgen Catabolism and Aromatization in Women With Metabolic Dysfunction. J Clin Endocrinol Metab 2022; 107:e3330-e3342. [PMID: 35511873 PMCID: PMC9282357 DOI: 10.1210/clinem/dgac261] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Indexed: 02/02/2023]
Abstract
CONTEXT Body fat distribution is a risk factor for obesity-associated comorbidities, and adipose tissue dysfunction plays a role in this association. In humans, there is a sex difference in body fat distribution, and steroid hormones are known to regulate several cellular processes within adipose tissue. OBJECTIVE Our aim was to investigate if intra-adipose steroid concentration and expression or activity of steroidogenic enzymes were associated with features of adipose tissue dysfunction in individuals with severe obesity. METHODS Samples from 40 bariatric candidates (31 women, 9 men) were included in the study. Visceral (VAT) and subcutaneous adipose tissue (SAT) were collected during surgery. Adipose tissue morphology was measured by a combination of histological staining and semi-automated quantification. Following extraction, intra-adipose and plasma steroid concentrations were determined by liquid chromatography electrospray ionization tandem mass spectrometry (LC-ESI-MS/MS). Aromatase activity was estimated using product over substrate ratio, while AKR1C2 activity was measured directly by fluorogenic probe. Gene expression was measured by quantitative PCR. RESULTS VAT aromatase activity was positively associated with VAT adipocyte hypertrophy (P valueadj < 0.01) and negatively with plasma high-density lipoprotein (HDL)-cholesterol (P valueadj < 0.01), while SAT aromatase activity predicted dyslipidemia in women even after adjustment for waist circumference, age, and hormonal contraceptive use. We additionally compared women with high and low visceral adiposity index (VAI) and found that VAT excess is characterized by adipose tissue dysfunction, increased androgen catabolism mirrored by increased AKR1C2 activity, and higher aromatase expression and activity indices. CONCLUSION In women, increased androgen catabolism or aromatization is associated with visceral adiposity and adipose tissue dysfunction.
Collapse
Affiliation(s)
- Giada Ostinelli
- Centre de recherche de l’Institut universitaire de cardiologie et pneumologie de Québec-Université Laval, Québec City, QC G1V 4G5, Canada
- École de nutrition, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Sofia Laforest
- Centre de recherche de l’Institut universitaire de cardiologie et pneumologie de Québec-Université Laval, Québec City, QC G1V 4G5, Canada
- École de nutrition, Université Laval, Québec City, QC G1V 0A6, Canada
- University of Strathclyde, Glasgow G1 1XQ, UK
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, University/BHF, Cardiovascular Sciences, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - Scott G Denham
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, University/BHF, Cardiovascular Sciences, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - Marie-Frederique Gauthier
- Centre de recherche de l’Institut universitaire de cardiologie et pneumologie de Québec-Université Laval, Québec City, QC G1V 4G5, Canada
| | | | - Emma Scott
- Faculté de médecine, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Frédéric-Simon Hould
- Centre de recherche de l’Institut universitaire de cardiologie et pneumologie de Québec-Université Laval, Québec City, QC G1V 4G5, Canada
- Faculté de médecine, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Simon Marceau
- Centre de recherche de l’Institut universitaire de cardiologie et pneumologie de Québec-Université Laval, Québec City, QC G1V 4G5, Canada
- Faculté de médecine, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Natalie Z M Homer
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, University/BHF, Cardiovascular Sciences, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - Catherine Bégin
- Centre de recherche de l’Institut universitaire de cardiologie et pneumologie de Québec-Université Laval, Québec City, QC G1V 4G5, Canada
- École de psychologie, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Ruth Andrew
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, University/BHF, Cardiovascular Sciences, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, EH16 4TJ, UK
- BHF/CVS, Queen’s Medical Research Institute, University of Edinburgh, EH16 4TJ, UK
| | - André Tchernof
- Correspondence: Andre Tchernof, PhD, Quebec Heart and Lung Institute, School of Nutrition, Laval University, 2725 Chemin Sainte-Foy (Y-4212), Québec, QC G1V 4G5, Canada.
| |
Collapse
|
18
|
SARZYNSKI MARKA, RICE TREVAK, DESPRÉS JEANPIERRE, PÉRUSSE LOUIS, TREMBLAY ANGELO, STANFORTH PHILIPR, TCHERNOF ANDRÉ, BARBER JACOBL, FALCIANI FRANCESCO, CLISH CLARY, ROBBINS JEREMYM, GHOSH SUJOY, GERSZTEN ROBERTE, LEON ARTHURS, SKINNER JAMESS, RAO DC, BOUCHARD CLAUDE. The HERITAGE Family Study: A Review of the Effects of Exercise Training on Cardiometabolic Health, with Insights into Molecular Transducers. Med Sci Sports Exerc 2022; 54:S1-S43. [PMID: 35611651 PMCID: PMC9012529 DOI: 10.1249/mss.0000000000002859] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The aim of the HERITAGE Family Study was to investigate individual differences in response to a standardized endurance exercise program, the role of familial aggregation, and the genetics of response levels of cardiorespiratory fitness and cardiovascular disease and diabetes risk factors. Here we summarize the findings and their potential implications for cardiometabolic health and cardiorespiratory fitness. It begins with overviews of background and planning, recruitment, testing and exercise program protocol, quality control measures, and other relevant organizational issues. A summary of findings is then provided on cardiorespiratory fitness, exercise hemodynamics, insulin and glucose metabolism, lipid and lipoprotein profiles, adiposity and abdominal visceral fat, blood levels of steroids and other hormones, markers of oxidative stress, skeletal muscle morphology and metabolic indicators, and resting metabolic rate. These summaries document the extent of the individual differences in response to a standardized and fully monitored endurance exercise program and document the importance of familial aggregation and heritability level for exercise response traits. Findings from genomic markers, muscle gene expression studies, and proteomic and metabolomics explorations are reviewed, along with lessons learned from a bioinformatics-driven analysis pipeline. The new opportunities being pursued in integrative -omics and physiology have extended considerably the expected life of HERITAGE and are being discussed in relation to the original conceptual model of the study.
Collapse
Affiliation(s)
- MARK A. SARZYNSKI
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC
| | - TREVA K. RICE
- Division of Biostatistics, Washington University in St. Louis School of Medicine, St. Louis, MO
| | - JEAN-PIERRE DESPRÉS
- Department of Kinesiology, Faculty of Medicine, Laval University, Quebec, QC, CANADA
- Quebec Heart and Lung Institute Research Center, Laval University, Québec, QC, CANADA
| | - LOUIS PÉRUSSE
- Department of Kinesiology, Faculty of Medicine, Laval University, Quebec, QC, CANADA
- Institute of Nutrition and Functional Foods (INAF), Laval University, Quebec, QC, CANADA
| | - ANGELO TREMBLAY
- Department of Kinesiology, Faculty of Medicine, Laval University, Quebec, QC, CANADA
- Institute of Nutrition and Functional Foods (INAF), Laval University, Quebec, QC, CANADA
| | - PHILIP R. STANFORTH
- Department of Kinesiology and Health Education, University of Texas at Austin, Austin, TX
| | - ANDRÉ TCHERNOF
- Quebec Heart and Lung Institute Research Center, Laval University, Québec, QC, CANADA
- School of Nutrition, Laval University, Quebec, QC, CANADA
| | - JACOB L. BARBER
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC
| | - FRANCESCO FALCIANI
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UNITED KINGDOM
| | - CLARY CLISH
- Metabolomics Platform, Broad Institute and Harvard Medical School, Boston, MA
| | - JEREMY M. ROBBINS
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
- Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, MA
| | - SUJOY GHOSH
- Cardiovascular and Metabolic Disorders Program and Centre for Computational Biology, Duke-National University of Singapore Medical School, SINGAPORE
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | - ROBERT E. GERSZTEN
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
- Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, MA
| | - ARTHUR S. LEON
- School of Kinesiology, University of Minnesota, Minneapolis, MN
| | | | - D. C. RAO
- Division of Biostatistics, Washington University in St. Louis School of Medicine, St. Louis, MO
| | - CLAUDE BOUCHARD
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| |
Collapse
|
19
|
Liu X, Bai Y, Cui R, He S, Ling Y, Wu C, Fang M. Integrated Analysis of the ceRNA Network and M-7474 Function in Testosterone-Mediated Fat Deposition in Pigs. Genes (Basel) 2022; 13:genes13040668. [PMID: 35456474 PMCID: PMC9032878 DOI: 10.3390/genes13040668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/30/2022] [Accepted: 04/06/2022] [Indexed: 02/01/2023] Open
Abstract
Castration can significantly enhance fat deposition in pigs, and the molecular mechanism of fat deposition caused by castration and its influence on fat deposition in different parts of pigs remain unclear. RNA-seq was performed on adipose tissue from different parts of castrated and intact Yorkshire pigs. Different ceRNA networks were constructed for different fat parts. GO and KEGG pathway annotations suggested that testosterone elevates cell migration and affects differentiation and apoptosis in back fat, while it predisposes animals to glycolipid metabolism disorders and increases the expression of inflammatory cytokines in abdominal fat. The interaction between M-7474, novel_miR_243 and SGK1 was verified by dual fluorescence experiments. This ceRNA relationship has also been demonstrated in porcine preadipocytes. Overexpression of M-7474 significantly inhibited the differentiation of preadipocytes compared to the control group. When 100 nM testosterone was added during preadipocyte differentiation, the expression of M-7474 was increased, and preadipocyte differentiation was significantly inhibited. Testosterone can affect preadipocyte differentiation by upregulating the expression of M-7474, sponging novel-miR-243, and regulating the expression of genes such as SGK1. At the same time, HSD11B1 and SLC2A4 may also be regulated by the corresponding lncRNA and miRNA, which ultimately affects glucose uptake by adipocytes and leads to obesity.
Collapse
Affiliation(s)
- Ximing Liu
- National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (X.L.); (R.C.); (S.H.); (Y.L.); (C.W.)
| | - Ying Bai
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan 056021, China;
| | - Ran Cui
- National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (X.L.); (R.C.); (S.H.); (Y.L.); (C.W.)
| | - Shuaihan He
- National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (X.L.); (R.C.); (S.H.); (Y.L.); (C.W.)
| | - Yao Ling
- National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (X.L.); (R.C.); (S.H.); (Y.L.); (C.W.)
| | - Changxin Wu
- National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (X.L.); (R.C.); (S.H.); (Y.L.); (C.W.)
| | - Meiying Fang
- National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (X.L.); (R.C.); (S.H.); (Y.L.); (C.W.)
- Sanya Institute of China Agricultural University, Sanya 572025, China
- Correspondence: ; Tel.: +86-10-62734943; Fax: +86-10-62734943
| |
Collapse
|
20
|
Schuh K, Häussler S, Sadri H, Prehn C, Lintelmann J, Adamski J, Koch C, Frieten D, Ghaffari MH, Dusel G, Sauerwein H. Blood and adipose tissue steroid metabolomics and mRNA expression of steroidogenic enzymes in periparturient dairy cows differing in body condition. Sci Rep 2022; 12:2297. [PMID: 35145150 PMCID: PMC8831572 DOI: 10.1038/s41598-022-06014-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 01/20/2022] [Indexed: 12/29/2022] Open
Abstract
In high-yielding dairy cows, the rapidly increasing milk production after parturition can result in a negative nutrient balance, since feed intake is insufficient to cover the needs for lactation. Mobilizing body reserves, mainly adipose tissue (AT), might affect steroid metabolism. We hypothesized, that cows differing in the extent of periparturient lipomobilization, will have divergent steroid profiles measured in serum and subcutaneous (sc)AT by a targeted metabolomics approach and steroidogenic enzyme profiles in scAT and liver. Fifteen weeks antepartum, 38 multiparous Holstein cows were allocated to a high (HBCS) or normal body condition (NBCS) group fed differently until week 7 antepartum to either increase (HBCS BCS: 3.8 ± 0.1 and BFT: 2.0 ± 0.1 cm; mean ± SEM) or maintain BCS (NBCS BCS: 3.0 ± 0.1 and BFT: 0.9 ± 0.1 cm). Blood samples, liver, and scAT biopsies were collected at week -7, 1, 3, and 12 relative to parturition. Greater serum concentrations of progesterone, androsterone, and aldosterone in HBCS compared to NBCS cows after parturition, might be attributed to the increased mobilization of AT. Greater glucocorticoid concentrations in scAT after parturition in NBCS cows might either influence local lipogenesis by differentiation of preadipocytes into mature adipocytes and/or inflammatory response.
Collapse
Affiliation(s)
- K Schuh
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115, Bonn, Germany
- Department of Life Sciences and Engineering, Animal Nutrition and Hygiene Unit, University of Applied Sciences Bingen, 55411, Bingen am Rhein, Germany
| | - S Häussler
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115, Bonn, Germany.
| | - H Sadri
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, 5166616471, Tabriz, Iran
| | - C Prehn
- Helmholtz Zentrum München, German Research Center for Environmental Health, Metabolomics and Proteomics Core, 85764, Neuherberg, Germany
| | - J Lintelmann
- Helmholtz Zentrum München, German Research Center for Environmental Health, Metabolomics and Proteomics Core, 85764, Neuherberg, Germany
| | - J Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - C Koch
- Educational and Research Centre for Animal Husbandry, Hofgut Neumuehle, 67728, Muenchweiler an der Alsenz, Germany
| | - D Frieten
- Thünen Institute of Organic Farming, 23847, Westerau, Germany
| | - M H Ghaffari
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115, Bonn, Germany
| | - G Dusel
- Department of Life Sciences and Engineering, Animal Nutrition and Hygiene Unit, University of Applied Sciences Bingen, 55411, Bingen am Rhein, Germany
| | - H Sauerwein
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115, Bonn, Germany
| |
Collapse
|
21
|
Wagner IV, Savchuk I, Sahlin L, Kulle A, Klöting N, Dietrich A, Holterhus PM, Dötsch J, Blüher M, Söder O. De Novo and Depot-Specific Androgen Production in Human Adipose Tissue: A Source of Hyperandrogenism in Women with Obesity. Obes Facts 2022; 15:281-291. [PMID: 34983051 PMCID: PMC9021649 DOI: 10.1159/000521571] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/15/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Obesity in women is often associated with hyperandrogenism, but the role of adipose tissue (AT) in androgen synthesis remains unclear. Therefore, we studied whether AT could be a source of androgens promoting hyperandrogenism. METHODS Subcutaneous and visceral (visc) AT was collected from lean and obese women. Androgen levels were evaluated in serum, AT, and cell-culture supernatant. Gene and protein expression of steroidogenic enzymes were determined. RESULTS Obese subjects had elevated serum androgen levels, which reduced after weight loss. Androgens were measurable in AT and in cell-culture supernatants of adipocytes. Steroids were higher in AT from obese women, with the highest difference for testosterone in visc AT (+7.9-fold, p = 0.032). Steroidogenic enzymes were expressed in human AT with depot-specific differences. Obese women showed a significantly higher expression of genes of the backdoor pathway and of CYP19 in visc AT. CONCLUSION The whole steroidogenic machinery of the classical and backdoor pathways of steroidogenesis, and the capacity for androgen biosynthesis, were found in both AT depots and cultured adipocytes. Therefore, we hypothesize that AT is a de novo site of androgen production and the backdoor pathway of steroidogenesis might be a new pathomechanism for hyperandrogenism in women with obesity.
Collapse
Affiliation(s)
- Isabel Viola Wagner
- Pediatric Endocrinology Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatrics, Medical Faculty, University of Cologne, Cologne, Germany
- Department of Pediatrics, Medical Faculty, UKSH, University of Lübeck, Lübeck, Germany
| | - Iuliia Savchuk
- Pediatric Endocrinology Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Lena Sahlin
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Alexandra Kulle
- Hormone Center for Pediatric Endocrinology Lab, University Hospital Kiel, Kiel, Germany
| | - Nora Klöting
- Department of Medicine and Department of Surgery, Integrated Research and Treatment Center (IFB Adiposity Diseases), University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum Muenchen at the University of Leipzig, Leipzig, Germany
| | - Arne Dietrich
- Department of Medicine and Department of Surgery, Integrated Research and Treatment Center (IFB Adiposity Diseases), University of Leipzig, Leipzig, Germany
| | - Paul-Martin Holterhus
- Hormone Center for Pediatric Endocrinology Lab, University Hospital Kiel, Kiel, Germany
| | - Jörg Dötsch
- Department of Pediatrics, Medical Faculty, University of Cologne, Cologne, Germany
| | - Matthias Blüher
- Department of Medicine and Department of Surgery, Integrated Research and Treatment Center (IFB Adiposity Diseases), University of Leipzig, Leipzig, Germany
| | - Olle Söder
- Pediatric Endocrinology Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
22
|
Overview of human 20 alpha-hydroxysteroid dehydrogenase (AKR1C1): Functions, regulation, and structural insights of inhibitors. Chem Biol Interact 2021; 351:109746. [PMID: 34780792 DOI: 10.1016/j.cbi.2021.109746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/28/2021] [Accepted: 11/10/2021] [Indexed: 11/22/2022]
Abstract
Human aldo-keto reductase family 1C1 (AKR1C1) is an important enzyme involved in human hormone metabolism, which is mainly responsible for the metabolism of progesterone in the human body. AKR1C1 is highly expressed and has an important relationship with the occurrence and development of various diseases, especially some cancers related to hormone metabolism. Nowadays, many inhibitors against AKR1C1 have been discovered, including some synthetic compounds and natural products, which have certain inhibitory activity against AKR1C1 at the target level. Here we briefly reviewed the physiological and pathological functions of AKR1C1 and the relationship with the disease, and then summarized the development of AKR1C1 inhibitors, elucidated the interaction between inhibitors and AKR1C1 through molecular docking results and existing co-crystal structures. Finally, we discussed the design ideals of selective AKR1C1 inhibitors from the perspective of AKR1C1 structure, discussed the prospects of AKR1C1 in the treatment of human diseases in terms of biomarkers, pre-receptor regulation and single nucleotide polymorphisms, aiming to provide new ideas for drug research targeting AKR1C1.
Collapse
|
23
|
Katzer K, Hill JL, McIver KB, Foster MT. Lipedema and the Potential Role of Estrogen in Excessive Adipose Tissue Accumulation. Int J Mol Sci 2021; 22:ijms222111720. [PMID: 34769153 PMCID: PMC8583809 DOI: 10.3390/ijms222111720] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 12/26/2022] Open
Abstract
Lipedema is a painful fat disorder that affects ~11% of the female population. It is characterized by bilateral, disproportionate accumulation of subcutaneous adipose tissue predominantly in the lower body. The onset of lipedema pathophysiology is thought to occur during periods of hormonal fluctuation, such as puberty, pregnancy, or menopause. Although the identification and characterization of lipedema have improved, the underlying disease etiology remains to be elucidated. Estrogen, a key regulator of adipocyte lipid and glucose metabolism, and female-associated body fat distribution are postulated to play a contributory role in the pathophysiology of lipedema. Dysregulation of adipose tissue accumulation via estrogen signaling likely occurs by two mechanisms: (1). altered adipocyte estrogen receptor distribution (ERα/ERß ratio) and subsequent metabolic signaling and/or (2). increased release of adipocyte-produced steroidogenic enzymes leading to increased paracrine estrogen release. These alterations could result in increased activation of peroxisome proliferator-activated receptor γ (PPARγ), free fatty acid entry into adipocytes, glucose uptake, and angiogenesis while decreasing lipolysis, mitochondriogenesis, and mitochondrial function. Together, these metabolic alterations would lead to increased adipogenesis and adipocyte lipid deposition, resulting in increased adipose depot mass. This review summarizes research characterizing estrogen-mediated adipose tissue metabolism and its possible relation to excessive adipose tissue accumulation associated with lipedema.
Collapse
|
24
|
Pott J, Horn K, Zeidler R, Kirsten H, Ahnert P, Kratzsch J, Loeffler M, Isermann B, Ceglarek U, Scholz M. Sex-Specific Causal Relations between Steroid Hormones and Obesity-A Mendelian Randomization Study. Metabolites 2021; 11:738. [PMID: 34822396 PMCID: PMC8624973 DOI: 10.3390/metabo11110738] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 12/15/2022] Open
Abstract
Steroid hormones act as important regulators of physiological processes including gene expression. They provide possible mechanistic explanations of observed sex-dimorphisms in obesity and coronary artery disease (CAD). Here, we aim to unravel causal relationships between steroid hormones, obesity, and CAD in a sex-specific manner. In genome-wide meta-analyses of four steroid hormone levels and one hormone ratio, we identified 17 genome-wide significant loci of which 11 were novel. Among loci, seven were female-specific, four male-specific, and one was sex-related (stronger effects in females). As one of the loci was the human leukocyte antigen (HLA) region, we analyzed HLA allele counts and found four HLA subtypes linked to 17-OH-progesterone (17-OHP), including HLA-B*14*02. Using Mendelian randomization approaches with four additional hormones as exposure, we detected causal effects of dehydroepiandrosterone sulfate (DHEA-S) and 17-OHP on body mass index (BMI) and waist-to-hip ratio (WHR). The DHEA-S effect was stronger in males. Additionally, we observed the causal effects of testosterone, estradiol, and their ratio on WHR. By mediation analysis, we found a direct sex-unspecific effect of 17-OHP on CAD while the other four hormone effects on CAD were mediated by BMI or WHR. In conclusion, we identified the sex-specific causal networks of steroid hormones, obesity-related traits, and CAD.
Collapse
Affiliation(s)
- Janne Pott
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, 04107 Leipzig, Germany; (K.H.); (H.K.); (P.A.); (M.L.)
- LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; (J.K.); (B.I.); (U.C.)
| | - Katrin Horn
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, 04107 Leipzig, Germany; (K.H.); (H.K.); (P.A.); (M.L.)
- LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; (J.K.); (B.I.); (U.C.)
| | - Robert Zeidler
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, 04103 Leipzig, Germany;
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, 04107 Leipzig, Germany; (K.H.); (H.K.); (P.A.); (M.L.)
- LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; (J.K.); (B.I.); (U.C.)
| | - Peter Ahnert
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, 04107 Leipzig, Germany; (K.H.); (H.K.); (P.A.); (M.L.)
- LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; (J.K.); (B.I.); (U.C.)
| | - Jürgen Kratzsch
- LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; (J.K.); (B.I.); (U.C.)
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, 04103 Leipzig, Germany;
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, 04107 Leipzig, Germany; (K.H.); (H.K.); (P.A.); (M.L.)
- LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; (J.K.); (B.I.); (U.C.)
| | - Berend Isermann
- LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; (J.K.); (B.I.); (U.C.)
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, 04103 Leipzig, Germany;
| | - Uta Ceglarek
- LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; (J.K.); (B.I.); (U.C.)
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, 04103 Leipzig, Germany;
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, 04107 Leipzig, Germany; (K.H.); (H.K.); (P.A.); (M.L.)
- LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; (J.K.); (B.I.); (U.C.)
| |
Collapse
|
25
|
Kothmann KH, Jacobsen V, Laffitte E, Bromfield C, Grizzaffi M, Jarboe M, Braundmeier-Fleming AG, Bahr JM, Nowak RA, Newell-Fugate AE. Virilizing doses of testosterone decrease circulating insulin levels and differentially regulate insulin signaling in liver and adipose tissue of females. Am J Physiol Endocrinol Metab 2021; 320:E1107-E1118. [PMID: 33900852 PMCID: PMC8285596 DOI: 10.1152/ajpendo.00281.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Transgender men undergoing hormone therapy are at risk for insulin resistance. However, how virilizing testosterone therapy affects serum insulin and peripheral insulin sensitivity in transgender men is unknown. This study assessed the effect of acute, virilizing testosterone on serum insulin concentrations and insulin signaling in liver, skeletal muscle, and white adipose tissue (WAT) of female pigs as a translational model for transgender men. Females received three doses of intramuscular testosterone cypionate (TEST females; 50 mg/day/pig) or corn oil (control) spaced 6 days apart starting on the day of estrus (D0). Fasting blood was collected on D0, D3, D5, D11, and D13, and females were euthanized on D13. On D13, TEST females had virilizing concentrations of serum testosterone with normal concentrations of serum estradiol. Virilizing serum testosterone concentrations (D13) were associated with decreased serum insulin and C-peptide concentrations. Blood glucose and serum glycerol concentrations were not altered by testosterone. Virilizing concentrations of testosterone downregulated AR and ESR1 in subcutaneous (sc) WAT and upregulated transcript levels of insulin-signaling pathway components in WAT and liver. At the protein level, virilizing testosterone concentrations were associated with increased PI3K 110α in liver and increased insulin receptor (INSR) and phospho(Ser256)-FOXO1 in visceral (v) WAT but decreased phospho(Ser473)-AKT in vWAT and scWAT. These results suggest that acute exposure to virilizing concentrations of testosterone suppresses circulating insulin levels and results in increased abundance of proteins in the insulin-signaling pathway in liver and altered phosphorylation of key proteins in control of insulin sensitivity in WAT.NEW & NOTEWORTHY Acute virilizing doses of testosterone administered to females suppress circulating insulin levels, upregulate components of the insulin-signaling pathway in liver, and suppress insulin signaling in white adipose tissue. These results suggest that insulin resistance in transgender men may be due to suppression of the insulin-signaling pathway and decreased insulin sensitivity in white adipose tissue.
Collapse
Affiliation(s)
- Kadden H Kothmann
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Victoria Jacobsen
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Emily Laffitte
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Corinne Bromfield
- Agricultural Animal Care and Use Program, Office of the Vice Chancellor for Research, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Matthew Grizzaffi
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Monica Jarboe
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois
| | - Andrea G Braundmeier-Fleming
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois
| | - Janice M Bahr
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Romana A Nowak
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Annie E Newell-Fugate
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| |
Collapse
|
26
|
Nigro P, Middelbeek RJW, Alves CRR, Rovira-Llopis S, Ramachandran K, Rowland LA, Møller AB, Takahashi H, Alves-Wagner AB, Vamvini M, Makarewicz NS, Albertson BG, Hirshman MF, Goodyear LJ. Exercise Training Promotes Sex-Specific Adaptations in Mouse Inguinal White Adipose Tissue. Diabetes 2021; 70:1250-1264. [PMID: 33563587 PMCID: PMC8275891 DOI: 10.2337/db20-0790] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022]
Abstract
Recent studies demonstrate that adaptations to white adipose tissue (WAT) are important components of the beneficial effects of exercise training on metabolic health. Exercise training favorably alters the phenotype of subcutaneous inguinal WAT (iWAT) in male mice, including decreasing fat mass, improving mitochondrial function, inducing beiging, and stimulating the secretion of adipokines. In this study, we find that despite performing more voluntary wheel running compared with males, these adaptations do not occur in the iWAT of female mice. Consistent with sex-specific adaptations, we report that mRNA expression of androgen receptor coactivators is upregulated in iWAT from trained male mice and that testosterone treatment of primary adipocytes derived from the iWAT of male, but not female mice, phenocopies exercise-induced metabolic adaptations. Sex specificity also occurs in the secretome profile, as we identify cysteine-rich secretory protein 1 (Crisp1) as a novel adipokine that is only secreted from male iWAT in response to exercise. Crisp1 expression is upregulated by testosterone and functions to increase glucose and fatty acid uptake. Our finding that adaptations to iWAT with exercise training are dramatically greater in male mice has potential clinical implications for understanding the different metabolic response to exercise training in males and females and demonstrates the importance of investigating both sexes in studies of adipose tissue biology.
Collapse
Affiliation(s)
- Pasquale Nigro
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Roeland J W Middelbeek
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Christiano R R Alves
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Susana Rovira-Llopis
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Krithika Ramachandran
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Leslie A Rowland
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Andreas B Møller
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Hirokazu Takahashi
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Ana B Alves-Wagner
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Maria Vamvini
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Nathan S Makarewicz
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Brent G Albertson
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Michael F Hirshman
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Laurie J Goodyear
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
27
|
Ostinelli G, Vijay J, Vohl MC, Grundberg E, Tchernof A. AKR1C2 and AKR1C3 expression in adipose tissue: Association with body fat distribution and regulatory variants. Mol Cell Endocrinol 2021; 527:111220. [PMID: 33675863 PMCID: PMC8052191 DOI: 10.1016/j.mce.2021.111220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Changes in androgen dynamics within adipose tissue have been proposed as modulators of body fat accumulation. In this context, AKR1C2 likely plays a significant role by inactivating 5α-dihydrotestosterone. AIM To characterize AKR1C2 expression patterns across adipose depots and cell populations and to provide insight into the link with body fat distribution and genetic regulation. METHODS We used RNA sequencing data from severely obese patients to assess patterns of AKR1C2 and AKR1C3 expression in abdominal adipose tissue depots and cell fractions. We additionally used data from 856 women to assess AKR1C2 heritability and to link its expression in adipose tissue with body fat distribution. Further, we used public resources to study AKR1C2 genetic regulation as well as reference epigenome data for regulatory element profiling and functional interpretation of genetic data. RESULTS We found that mature adipocytes and adipocyte-committed adipocyte progenitor cells (APCs) had enriched expression of AKR1C2. We found adipose tissue AKR1C2 and AKR1C3 expression to be significantly and positively associated with percentage trunk fat mass in women. We identified strong genetic regulation of AKR1C2 by rs28571848 and rs34477787 located on the binding sites of two nuclear transcription factors, namely retinoid acid-related orphan receptor alpha and the glucocorticoid receptor. CONCLUSION We confirm the link between AKR1C2, adipogenic differentiation and adipose tissue distribution. We provide insight into genetic regulation of AKR1C2 by identifying regulatory variants mapping to binding sites for the glucocorticoid receptor and retinoid acid-related orphan receptor alpha which may in part mediate the effect of AKR1C2 expression on body fat distribution.
Collapse
Affiliation(s)
- Giada Ostinelli
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec-Université Laval, 2725 Chemin Sainte-Foy, G1V 4G5, Québec City, Québec, Canada; École de Nutrition, Université Laval, 2425 Rue de l'Agriculture, G1V 0A6, Québec City, Québec, Canada
| | - Jinchu Vijay
- Department of Human Genetics, McGill University, Montréal, Quebec, Canada
| | - Marie-Claude Vohl
- École de Nutrition, Université Laval, 2425 Rue de l'Agriculture, G1V 0A6, Québec City, Québec, Canada; Centre Nutrition, Santé et Societé (NUTRISS)-Insitut sur la Nutrition et les Aliments Fonctionnells (INAF), Université Laval, Québec City, Québec, Canada
| | - Elin Grundberg
- Department of Human Genetics, McGill University, Montréal, Quebec, Canada; Children's Mercy Research Institute, Children's Mercy Kansas City, Kansas City, MO, USA.
| | - Andre Tchernof
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec-Université Laval, 2725 Chemin Sainte-Foy, G1V 4G5, Québec City, Québec, Canada; École de Nutrition, Université Laval, 2425 Rue de l'Agriculture, G1V 0A6, Québec City, Québec, Canada.
| |
Collapse
|
28
|
Hetemäki N, Mikkola TS, Tikkanen MJ, Wang F, Hämäläinen E, Turpeinen U, Haanpää M, Vihma V, Savolainen-Peltonen H. Adipose tissue estrogen production and metabolism in premenopausal women. J Steroid Biochem Mol Biol 2021; 209:105849. [PMID: 33610799 DOI: 10.1016/j.jsbmb.2021.105849] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 02/03/2021] [Accepted: 02/07/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Although the ovaries produce the majority of estrogens in women before menopause, estrogen is also synthesized in peripheral tissues such as adipose tissue (AT). The typical female AT distribution, concentrated in subcutaneous and femoro-gluteal regions, is estrogen-mediated, but the significance of estrogen synthesis in AT of premenopausal women is poorly understood. DESIGN AND METHODS Serum and subcutaneous and visceral AT homogenates from 28 premenopausal women undergoing non-malignant surgery were analyzed for estrone, estradiol, and serum estrone sulfate (E1S) concentrations with liquid chromatography-tandem mass spectrometry. Isotopic precursors were used to measure enzyme activities of estrone-producing steroid sulfatase and estradiol-producing 17β-hydroxysteroid dehydrogenases (17β-HSD). Messenger RNA (mRNA) expression levels of genes for estrogen-metabolizing enzymes were analyzed using real-time reverse transcription quantitative polymerase chain reaction. RESULTS While estradiol was the predominant circulating active estrogen, estrone dominated in AT, with a higher concentration in visceral than subcutaneous AT (median, 2657 vs 1459 pmol/kg; P = 0.002). Both AT depots converted circulating E1S to estrone, and estrone to estradiol. Median levels of estrone were five to ten times higher in subcutaneous and visceral AT than in serum (P < 0.001) and the estradiol level in visceral AT was 1.3 times higher than in serum (P < 0.005). The local estrone concentration in visceral AT correlated positively with mRNA expression of estrone-producing enzyme aromatase (r = 0.65, P = 0.003). Waist circumference correlated positively with increased estradiol production in subcutaneous AT (r = 0.60, P = 0.039). CONCLUSIONS Premenopausal AT demonstrated high estrogenic enzyme activity and considerable local estrogen concentrations. This may be a factor promoting female-typical AT distribution in premenopausal women.
Collapse
Affiliation(s)
- Natalia Hetemäki
- Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, FIN-00029 HUS, Helsinki, Finland; Folkhälsan Research Center, University of Helsinki, FIN-00014, Helsinki, Finland
| | - Tomi S Mikkola
- Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, FIN-00029 HUS, Helsinki, Finland; Folkhälsan Research Center, University of Helsinki, FIN-00014, Helsinki, Finland
| | - Matti J Tikkanen
- Folkhälsan Research Center, University of Helsinki, FIN-00014, Helsinki, Finland; Heart and Lung Center, University of Helsinki and Helsinki University Hospital, FIN-00029 HUS, Helsinki, Finland
| | - Feng Wang
- Folkhälsan Research Center, University of Helsinki, FIN-00014, Helsinki, Finland
| | - Esa Hämäläinen
- Department of Clinical Chemistry, University of Helsinki, FIN-00029 HUS, Helsinki, Finland
| | - Ursula Turpeinen
- HUSLAB, Helsinki University Hospital, FIN-00029 HUS, Helsinki, Finland
| | - Mikko Haanpää
- HUSLAB, Helsinki University Hospital, FIN-00029 HUS, Helsinki, Finland
| | - Veera Vihma
- Folkhälsan Research Center, University of Helsinki, FIN-00014, Helsinki, Finland; Department of General Practice and Primary Health Care, University of Helsinki, FIN-00014, Helsinki, Finland
| | - Hanna Savolainen-Peltonen
- Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, FIN-00029 HUS, Helsinki, Finland; Folkhälsan Research Center, University of Helsinki, FIN-00014, Helsinki, Finland.
| |
Collapse
|
29
|
Kaikaew K, Grefhorst A, Visser JA. Sex Differences in Brown Adipose Tissue Function: Sex Hormones, Glucocorticoids, and Their Crosstalk. Front Endocrinol (Lausanne) 2021; 12:652444. [PMID: 33927694 PMCID: PMC8078866 DOI: 10.3389/fendo.2021.652444] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/23/2021] [Indexed: 12/12/2022] Open
Abstract
Excessive fat accumulation in the body causes overweight and obesity. To date, research has confirmed that there are two types of adipose tissue with opposing functions: lipid-storing white adipose tissue (WAT) and lipid-burning brown adipose tissue (BAT). After the rediscovery of the presence of metabolically active BAT in adults, BAT has received increasing attention especially since activation of BAT is considered a promising way to combat obesity and associated comorbidities. It has become clear that energy homeostasis differs between the sexes, which has a significant impact on the development of pathological conditions such as type 2 diabetes. Sex differences in BAT activity may contribute to this and, therefore, it is important to address the underlying mechanisms that contribute to sex differences in BAT activity. In this review, we discuss the role of sex hormones in the regulation of BAT activity under physiological and some pathological conditions. Given the increasing number of studies suggesting a crosstalk between sex hormones and the hypothalamic-pituitary-adrenal axis in metabolism, we also discuss this crosstalk in relation to sex differences in BAT activity.
Collapse
Affiliation(s)
- Kasiphak Kaikaew
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Aldo Grefhorst
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, Netherlands
| | - Jenny A. Visser
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- *Correspondence: Jenny A. Visser,
| |
Collapse
|
30
|
Ernst J, Gert K, Kraus FB, Rolle-Kampczyk UE, Wabitsch M, Dehghani F, Schaedlich K. Androstenedione changes steroidogenic activity of SGBS cells. Endocr Connect 2020; 9:587-598. [PMID: 32580160 PMCID: PMC7354720 DOI: 10.1530/ec-19-0549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 06/03/2020] [Indexed: 12/26/2022]
Abstract
The rapid increase of obesity during the last decades and its future prospects are alarming. Besides the general discussed causes of obesity, the 'Developmental Origins of Health and Disease' (DOHaD) hypothesis received more attention in recent years. This hypothesis postulates an adverse influence during early development that programs the unborn child for metabolic dysfunctions later in life. Childhood obesity - an as much increasing problem - can be predisposed by maternal overweight and diabetes. Both, obesity and hyperinsulinemia are major causes of female hyperandrogenemia. As predicted by the DOHaD hypothesis and shown in animal models, developmental androgen excess can lead to metabolic abnormalities in offspring. In this study, we investigated, if androgen exposure adversely affects the adipogenic differentiation of preadipocytes and the endocrine function of adult adipocytes. The human SGBS preadipocyte model was used to affirm the de novo biosynthesis of steroid hormones under normal adipogenesis conditions. Normal adipogenesis was paralleled by an increase of corticosteroids and androgens, whereas estrogen remained at a steady level. Treatment with androstenedione had no effect on SGBS proliferation and differentiation, but adult adipocytes exhibited a significant higher accumulation of triglycerides. Progesterone (up to 2-fold), testosterone (up to 38-fold) and cortisone (up to 1.4-fold) - but not cortisol - were elevated by androstenedione administration in adult adipocytes. Estrogen was not altered. Data suggest that androgen does not negatively influence adipogenic differentiation, but steroidogenic function of SGBS adipocytes.
Collapse
Affiliation(s)
- Jana Ernst
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse, Halle (Saale), Germany
- Correspondence should be addressed to J Ernst:
| | - Katharina Gert
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse, Halle (Saale), Germany
| | - Frank Bernhard Kraus
- Central Laboratory, University Hospital Halle (Saale), Ernst-Grube-Strasse, Halle (Saale), Germany
| | | | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Faramarz Dehghani
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse, Halle (Saale), Germany
| | - Kristina Schaedlich
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse, Halle (Saale), Germany
| |
Collapse
|
31
|
Laforest S, Pelletier M, Denver N, Poirier B, Nguyen S, Walker BR, Durocher F, Homer NZM, Diorio C, Andrew R, Tchernof A. Estrogens and Glucocorticoids in Mammary Adipose Tissue: Relationships with Body Mass Index and Breast Cancer Features. J Clin Endocrinol Metab 2020; 105:5680713. [PMID: 31853538 PMCID: PMC7065843 DOI: 10.1210/clinem/dgz268] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 12/17/2019] [Indexed: 12/16/2022]
Abstract
CONTEXT Adipose tissue is an important site for extragonadal steroid hormone biosynthesis through the expression and activity of P450 aromatase, 11β-hydroxysteroid dehydrogenase (HSD) 1, and 17β-HSDs. The contribution of steroid hormones produced by adjacent adipose tissue for the progression and survival of breast tumors is unknown. OBJECTIVE To quantify estrogens (estradiol, estrone) and glucocorticoids (cortisol, cortisone) in breast adipose tissue from both healthy and diseased women and their relationships with adiposity indices and breast cancer prognostic markers. DESIGN AND SETTING Breast adipose tissue was collected at time of surgery. PATIENTS Pre- and postmenopausal women undergoing partial mastectomy for treatment of breast cancer (n = 17) or reduction mammoplasty (n = 6) were studied. INTERVENTIONS Relative estrogen and glucocorticoid amounts were determined by liquid chromatography tandem mass spectrometry. RESULTS The targeted steroids were reliably detected and quantified in mammary adipose tissues. Women with ER+/PR+ tumor had higher relative estradiol amount than women with ER-/PR- tumor (P < .05). The ratio of estradiol-to-estrone was higher in lean women than in women with a body mass index (BMI) ≥ 25 kg/m2 (P < .05). Mixed-model analyses showed that estradiol, cortisone, and cortisol were negatively associated with tumor size (P < .05). Relationships between glucocorticoids and tumor size remained significant after adjustment for BMI. The cortisol-to-cortisone ratio was negatively associated with tumor stage (P < .05) independently of BMI. CONCLUSIONS We reliably quantified estrogens and glucocorticoids in breast adipose tissue from healthy women and women suffering from breast cancer. Our findings suggest that smaller breast tumors are associated with higher relative amounts of estradiol and cortisol in adipose tissue.
Collapse
Affiliation(s)
- Sofia Laforest
- CHU de Québec-Université Laval Research Center (Endocrinology and Nephrology division), School of Nutrition, Faculty of Agriculture and Food Sciences, Université Laval, Québec, Canada
- Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen’s Medical Research Institute, Edinburgh, UK
| | - Mélissa Pelletier
- CHU de Québec-Université Laval Research Center (Endocrinology and Nephrology division), School of Nutrition, Faculty of Agriculture and Food Sciences, Université Laval, Québec, Canada
- Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada
| | - Nina Denver
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen’s Medical Research Institute, Edinburgh, UK
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, University Avenue, Glasgow, UK
| | - Brigitte Poirier
- CHU de Québec-Université Laval Research Center (Oncology division), Université Laval Cancer Research Center and Department of Surgery, Faculty of Medicine, Université Laval, Québec, Canada
- Centre des maladies du sein Deschênes-Fabia, Hôpital Saint-Sacrement, Québec, Canada
| | - Sébastien Nguyen
- CHU de Québec-Université Laval Research Center (Oncology division), Université Laval Cancer Research Center and Department of Surgery, Faculty of Medicine, Université Laval, Québec, Canada
| | - Brian R Walker
- University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Francine Durocher
- CHU de Québec-Université Laval Research Center (Endocrinology and Nephrology division), Université Laval Cancer Research Center and Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, Canada
| | - Natalie Z M Homer
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen’s Medical Research Institute, Edinburgh, UK
- University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Caroline Diorio
- Centre des maladies du sein Deschênes-Fabia, Hôpital Saint-Sacrement, Québec, Canada
- CHU de Québec-Université Laval Research Center (Oncology division), Université Laval Cancer Research Center and Department of Social and Preventive Medicine, Faculty of Medicine, Université Laval, Québec, Canada
| | - Ruth Andrew
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen’s Medical Research Institute, Edinburgh, UK
- University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - André Tchernof
- CHU de Québec-Université Laval Research Center (Endocrinology and Nephrology division), School of Nutrition, Faculty of Agriculture and Food Sciences, Université Laval, Québec, Canada
- Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada
- Correspondence and Reprint Requests: André Tchernof, PhD, Institut universitaire de cardiologie et de pneumologie de Québec, 2725 Chemin Ste-Foy, Y4212, Québec, QC, Canada G1V 4G5. E-mail:
| |
Collapse
|
32
|
Jiang Y, Li Y, Cheng J, Ma J, Li Q, Pang T. Upregulation of AKR1C1 in mesenchymal stromal cells promotes the survival of acute myeloid leukaemia cells. Br J Haematol 2020; 189:694-706. [PMID: 31943135 DOI: 10.1111/bjh.16253] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/01/2019] [Indexed: 01/03/2023]
Abstract
The leukaemic bone marrow microenvironment, comprising abnormal mesenchymal stromal cells (MSCs), is responsible for the poor prognosis of acute myeloid leukaemia (AML). Therefore, it is essential to determine the mechanisms underlying the supportive role of MSCs in the survival of leukaemia cells. Through in silico analyses, we identified a total of 271 aberrantly expressed genes in the MSCs derived from acute myeloid leukemia (AML) patients that were associated with adipogenic differentiation, of which aldo-keto reductase 1C1 (AKR1C1) was significantly upregulated in the AML-MSCs. Knockdown of AKR1C1 in the MSCs suppressed adipogenesis and promoted osteogenesis, and inhibited the growth of co-cultured AML cell lines compared to the situation in wild- type AML-derived MSCs. Introduction of recombinant human AKR1C1 in the MSCs partially alleviated the effects of AKR1C1 knockdown. In addition, the absence of AKR1C1 reduced secretion of cytokines such as MCP-1, IL-6 and G-CSF from the MSCs, along with inactivation of STAT3 and ERK1/2 in the co-cultured AML cells. AKR1C1 is an essential factor driving the adipogenic differentiation of leukaemic MSCs and mediates its pro-survival effects on AML cells by promoting cytokine secretion and activating the downstream pathways in the AML cells.
Collapse
Affiliation(s)
- Yajing Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Ying Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jingying Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jiao Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Qinghua Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Tianxiang Pang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
33
|
Laforest S, Pelletier M, Denver N, Poirier B, Nguyen S, Walker BR, Durocher F, Homer NZM, Diorio C, Tchernof A, Andrew R. Simultaneous quantification of estrogens and glucocorticoids in human adipose tissue by liquid-chromatography-tandem mass spectrometry. J Steroid Biochem Mol Biol 2019; 195:105476. [PMID: 31561001 PMCID: PMC7099401 DOI: 10.1016/j.jsbmb.2019.105476] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/30/2019] [Accepted: 09/18/2019] [Indexed: 12/13/2022]
Abstract
The presence of estrogens, androgens and glucocorticoids as well as their receptors and steroid converting enzymes in adipose tissue has been established. Their contribution to diseases such as obesity, diabetes and hormone-dependent cancers is an active area of research. Our objective was to develop a LC-MS/MS method to quantify bioactive estrogens and glucocorticoids simultaneously in human adipose tissue. Estrogens and glucocorticoids were extracted from adipose tissue samples using solid-phase extraction. Estrogens were derivatized using 1-(2,4-dinitro-5-fluorophenyl)-4-methylpiperazine (PPZ) and methyl iodide to generate a permanently charged molecule (MPPZ). Steroids were separated and quantified by LC-MS/MS. The limit of quantitation for the steroids was between 15 and 100 pg per sample. Accuracy and precision were acceptable (<20%). Using this method, estradiol, estrone, cortisone and cortisol were quantified in adipose tissue from women with and without breast cancer. This novel assay of estrogens and glucocorticoids by LC-MS/MS coupled with derivatization allowed simultaneous quantification of a panel of steroids in human adipose tissue across the endogenous range of concentrations encountered in health and disease.
Collapse
Affiliation(s)
- Sofia Laforest
- CHU de Québec-Université Laval Research Center (Endocrinology and Nephrology Division), School of Nutrition, Faculty of Agriculture and Food Sciences, Université Laval, Québec, Canada; Quebec Heart Lung Institute, Québec, Canada
| | - Mélissa Pelletier
- CHU de Québec-Université Laval Research Center (Endocrinology and Nephrology Division), School of Nutrition, Faculty of Agriculture and Food Sciences, Université Laval, Québec, Canada; Quebec Heart Lung Institute, Québec, Canada
| | - Nina Denver
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research Institute, Edinburgh, United Kingdom; Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, University Avenue, Glasgow, United Kingdom; Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Cathedral Street, Glasgow, United Kingdom
| | - Brigitte Poirier
- CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center and Department of Surgery, Faculty of Medicine, Université Laval, Québec, Canada; Centre des maladies du sein Deschênes-Fabia, Hôpital Saint-Sacrement, Québec, Canada
| | - Sébastien Nguyen
- CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center and Department of Surgery, Faculty of Medicine, Université Laval, Québec, Canada
| | - Brian R Walker
- University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47, Little France Crescent, Edinburgh, EH16 4TJ, UK; Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Francine Durocher
- CHU de Québec-Université Laval Research Center (Endocrinology and Nephrology Division), Université Laval Cancer Research Center and Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, Canada
| | - Natalie Z M Homer
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research Institute, Edinburgh, United Kingdom; University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47, Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Caroline Diorio
- Centre des maladies du sein Deschênes-Fabia, Hôpital Saint-Sacrement, Québec, Canada; CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center and Department of Social and Preventive Medicine, Faculty of Medicine, Université Laval, Québec, Canada
| | - André Tchernof
- CHU de Québec-Université Laval Research Center (Endocrinology and Nephrology Division), School of Nutrition, Faculty of Agriculture and Food Sciences, Université Laval, Québec, Canada; Quebec Heart Lung Institute, Québec, Canada
| | - Ruth Andrew
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research Institute, Edinburgh, United Kingdom; University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47, Little France Crescent, Edinburgh, EH16 4TJ, UK.
| |
Collapse
|
34
|
Tomaselli G, Vallée M. Stress and drug abuse-related disorders: The promising therapeutic value of neurosteroids focus on pregnenolone-progesterone-allopregnanolone pathway. Front Neuroendocrinol 2019; 55:100789. [PMID: 31525393 DOI: 10.1016/j.yfrne.2019.100789] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/14/2019] [Accepted: 09/09/2019] [Indexed: 02/06/2023]
Abstract
The pregnenolone-progesterone-allopregnanolone pathway is receiving increasing attention in research on the role of neurosteroids in pathophysiology, particularly in stress-related and drug use disorders. These disorders involve an allostatic change that may result from deficiencies in allostasis or adaptive responses, and may be downregulated by adjustments in neurotransmission by neurosteroids. The following is an overview of findings that assess how pregnenolone and/or allopregnanolone concentrations are altered in animal models of stress and after consumption of alcohol or cannabis-type drugs, as well as in patients with depression, anxiety, post-traumatic stress disorder or psychosis and/or in those diagnosed with alcohol or cannabis use disorders. Preclinical and clinical evidence shows that pregnenolone and allopregnanolone, operating according to a different or common pharmacological profile involving GABAergic and/or endocannabinoid system, may be relevant biomarkers of psychiatric disorders for therapeutic purposes. Hence, ongoing clinical trials implicate synthetic analogs of pregnenolone or allopregnanolone, and also modulators of neurosteroidogenesis.
Collapse
Affiliation(s)
- Giovanni Tomaselli
- INSERM U1215, Neurocentre Magendie, Group "Physiopathology and Therapeutic Approaches of Stress-Related Disease", 146 Rue Léo Saignat, 33000 Bordeaux, France; University of Bordeaux, 33000 Bordeaux, France
| | - Monique Vallée
- INSERM U1215, Neurocentre Magendie, Group "Physiopathology and Therapeutic Approaches of Stress-Related Disease", 146 Rue Léo Saignat, 33000 Bordeaux, France; University of Bordeaux, 33000 Bordeaux, France.
| |
Collapse
|
35
|
Bourgeois R, Piché ME, Auclair A, Grenier-Larouche T, Mitchell PL, Poirier P, Biertho L, Marceau S, Hould FS, Biron S, Lebel S, Lescelleur O, Julien F, Martin J, Tchernof A, Mathieu P, Carpentier AC, Arsenault BJ. Acute and chronic effect of bariatric surgery on circulating autotaxin levels. Physiol Rep 2019; 7:e14004. [PMID: 30821134 PMCID: PMC6395307 DOI: 10.14814/phy2.14004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/09/2019] [Accepted: 01/11/2019] [Indexed: 01/02/2023] Open
Abstract
Autotaxin (ATX), an adipose tissue-derived lysophospholipase, has been involved in the pathophysiology of cardiometabolic diseases. The impact of bariatric surgery on circulating ATX levels is unknown. We examined the short- (24 h, 5 days) and longer-term (6 and 12 months) impact of bariatric surgery; as well as the short-term effect of caloric restriction (CR) on plasma ATX levels in patients with severe obesity. We measured ATX levels in 69 men and women (mean age: 41 ± 11 years, body mass index: 49.8 ± 7.1 kg/m2 ), before and after biliopancreatic diversion with duodenal switch surgery (BPD-DS) as well as in a control group (patients with severe obesity without surgery; n = 34). We also measured ATX levels in seven patients with severe obesity and type 2 diabetes who underwent a 3-day CR protocol before their BPD-DS. At baseline, ATX levels were positively associated with body mass index, fat mass, insulin resistance (HOMA-IR) as well as insulin and leptin levels and negatively with fat-free mass. ATX concentrations decreased 26.2% at 24 h after BPD-DS (342.9 ± 152.3 pg/mL to 253.2 ± 68.9 pg/mL, P < 0.0001) and by 16.4% at 12 months after BPD-DS (342.9 ± 152.3 pg/mL to 286.8 ± 182.6 pg/mL, P = 0.04). ATX concentrations were unchanged during follow-up in the control group (P = 0.4), and not influenced by short-term CR. In patients with severe obesity, bariatric surgery induced a rapid and sustained decrease in plasma ATX levels. Acute changes in ATX may not be explained by bariatric surgery-induced CR.
Collapse
Affiliation(s)
- Raphaëlle Bourgeois
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada
| | - Marie-Eve Piché
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada
| | - Audrey Auclair
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
| | - Thomas Grenier-Larouche
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
| | - Patricia L Mitchell
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
| | - Paul Poirier
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
- Faculty of Pharmacy, Université Laval, Québec, Canada
| | - Laurent Biertho
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
| | - Simon Marceau
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
| | - Frédéric-Simon Hould
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
| | - Simon Biron
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
| | - Stéfane Lebel
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
| | - Odette Lescelleur
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
| | - François Julien
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
| | - Julie Martin
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
| | - André Tchernof
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
- School of Nutrition, Université Laval, Québec, Canada
| | - Patrick Mathieu
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, Canada
| | - André C Carpentier
- Department of Medicine, Division of Endocrinology, Centre de recherche du CHUS, Université de Sherbrooke, Canada
| | - Benoit J Arsenault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada
| |
Collapse
|
36
|
Kvernebo Sunnergren K, Ankarberg-Lindgren C, Dahlgren J. Adrenal and Gonadal Activity, Androgen Concentrations, and Adult Height Outcomes in Boys With Silver-Russell Syndrome. Front Endocrinol (Lausanne) 2019; 10:829. [PMID: 31920957 PMCID: PMC6914679 DOI: 10.3389/fendo.2019.00829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 11/13/2019] [Indexed: 11/24/2022] Open
Abstract
Background: We have previously shown that adult height (AH) in males with Silver-Russell syndrome (SRS) correlated negatively with prepubertal estradiol concentrations. We aimed to identify the source of estradiol by analyzing androgen secretion profiles and measuring anti-Müllerian hormone (AMH) and inhibin B concentrations during childhood and puberty in this group of patients. Methods: In a retrospective longitudinal single-center study, 13 males with SRS were classified as non-responders (NRs = 8) or responders (Rs = 5), depending on the AH outcome. From 6 years of age, androgens were determined by mass spectrometry, and AMH, inhibin B and sex hormone-binding globulin concentrations were analyzed by immunoassays. Results: AH outcome correlated negatively with dehydroepiandrosterone-sulfate (DHEAS) at 8 (r = -0.72), 10 (r = -0.79), and 12 years (r = -0.72); testosterone at 10 (r = -0.94), 12 (r = -0.70) and 14 years (r = -0.64); dihydrotestosterone (DHT) at 10 (r = -0.62) and 12 years; (r = -0.57) and AMH at 12 years (r = 0.62) of age. Compared with Rs, NRs had higher median concentrations of DHEAS (μmol/L) at 10 years (2.9 vs. 1.0); androstenedione (nmol/L) at 10 (1.1 vs. 0.6) and 12 years (1.7 vs. 0.8); testosterone (nmol/L) at 10 (0.3 vs. 0.1), 12 (7.8 vs. 0.2) and 14 years (15.6 vs. 10.4); and DHT (pmol/L) at 10 (122 vs. 28) and 12 years (652 vs. 59) of age. AMH (ng/mL) was lower in NRs than in Rs at 12 years of age (11 vs. 50). No significant differences were observed in the inhibin B concentrations at any age. Conclusions: The elevated androgen concentrations before and during puberty, originated from both adrenal and gonadal secretion and correlated negatively with AH outcomes in males with SRS.
Collapse
Affiliation(s)
- Kjersti Kvernebo Sunnergren
- Göteborg Pediatric Growth Research Center (GP-GRC), Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Pediatrics, Ryhov County Hospital, Jönköping, Sweden
| | - Carina Ankarberg-Lindgren
- Göteborg Pediatric Growth Research Center (GP-GRC), Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jovanna Dahlgren
- Göteborg Pediatric Growth Research Center (GP-GRC), Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Endocrinology, Sahlgrenska University Hospital, Queen Silva Hospital, Gothenburg, Sweden
| |
Collapse
|
37
|
Vihma V, Heinonen S, Naukkarinen J, Kaprio J, Rissanen A, Turpeinen U, Hämäläinen E, Hakkarainen A, Lundbom J, Lundbom N, Mikkola TS, Tikkanen MJ, Pietiläinen KH. Increased body fat mass and androgen metabolism - A twin study in healthy young women. Steroids 2018; 140:24-31. [PMID: 30149073 DOI: 10.1016/j.steroids.2018.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/15/2018] [Accepted: 08/21/2018] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Obesity may alter serum steroid concentrations and metabolism. We investigated this in healthy young women with increased body fat and their leaner co-twin sisters. DESIGN Age and genetic background both strongly influence serum steroid levels and body composition. This is a cross-sectional study of 13 female monozygotic twin pairs (age, 23-36 years), ten of which were discordant for body mass index (median difference in body weight between the co-twins, 19 kg). METHODS We determined body composition by dual energy X-ray absorptiometry and magnetic resonance imaging, serum androgens by liquid chromatography-tandem mass spectrometry, and mRNA expression of genes in subcutaneous adipose tissue and adipocytes. RESULTS The heavier women had lower serum dehydroepiandrosterone (DHEA), dihydrotestosterone (DHT), and sex hormone-binding globulin (SHBG) (P < 0.05 for all) compared to their leaner co-twins with no differences in serum testosterone or androstenedione levels. Serum DHEA correlated inversely with %body fat (r = -0.905, P = 0.002), and DHT positively with SHBG (r = 0.842, P = 0.002). In adipose tissue or adipocytes, expressions of STS (steroid sulfatase) and androgen-related genes were significantly higher in the heavier compared to the leaner co-twin, and within pairs, correlated positively with adiposity but were not related to serum androgen levels. None of the serum androgen or SHBG levels correlated with indices of insulin resistance. CONCLUSIONS Serum DHEA levels were best predicted by %body fat, and serum DHT by SHBG. These or other serum androgen concentrations did not reflect differences in androgen-related genes in adipose tissue. General or intra-abdominal adiposity were not associated with increased androgenicity in young women.
Collapse
Affiliation(s)
- Veera Vihma
- University of Helsinki and Helsinki University Hospital, Heart and Lung Center, Biomedicum C315a, Haartmaninkatu 8, 00290 Helsinki, Finland; Folkhälsan Research Center, P.O. Box 63, 00014 University of Helsinki, Finland.
| | - Sini Heinonen
- University of Helsinki, Research Programs Unit, Diabetes and Obesity, Obesity Research Unit, P.O. Box 63, 00014 University of Helsinki, Finland
| | - Jussi Naukkarinen
- University of Helsinki, Research Programs Unit, Diabetes and Obesity, Obesity Research Unit, P.O. Box 63, 00014 University of Helsinki, Finland
| | - Jaakko Kaprio
- University of Helsinki, FIMM, Institute for Molecular Medicine Finland, and Department of Public Health, P.O. Box 20, 00014 University of Helsinki, Finland
| | - Aila Rissanen
- University of Helsinki, Research Programs Unit, Diabetes and Obesity, Obesity Research Unit, P.O. Box 63, 00014 University of Helsinki, Finland
| | - Ursula Turpeinen
- Helsinki University Hospital, HUSLAB, P.O. Box 720, 00029 HUS, Helsinki, Finland
| | - Esa Hämäläinen
- Helsinki University Hospital, HUSLAB, P.O. Box 720, 00029 HUS, Helsinki, Finland
| | - Antti Hakkarainen
- University of Helsinki and HUS Medical Imaging Center, Helsinki University Hospital, Haartmaninkatu 4, 00290 Helsinki, Finland
| | - Jesper Lundbom
- University of Helsinki and HUS Medical Imaging Center, Helsinki University Hospital, Haartmaninkatu 4, 00290 Helsinki, Finland
| | - Nina Lundbom
- University of Helsinki and HUS Medical Imaging Center, Helsinki University Hospital, Haartmaninkatu 4, 00290 Helsinki, Finland
| | - Tomi S Mikkola
- Folkhälsan Research Center, P.O. Box 63, 00014 University of Helsinki, Finland; Helsinki University Hospital, Obstetrics and Gynecology, P.O. Box 140, 00029 HUS, Helsinki, Finland
| | - Matti J Tikkanen
- University of Helsinki and Helsinki University Hospital, Heart and Lung Center, Biomedicum C315a, Haartmaninkatu 8, 00290 Helsinki, Finland; Folkhälsan Research Center, P.O. Box 63, 00014 University of Helsinki, Finland
| | - Kirsi H Pietiläinen
- University of Helsinki, Research Programs Unit, Diabetes and Obesity, Obesity Research Unit, P.O. Box 63, 00014 University of Helsinki, Finland; Helsinki University Hospital, Endocrinology, Abdominal Center, P.O. Box 340, 00029 HUS, Helsinki, Finland
| |
Collapse
|
38
|
Tchernof A, Brochu D, Maltais‐Payette I, Mansour MF, Marchand GB, Carreau A, Kapeluto J. Androgens and the Regulation of Adiposity and Body Fat Distribution in Humans. Compr Physiol 2018; 8:1253-1290. [DOI: 10.1002/cphy.c170009] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
39
|
Schiffer L, Arlt W, Storbeck KH. Intracrine androgen biosynthesis, metabolism and action revisited. Mol Cell Endocrinol 2018; 465:4-26. [PMID: 28865807 PMCID: PMC6565845 DOI: 10.1016/j.mce.2017.08.016] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 08/28/2017] [Accepted: 08/28/2017] [Indexed: 12/19/2022]
Abstract
Androgens play an important role in metabolic homeostasis and reproductive health in both men and women. Androgen signalling is dependent on androgen receptor activation, mostly by testosterone and 5α-dihydrotestosterone. However, the intracellular or intracrine activation of C19 androgen precursors to active androgens in peripheral target tissues of androgen action is of equal importance. Intracrine androgen synthesis is often not reflected by circulating androgens but rather by androgen metabolites and conjugates. In this review we provide an overview of human C19 steroid biosynthesis including the production of 11-oxygenated androgens, their transport in circulation and uptake into peripheral tissues. We conceptualise the mechanisms of intracrinology and review the intracrine pathways of activation and inactivation in selected human tissues. The contribution of liver and kidney as organs driving androgen inactivation and renal excretion are also highlighted. Finally, the importance of quantifying androgen metabolites and conjugates to assess intracrine androgen production is discussed.
Collapse
Affiliation(s)
- Lina Schiffer
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| | - Karl-Heinz Storbeck
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; Department of Biochemistry, Stellenbosch University, Stellenbosch 7600, South Africa
| |
Collapse
|
40
|
Kvernebo-Sunnergren K, Ankarberg-Lindgren C, Åkesson K, Andersson MX, Samuelsson L, Lovmar L, Dahlgren J. Hyperestrogenism Affects Adult Height Outcome in Growth Hormone Treated Boys With Silver-Russell Syndrome. Front Endocrinol (Lausanne) 2018; 9:780. [PMID: 30622515 PMCID: PMC6308318 DOI: 10.3389/fendo.2018.00780] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 12/11/2018] [Indexed: 11/29/2022] Open
Abstract
Background: Intrauterine growth retardation and short stature are common features in Silver-Russell syndrome (SRS). Despite recombinant growth hormone (rGH) treatment, poor pubertal height gain, affecting adult height (AH), is common. This study investigated whether growth patterns and estrogen concentrations are associated with AH outcome in rGH treated SRS males. Methods: In this retrospective longitudinal single-center study, 11 males with SRS were classified as non-responders (NR = 6) or responders (R = 5), depending on AH adjusted for midparental height. Epigenetic analysis and longitudinal growth measures, including bone age, rGH related parameters, pubertal development, gonadotropins and estrogen concentrations, were analyzed until AH. Results: Pubarche before 9 years was only observed in one NR. At 10 years of age, there was no difference in gonadotropins between NR and R. However, estradiol (E2) concentrations at 10 years of age showed a strong association to AH adjusted for MPH (r = -0.78, p < 0.001). Serum E2 (pmol/L) was significantly higher in NR at ages 10 years [median (range) 2 (<2-5) vs. <2 (<2)], 12 years [23 (10-57) vs. 2 (<2-2)] and 14 years [77 (54-87) vs. 24 (<2-38)] but not at 16 years. Birth weight standard deviation score (SDS) was lower in NR [-4.1 (-4.7 to -2.1) vs. -2.7 (-3.3 to -1.7)]. Weight gain (SDS) until pubertal onset was greater in NR [2.4 (1.4-3.5) vs. 0.8 (-0.4 to 1.7)] and pubertal height gain (SDS) was lower in NR [-1.0 (-2.7-0.4) vs. 0.1 (-0.1 to 1.1)]. At AH, a number of NR and R had high E2 concentrations and small testes. Conclusion: Increased E2 concentrations at age 10, 12, and 14 years were associated to less pubertal height gain, thus affecting AH. Due to the small number of patients, the results need to be confirmed in larger cohorts. The finding of impaired testicular development stresses the need of hormonal evaluation as a complement to clinical and radiological assessment when predicting AH in males with SRS.
Collapse
Affiliation(s)
- Kjersti Kvernebo-Sunnergren
- Department of Pediatrics, Ryhov County Hospital, Jönköping, Sweden
- Department of Pediatrics, Göteborg Pediatric Growth Research Center, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- *Correspondence: Kjersti Kvernebo-Sunnergren
| | - Carina Ankarberg-Lindgren
- Department of Pediatrics, Göteborg Pediatric Growth Research Center, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karin Åkesson
- Department of Pediatrics, Ryhov County Hospital, Jönköping, Sweden
- Division of Pediatrics, Department of Clinical and Experimental Medicine, Linköping University Hospital, Linköping, Sweden
| | - Mats X. Andersson
- Department of Pediatrics, Göteborg Pediatric Growth Research Center, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Lena Samuelsson
- Department of Clinical Pathology and Genetics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lovisa Lovmar
- Department of Clinical Pathology and Genetics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jovanna Dahlgren
- Department of Pediatrics, Göteborg Pediatric Growth Research Center, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
41
|
Galligan TM, Schwacke LH, McFee WE, Boggs ASP. Evidence for cortisol-cortisone metabolism by marine mammal blubber. MARINE BIOLOGY 2018; 165:10.1007/s00227-018-3373-4. [PMID: 31579267 PMCID: PMC6774200 DOI: 10.1007/s00227-018-3373-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/11/2018] [Indexed: 05/06/2023]
Abstract
Blubber, a specialized hyperdermic adipose tissue found in marine mammals, has been identified as a useful tissue for the assessment of steroid hormone homeostasis in cetaceans. However, blubber cortisol measurements are not quantitatively predictive of circulating cortisol concentrations in bottlenose dolphins. In other mammals, adipose tissue metabolizes steroid hormones. Thus, it is proposed that the disagreement between blubber and blood cortisol in bottlenose dolphins could be due in part to metabolism of corticosteroids in blubber. The purpose of this study is to characterize the ability of blubber to interconvert cortisol and cortisone using an in vitro design. Results demonstrate that bottlenose dolphin blubber microsomes interconvert cortisol and cortisone, an effect that is abated by denaturing the microsomes, indicating this is an enzymatic process. These findings lead to the conclusion that blubber is likely a site of active steroid metabolism, which should be considered in future studies utilizing blubber as a matrix for endocrine assessment.
Collapse
Affiliation(s)
- Thomas M. Galligan
- College of Graduate Studies, Medical University of South Carolina, 331 Fort Johnson Road, Charleston, SC 29412, USA
- JHT, Inc. Under Contract to the National Oceanic and Atmospheric Administration, National Centers for Coastal Ocean Science, Hollings Marine Laboratory, 331 Fort Johnson Road, Charleston, SC 29412, USA
- Department of Fish and Wildlife Conservation, Virginia Polytechnic Institute and State University, 310 West Campus Drive, Cheatham Hall Rm 101, Blacksburg 24060, VA, USA
| | - Lori H. Schwacke
- National Marine Mammal Foundation, 2240 Shelter Island Drive Suite 200, San Diego, CA 92106, USA
| | - Wayne E. McFee
- National Oceanic and Atmospheric Administration, National Ocean Service, Center for Coastal Environmental Health and Biomolecular Research, 219 Fort Johnson Road, Charleston 29412, SC, USA
| | - Ashley S. P. Boggs
- National Institute of Standards and Technology, Chemical Sciences Division, Hollings Marine Laboratory, 331 Fort Johnson Road, Charleston, SC 29412, USA
| |
Collapse
|
42
|
Hennig K, Antignac JP, Bichon E, Morvan ML, Miran I, Delaloge S, Feunteun J, Le Bizec B. Steroid hormone profiling in human breast adipose tissue using semi-automated purification and highly sensitive determination of estrogens by GC-APCI-MS/MS. Anal Bioanal Chem 2017; 410:259-275. [PMID: 29147745 DOI: 10.1007/s00216-017-0717-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 09/22/2017] [Accepted: 10/18/2017] [Indexed: 01/08/2023]
Abstract
Body mass index is a known breast cancer risk factor due to, among other mechanisms, adipose-derived hormones. We developed a method for steroid hormone profiling in adipose tissue to evaluate healthy tissue around the tumor and define new biomarkers for cancer development. A semi-automated sample preparation method based on gel permeation chromatography and subsequent derivatization with trimethylsilyl (TMS) is presented. Progestagens and androgens were determined by GC-EI-MS/MS (LOQ 0.5 to 10 ng/g lipids). For estrogen measurement, a highly sensitive GC-APCI-MS/MS method was developed to reach the required lower limits of detection (0.05 to 0.1 ng/g lipids in matrix, 100-200 fg on column for pure standards). The combination of the two methods allows the screening of 27 androgens and progestagens and 4 estrogens from a single sample. Good accuracies and repeatabilities were achieved for each compound class at their respective limit of detection. The method was applied to determine steroid hormone profiles in adipose tissue of 51 patients, collected both at proximity and distant to the tumor. Out of the 31 tested steroid hormones, 14 compounds were detected in human samples. Pregnenolone, 17-hydroxypregnenolone, dehydroepiandrosterone (DHEA), and androstendione accounted together for 80% of the observed steroid hormone profiles, whereas the estrogens accounted for only 1%. These profiles did not differ based on sampling location, except for ß-estradiol; steroid hormone conversions from androgens to estrogens that potentially take place in adipose or tumoral tissue might not be detectable due a factor 100 difference in concentration of for example DHEA and ß-estradiol. Graphical Abstract Schematic overview of the determination of steroid hormones and metabolites in adipose tissue in proximity and distal to the tumor.
Collapse
Affiliation(s)
- Kristin Hennig
- Laboratoire d'Etude des Résidus et Contaminants dans les Aliments (LABERCA), ONIRIS, LUNAM Université, 44307, Nantes, France
| | - Jean Philippe Antignac
- Laboratoire d'Etude des Résidus et Contaminants dans les Aliments (LABERCA), ONIRIS, LUNAM Université, 44307, Nantes, France.
| | - Emmanuelle Bichon
- Laboratoire d'Etude des Résidus et Contaminants dans les Aliments (LABERCA), ONIRIS, LUNAM Université, 44307, Nantes, France
| | - Marie-Line Morvan
- Laboratoire d'Etude des Résidus et Contaminants dans les Aliments (LABERCA), ONIRIS, LUNAM Université, 44307, Nantes, France
| | - Isabelle Miran
- UMR981 INSERM, Gustave Roussy, Paris-Saclay University, 94805, Villejuif, France
| | - Suzette Delaloge
- Breast Cancer Group, Gustave Roussy Cancer Campus, 94805, Villejuif, France
| | - Jean Feunteun
- UMR8200 CNRS, Gustave Roussy, Paris-Saclay University, 94805, Villejuif, France
| | - Bruno Le Bizec
- Laboratoire d'Etude des Résidus et Contaminants dans les Aliments (LABERCA), ONIRIS, LUNAM Université, 44307, Nantes, France
| |
Collapse
|
43
|
Guénard F, Tchernof A, Deshaies Y, Biron S, Lescelleur O, Biertho L, Marceau S, Pérusse L, Vohl MC. Genetic regulation of differentially methylated genes in visceral adipose tissue of severely obese men discordant for the metabolic syndrome. Transl Res 2017; 184:1-11.e2. [PMID: 28219716 DOI: 10.1016/j.trsl.2017.01.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/19/2016] [Accepted: 01/24/2017] [Indexed: 01/01/2023]
Abstract
A genetic influence on methylation levels has been reported and methylation quantitative trait loci (meQTL) have been identified in various tissues. The contribution of genetic and epigenetic factors in the development of the metabolic syndrome (MetS) has also been noted. To pinpoint candidate genes for testing the association of SNPs with MetS and its components, we aimed to evaluate the contribution of genetic variations to differentially methylated CpG sites in severely obese men discordant for MetS. A genome-wide differential methylation analysis was conducted in visceral adipose tissue (VAT) of 31 severely obese men discordant for MetS (16 with and 15 without MetS) and identified ∼17,800 variable CpG sites. The genome-wide association study conducted to identify the SNPs (meQTL) associated with methylation levels at variable CpG sites revealed 2292 significant associations (P < 2.22 × 10-11) involving 2182 unique meQTLs regulating the methylation levels of 174 variable CpG sites. Two meQTLs disrupting CpG sites located within the collagen-encoding COL11A2 gene were tested for associations with MetS and its components in a cohort of 3021 obese individuals. Rare alleles of these meQTLs showed association with plasma fasting glucose levels. Further analysis conducted on these meQTL suggested a biological impact mediated through the disruption of transcription factor (TF)-binding sites based on the prediction of TF-binding affinities. The current study identified meQTL in the VAT of severely obese men and revealed associations of two COL11A2 meQTL with fasting glucose levels.
Collapse
Affiliation(s)
- Frédéric Guénard
- Institute of Nutrition and Functional Foods (INAF), Québec, Canada; School of Nutrition, Laval University, Québec, Canada
| | - André Tchernof
- School of Nutrition, Laval University, Québec, Canada; Québec Heart and Lung Institute, Québec, Canada
| | - Yves Deshaies
- Québec Heart and Lung Institute, Québec, Canada; Department of Medicine, Laval University, Québec, Canada
| | - Simon Biron
- Department of Surgery, Laval University, Québec, Canada
| | | | | | - Simon Marceau
- Department of Surgery, Laval University, Québec, Canada
| | - Louis Pérusse
- Institute of Nutrition and Functional Foods (INAF), Québec, Canada; Department of Kinesiology, Laval University, Québec, Canada
| | - Marie-Claude Vohl
- Institute of Nutrition and Functional Foods (INAF), Québec, Canada; School of Nutrition, Laval University, Québec, Canada.
| |
Collapse
|
44
|
Swerdloff RS, Dudley RE, Page ST, Wang C, Salameh WA. Dihydrotestosterone: Biochemistry, Physiology, and Clinical Implications of Elevated Blood Levels. Endocr Rev 2017; 38:220-254. [PMID: 28472278 PMCID: PMC6459338 DOI: 10.1210/er.2016-1067] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 04/20/2017] [Indexed: 02/07/2023]
Abstract
Benefits associated with lowered serum DHT levels after 5α-reductase inhibitor (5AR-I) therapy in men have contributed to a misconception that circulating DHT levels are an important stimulus for androgenic action in target tissues (e.g., prostate). Yet evidence from clinical studies indicates that intracellular concentrations of androgens (particularly in androgen-sensitive tissues) are essentially independent of circulating levels. To assess the clinical significance of modest elevations in serum DHT and the DHT/testosterone (T) ratio observed in response to common T replacement therapy, a comprehensive review of the published literature was performed to identify relevant data. Although the primary focus of this review is about DHT in men, we also provide a brief overview of DHT in women. The available published data are limited by the lack of large, well-controlled studies of long duration that are sufficiently powered to expose subtle safety signals. Nonetheless, the preponderance of available clinical data indicates that modest elevations in circulating levels of DHT in response to androgen therapy should not be of concern in clinical practice. Elevated DHT has not been associated with increased risk of prostate disease (e.g., cancer or benign hyperplasia) nor does it appear to have any systemic effects on cardiovascular disease safety parameters (including increased risk of polycythemia) beyond those commonly observed with available T preparations. Well-controlled, long-term studies of transdermal DHT preparations have failed to identify safety signals unique to markedly elevated circulating DHT concentrations or signals materially different from T.
Collapse
Affiliation(s)
- Ronald S Swerdloff
- Division of Endocrinology, Department of Medicine, David Geffen School of Medicine at UCLA, Torrance, California 90502
| | | | - Stephanie T Page
- Division of Metabolism, Endocrinology, and Nutrition, University of Washington School of Medicine, Seattle, Washington 98195
| | - Christina Wang
- Division of Endocrinology, Department of Medicine, David Geffen School of Medicine at UCLA, Torrance, California 90502
- UCLA Clinical and Translational Science Institute, Harbor-UCLA Medical Center, and Los Angeles Biomedical Research Institute, David Geffen School of Medicine at UCLA, Torrance, California 90509
| | - Wael A Salameh
- Division of Endocrinology, Department of Medicine, David Geffen School of Medicine at UCLA, Torrance, California 90502
| |
Collapse
|
45
|
Chen TS, Chen YT, Liu CH, Sun CC, Mao FC. Steroidogenic enzymes of adipose tissue in modulation of trivalent chromium in a mouse model of PCOS. Gynecol Endocrinol 2017; 33:48-52. [PMID: 27449565 DOI: 10.1080/09513590.2016.1205579] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a type of endocrine metabolic disorder with many different consequences to health, most commonly infertility, obesity and insulin resistance. Trivalent chromium (Cr3+) was previously found to improve the metabolic profiles of patients with PCOS. The aim of this study was to explore the effect of Cr on regulating steroidogenic enzymes in adipose tissue. Female BALB/c mice were divided into three groups (n = 6 per group): the control group, PCOS + placebo milk group and PCOS + Cr-containing milk group. The dietary intake of Cr significantly decreased fasting blood sugar (FBS) and homeostasis model assessment of insulin resistance levels in the murine model of PCOS. Importantly, we found significant correlations among the levels of Cr, insulin and dehydroepiandrosterone (DHEA). In adipose tissue, decreases in the enzyme expressions of 3β-hydroxysteroid dehydrogenase (3β-HSD) and 17β-hydroxysteroid dehydrogenase, but not of aromatase, were observed. By understanding the role of steroidogenic enzymes in PCOS in normal and pathological states, trace elements may be used as a form of adjunctive therapy in the management of patients with PCOS.
Collapse
Affiliation(s)
- Tsung-Sheng Chen
- a Department of Veterinary Medicine , College of Veterinary Medicine, National Chung Hsing University , Taichung , Taiwan
| | - Yi-Ting Chen
- a Department of Veterinary Medicine , College of Veterinary Medicine, National Chung Hsing University , Taichung , Taiwan
| | - Chia-Hsin Liu
- a Department of Veterinary Medicine , College of Veterinary Medicine, National Chung Hsing University , Taichung , Taiwan
| | - Chi-Ching Sun
- a Department of Veterinary Medicine , College of Veterinary Medicine, National Chung Hsing University , Taichung , Taiwan
| | - Frank Chiahung Mao
- a Department of Veterinary Medicine , College of Veterinary Medicine, National Chung Hsing University , Taichung , Taiwan
| |
Collapse
|
46
|
Robic A, Feve K, Riquet J, Prunier A. Transcript levels of genes implicated in steroidogenesis in the testes and fat tissue in relation to androstenone accumulation in fat of pubertal pigs. Domest Anim Endocrinol 2016; 57:1-9. [PMID: 27285831 DOI: 10.1016/j.domaniend.2016.03.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 03/15/2016] [Accepted: 03/27/2016] [Indexed: 12/19/2022]
Abstract
The present study was performed to measure messenger RNA levels of steroidogenic enzymes in testes and fat tissue and determine whether they are related to fat androstenone level. Real-time polymerase chain reaction experiments were performed on 26 testes and 12 adipose tissue samples from pubertal boars using 21 genes. The absence of significant correlations between fat androstenone and the transcriptional activity of the SRD5A2 and SRD5A3 genes but the high correlation coefficient with that of the SRD5A1 gene (r = 0.62, P < 0.05) suggests that the enzyme coded by SRD5A1 is mainly responsible for the last step of androstenone synthesis. The testicular transcriptional activities of CYP17, CYP11A1, CYP19A, AKR1C-pig6, SRD5A1, LHCGR, and AR were significantly correlated. Only transcriptional levels of CYP17, CYP11A1, CYP19A, SRD5A1, and AKR1C-pig6 were correlated with the fat concentration of androstenone (0.57 < r < 0.70, P < 0.05) confirming that the amount of androstenone stored in fat is related to the production in testes of androstenone and more generally to all sex steroids. Altogether, our data are in favor of a preponderant role of AKR1C-pig6 instead of HSD17B3 for testicular synthesis of steroids. Concerning fat tissue, our data do not support a significant de novo biosynthesis of steroids in porcine adipose tissues. The presence of transcripts coding for steroid enzymes, especially those of AKR1C-pig6, suggests that steroids can be transformed. None of transcript abundance was related to androstenone accumulation (P > 0.1). Therefore, steroids synthesized elsewhere can be transformed in fat tissue but synthesis of androstenone is unlikely.
Collapse
Affiliation(s)
- A Robic
- GenPhySE, Université de Toulouse, INRA, INPT, ENVT, Castanet-Tolosan 31320, France.
| | - K Feve
- GenPhySE, Université de Toulouse, INRA, INPT, ENVT, Castanet-Tolosan 31320, France
| | - J Riquet
- GenPhySE, Université de Toulouse, INRA, INPT, ENVT, Castanet-Tolosan 31320, France
| | - A Prunier
- PEGASE, Agrocampus Ouest, INRA, Saint-Gilles 35590, France
| |
Collapse
|
47
|
Vallée M. Neurosteroids and potential therapeutics: Focus on pregnenolone. J Steroid Biochem Mol Biol 2016; 160:78-87. [PMID: 26433186 DOI: 10.1016/j.jsbmb.2015.09.030] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 09/18/2015] [Accepted: 09/21/2015] [Indexed: 12/26/2022]
Abstract
Considerable evidence from preclinical and clinical studies shows that steroids and in particular neurosteroids are important endogenous modulators of several brain-related functions. In this context, it remains to be elucidated whether neurosteroids may serve as biomarkers in the diagnosis of disorders and might have therapeutic potential for the treatment of these disorders. Pregnenolone (PREG) is the main steroid synthesized from cholesterol in mammals and invertebrates. PREG has three main sources of synthesis, the gonads, adrenal glands and brain and is submitted to various metabolizing pathways which are modulated depending on various factors including species, steroidogenic tissues and steroidogenic enzymes. Looking at the whole picture of steroids, PREG is often known as the precursor to other steroids and not as an active steroid per se. Actually, physiological and brain functions have been studied mainly for steroids that are very active either binding to specific intracellular receptors, or modulating with high affinity the abundant membrane receptors, GABAA or NMDA receptors. However, when high sensitive and specific methodological approaches were available to analyze low concentrations of steroids and then match endogenous levels of different steroid metabolomes, several studies have reported more significant alterations in PREG than in other steroids in extraphysiological or pathological conditions, suggesting that PREG could play a functional role as well. Additionally, several molecular targets of PREG were revealed in the mammalian brain and beneficial effects of PREG have been demonstrated in preclinical and clinical studies. On this basis, this review will be divided into three parts. The first provides a brief overview of the molecular targets of PREG and the pharmacological effects observed in animal and human studies. The second will focus on the possible functional role of PREG with an outline of the modulation of PREG levels in animal and in human research. Finally, the review will highlight the possible therapeutic uses of PREG that point towards the development of pregnenolone-like molecules.
Collapse
Affiliation(s)
- Monique Vallée
- INSERM U862, Neurocentre Magendie, Pathophysiology of Addiction, Bordeaux F33077, France; Université de Bordeaux, Bordeaux F33077, France.
| |
Collapse
|
48
|
Paatela H, Wang F, Vihma V, Savolainen-Peltonen H, Mikkola TS, Turpeinen U, Hämäläinen E, Jauhiainen M, Tikkanen MJ. Steroid sulfatase activity in subcutaneous and visceral adipose tissue: a comparison between pre- and postmenopausal women. Eur J Endocrinol 2016; 174:167-75. [PMID: 26553725 DOI: 10.1530/eje-15-0831] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 11/06/2015] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Adipose tissue is an important extragonadal site for steroid hormone biosynthesis. After menopause, estrogens are synthesized exclusively in peripheral tissues from circulating steroid precursors, of which the most abundant is dehydroepiandrosterone sulfate (DHEAS). Our aim was to study activity of steroid sulfatase, an enzyme hydrolyzing DHEAS, and expression of steroid-converting enzyme genes in subcutaneous and visceral adipose tissue derived from pre- and postmenopausal women. DESIGN Serum and paired abdominal subcutaneous and visceral adipose tissue samples were obtained from 18 premenopausal and seven postmenopausal women undergoing elective surgery for non-malignant reasons in Helsinki University Central Hospital. METHODS To assess steroid sulfatase activity, radiolabeled DHEAS was incubated in the presence of adipose tissue homogenate and the liberated dehydroepiandrosterone (DHEA) was measured. Gene mRNA expressions were analyzed by quantitative RT-PCR. Serum DHEAS, DHEA, and estrogen concentrations were determined by liquid chromatography-tandem mass spectrometry. RESULTS Steroid sulfatase activity was higher in postmenopausal compared to premenopausal women in subcutaneous (median 379 vs 257 pmol/kg tissue per hour; P=0.006) and visceral (545 vs 360 pmol/kg per hour; P=0.004) adipose tissue. Visceral fat showed higher sulfatase activity than subcutaneous fat in premenopausal (P=0.035) and all (P=0.010) women. The mRNA expression levels of two estradiol-producing enzymes, aromatase and 17β-hydroxysteroid dehydrogenase type 12, were higher in postmenopausal than in premenopausal subcutaneous adipose tissue. CONCLUSIONS Steroid sulfatase activity in adipose tissue was higher in postmenopausal than in premenopausal women suggesting that DHEAS, derived from the circulation, could be more efficiently utilized in postmenopausal adipose tissue for the formation of biologically active sex hormones.
Collapse
Affiliation(s)
- Hanna Paatela
- Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland
| | - Feng Wang
- Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland
| | - Veera Vihma
- Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland
| | - Hanna Savolainen-Peltonen
- Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland
| | - Tomi S Mikkola
- Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland
| | - Ursula Turpeinen
- Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland
| | - Esa Hämäläinen
- Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland
| | - Matti Jauhiainen
- Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland
| | - Matti J Tikkanen
- Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland Folkhälsan Research CenterBiomedicum Helsinki, 00290 Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalHeart and Lung Center, Helsinki, FinlandUniversity of Helsinki and Helsinki University Central HospitalObstetrics and Gynecology, Helsinki, FinlandHelsinki University Central HospitalHUSLAB, Helsinki, FinlandNational Institute for Health and WelfareGenomics and Biomarkers Unit, Helsinki, Finland
| |
Collapse
|
49
|
Martinez-Useros J, Garcia-Foncillas J. Obesity and colorectal cancer: molecular features of adipose tissue. J Transl Med 2016; 14:21. [PMID: 26801617 PMCID: PMC4722674 DOI: 10.1186/s12967-016-0772-5] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/05/2016] [Indexed: 02/06/2023] Open
Abstract
The huge part of population in developed countries is overweight or obese. Obesity is often determined by body mass index (BMI) but new accurate methods and ratios have recently appeared to measure body fat or fat located in the intestines. Early diagnosis of obesity is crucial since it is considered an increasing colorectal cancer risk factor. On the one hand, colorectal cancer has been strongly associated with lifestyle factors. A diet rich in red and processed meats may increase colorectal cancer risk; however, high-fiber diets (grains, cereals and fruits) have been associated with a decreased risk of colorectal cancer. Other life-style factors associated with obesity that also increase colorectal cancer risk are physical inactivity, smoking and high alcohol intake. Cutting-edge studies reported that high-risk transformation ability of adipose tissue is due to production of different pro-inflammatory cytokines like IL-8, IL-6 or IL-2 and other enzymes like lactate dehydrogenase (LDH) and tumour necrosis factor alpha (TNFα). Furthermore, oxidative stress produces fatty-acid peroxidation whose metabolites possess very high toxicities and mutagenic properties. 4-hydroxy-2-nonenal (4-HNE) is an active compounds that upregulates prostaglandin E2 which is directly associated with high proliferative colorectal cancer. Moreover, 4-HNE deregulates cell proliferation, cell survival, differentiation, autophagy, senescence, apoptosis and necrosis via mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase (PIK3CA)—AKT and protein kinase C pathways. Other product of lipid peroxidation is malondialdehyde (MDA) being able to regulate insulin through WNT-pathway as well as having demonstrated its mutagenic capability. Accumulation of point mutation enables genomic evolution of colorectal cancer described in the model of Fearon and Vogelstein. In this review, we will summarize different determination methods and techniques to assess a truthfully diagnosis and we will explain some of the capabilities that performs adipocytes as the largest endocrine organ.
Collapse
Affiliation(s)
- Javier Martinez-Useros
- Translational Oncology Division, Oncohealth Institute, FIIS-Fundacion Jimenez Diaz, Av. Reyes Catolicos 2, 28040, Madrid, Spain.
| | - Jesus Garcia-Foncillas
- Translational Oncology Division, Oncohealth Institute, FIIS-Fundacion Jimenez Diaz, Av. Reyes Catolicos 2, 28040, Madrid, Spain.
| |
Collapse
|
50
|
Li J, Papadopoulos V, Vihma V. Steroid biosynthesis in adipose tissue. Steroids 2015; 103:89-104. [PMID: 25846979 DOI: 10.1016/j.steroids.2015.03.016] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/20/2015] [Accepted: 03/24/2015] [Indexed: 12/25/2022]
Abstract
Tissue-specific expression of steroidogenic enzymes allows the modulation of active steroid levels in a local manner. Thus, the measurement of local steroid concentrations, rather than the circulating levels, has been recognized as a more accurate indicator of the steroid action within a specific tissue. Adipose tissue, one of the largest endocrine tissues in the human body, has been established as an important site for steroid storage and metabolism. Locally produced steroids, through the enzymatic conversion from steroid precursors delivered to adipose tissue, have been proven to either functionally regulate adipose tissue metabolism, or quantitatively contribute to the whole body's steroid levels. Most recently, it has been suggested that adipose tissue may contain the steroidogenic machinery necessary for the initiation of steroid biosynthesis de novo from cholesterol. This review summarizes the evidence indicating the presence of the entire steroidogenic apparatus in adipose tissue and discusses the potential roles of local steroid products in modulating adipose tissue activity and other metabolic parameters.
Collapse
Affiliation(s)
- Jiehan Li
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada; Department of Medicine, McGill University, Montreal, Canada; Department of Biochemistry, McGill University, Montreal, Canada
| | - Vassilios Papadopoulos
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada; Department of Medicine, McGill University, Montreal, Canada; Department of Biochemistry, McGill University, Montreal, Canada.
| | - Veera Vihma
- Folkhälsan Research Center, Helsinki, Finland; University of Helsinki and Helsinki University Central Hospital, Heart and Lung Center, Helsinki, Finland.
| |
Collapse
|