37 results on '"Ohlsson C"'
Search Results
2. Identification of Target Cells for the Genomic Effects of Estrogens in Bone
- Author
-
Windahl, S H., Lagerquist, M K., Andersson, N, Jochems, C, Kallkopf, A, Håkansson, C, Inzunza, J, Gustafsson, J -Å., van der Saag, P T., Carlsten, H, Pettersson, K, and Ohlsson, C
- Published
- 2007
3. Female Mice Lacking Estrogen Receptor-α in Hypothalamic Proopiomelanocortin (POMC) Neurons Display Enhanced Estrogenic Response on Cortical Bone Mass
- Author
-
Farman, H. H., primary, Windahl, S. H., primary, Westberg, L., primary, Isaksson, H., primary, Egecioglu, E., primary, Schele, E., primary, Ryberg, H., primary, Jansson, J. O., primary, Tuukkanen, J., primary, Koskela, A., primary, Xie, S. K., primary, Hahner, L., primary, Zehr, J., primary, Clegg, D. J., primary, Lagerquist, M. K., primary, and Ohlsson, C., primary
- Published
- 2016
- Full Text
- View/download PDF
4. The Role of the Growth Hormone/Insulin-Like Growth Factor I Axis in Stimulation of Protein Synthesis in Skeletal Muscles Following Oral Refeeding
- Author
-
Svanberg, E., primary, Powell-Braxton, L., additional, Ohlsson, C., additional, Zachrisson, H., additional, and Lundholm, K., additional
- Published
- 1998
- Full Text
- View/download PDF
5. Elevated levels of growth hormone increase bone mineral content in normal young mice, but not in ovariectomized mice.
- Author
-
Sandstedt, J, primary, Törnell, J, additional, Norjavaara, E, additional, Isaksson, O G, additional, and Ohlsson, C, additional
- Published
- 1996
- Full Text
- View/download PDF
6. Insulin-like growth factor binding proteins-2 and -3 stimulate growth hormone receptor binding and mitogenesis in rat osteosarcoma cells.
- Author
-
Slootweg, M C, primary, Ohlsson, C, additional, Salles, J P, additional, de Vries, C P, additional, and Netelenbos, J C, additional
- Published
- 1995
- Full Text
- View/download PDF
7. Disproportional bone growth and reduced weight gain in gonadectomized male bovine growth hormone transgenic and normal mice.
- Author
-
Sandstedt, J, primary, Ohlsson, C, additional, Norjavaara, E, additional, Nilsson, J, additional, and Törnell, J, additional
- Published
- 1994
- Full Text
- View/download PDF
8. Embryonic stem cells express growth hormone receptors: regulation by retinoic acid.
- Author
-
Ohlsson, C, primary, Lövstedt, K, additional, Holmes, P V, additional, Nilsson, A, additional, Carlsson, L, additional, and Törnell, J, additional
- Published
- 1993
- Full Text
- View/download PDF
9. HSD17B1 Compensates for HSD17B3 Deficiency in Fetal Mouse Testis but Not in Adults.
- Author
-
Junnila A, Zhang FP, Martínez Nieto G, Hakkarainen J, Mäkelä JA, Ohlsson C, Sipilä P, and Poutanen M
- Subjects
- Animals, Male, Mice, Estradiol Dehydrogenases metabolism, Estradiol Dehydrogenases genetics, Fetus metabolism, 17-Hydroxysteroid Dehydrogenases metabolism, 17-Hydroxysteroid Dehydrogenases genetics, 17-Hydroxysteroid Dehydrogenases deficiency, Mice, Knockout, Testis metabolism, Testis embryology, Testosterone blood, Testosterone metabolism
- Abstract
Hydroxysteroid (17β) dehydrogenase (HSD17B) enzymes convert 17-ketosteroids to 17beta-hydroxysteroids, an essential step in testosterone biosynthesis. Human XY individuals with inactivating HSD17B3 mutations are born with female-appearing external genitalia due to testosterone deficiency. However, at puberty their testosterone production reactivates, indicating HSD17B3-independent testosterone synthesis. We have recently shown that Hsd17b3 knockout (3-KO) male mice display a similar endocrine imbalance, with high serum androstenedione and testosterone in adulthood, but milder undermasculinization than humans. Here, we studied whether HSD17B1 is responsible for the remaining HSD17B activity in the 3-KO male mice by generating a Ser134Ala point mutation that disrupted the enzymatic activity of HSD17B1 (1-KO) followed by breeding Hsd17b1/Hsd17b3 double-KO (DKO) mice. In contrast to 3-KO, inactivation of both HSD17B3 and HSD17B1 in mice results in a dramatic drop in testosterone synthesis during the fetal period. This resulted in a female-like anogenital distance at birth, and adult DKO males displayed more severe undermasculinization than 3-KO, including more strongly reduced weight of seminal vesicles, levator ani, epididymis, and testis. However, qualitatively normal spermatogenesis was detected in adult DKO males. Furthermore, similar to 3-KO mice, high serum testosterone was still detected in adult DKO mice, accompanied by upregulation of various steroidogenic enzymes. The data show that HSD17B1 compensates for HSD17B3 deficiency in fetal mouse testis but is not the enzyme responsible for testosterone synthesis in adult mice with inactivated HSD17B3. Therefore, other enzymes are able to convert androstenedione to testosterone in the adult mouse testis and presumably also in the human testis., (© The Author(s) 2024. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For commercial re-use, please contact reprints@oup.com for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
10. Dietary Progesterone Contributes to Intratissue Levels of Progesterone in Male Mice.
- Author
-
Colldén H, Hagberg Thulin M, Landin A, Horkeby K, Lagerquist M, Wu J, Nilsson KH, Grahnemo L, Poutanen M, Ryberg H, Vandenput L, and Ohlsson C
- Subjects
- Humans, Cattle, Mice, Male, Animals, Progesterone, Androgen Antagonists, Adrenalectomy, Orchiectomy, Androgens, Prostatic Neoplasms
- Abstract
Progesterone serum levels have been identified as a potential predictor for treatment effect in men with advanced prostate cancer, which is an androgen-driven disease. Although progesterone is the most abundant sex steroid in orchiectomized (ORX) male mice, the origins of progesterone in males are unclear. To determine the origins of progesterone and androgens, we first determined the effect of ORX, adrenalectomy (ADX), or both (ORX + ADX) on progesterone levels in multiple male mouse tissues. As expected, intratissue androgen levels were mainly testicular derived. Interestingly, progesterone levels remained high after ORX and ORX + ADX with the highest levels in white adipose tissue and in the gastrointestinal tract. High progesterone levels were observed in mouse chow and exceptionally high progesterone levels were observed in food items such as dairy, eggs, and beef, all derived from female animals of reproductive age. To determine if orally ingested progesterone contributes to tissue levels of progesterone in males, we treated ORX + ADX and sham mice with isotope-labeled progesterone or vehicle by oral gavage. We observed a significant uptake of labeled progesterone in white adipose tissue and prostate, suggesting that dietary progesterone may contribute to tissue levels of progesterone. In conclusion, although adrenal-derived progesterone contributes to intratissue progesterone levels in males, nonadrenal progesterone sources also contribute. We propose that dietary progesterone is absorbed and contributes to intratissue progesterone levels in male mice. We speculate that food with high progesterone content could be a significant source of progesterone in males, possibly with consequences for men undergoing androgen deprivation therapy for prostate cancer., (© The Author(s) 2023. Published by Oxford University Press on behalf of the Endocrine Society.)
- Published
- 2023
- Full Text
- View/download PDF
11. Bone-Derived IGF-I Regulates Radial Bone Growth in Adult Male Mice.
- Author
-
Svensson J, Sjögren K, Lawenius L, Koskela A, Tuukkanen J, Nilsson KH, Movérare-Skrtic S, and Ohlsson C
- Subjects
- Humans, Mice, Male, Animals, Aged, Infant, Bone Development genetics, Cancellous Bone diagnostic imaging, Cancellous Bone metabolism, Disease Models, Animal, Tamoxifen pharmacology, Bone Density genetics, Insulin-Like Growth Factor I metabolism, Bone and Bones diagnostic imaging, Bone and Bones metabolism
- Abstract
Insulin-like growth factor-I (IGF-I) levels, which are reduced by age, and cortical bone dimensions are major determinants of fracture risk in elderly subjects. Inactivation of liver-derived circulating IGF-I results in reduced periosteal bone expansion in young and older mice. In mice with lifelong depletion of IGF-I in osteoblast lineage cells, the long bones display reduced cortical bone width. However, it has not previously been investigated whether inducible inactivation of IGF-I locally in bone in adult/old mice affects the bone phenotype. Adult tamoxifen-inducible inactivation of IGF-I using a CAGG-CreER mouse model (inducible IGF-IKO mice) substantially reduced IGF-I expression in bone (-55%) but not in liver. Serum IGF-I and body weight were unchanged. We used this inducible mouse model to assess the effect of local IGF-I on the skeleton in adult male mice, avoiding confounding developmental effects. After tamoxifen-induced inactivation of the IGF-I gene at 9 months of age, the skeletal phenotype was determined at 14 months of age. Computed tomography analyses of tibia revealed that the mid-diaphyseal cortical periosteal and endosteal circumferences and calculated bone strength parameters were decreased in inducible IGF-IKO mice compared with controls. Furthermore, 3-point bending showed reduced tibia cortical bone stiffness in inducible IGF-IKO mice. In contrast, the tibia and vertebral trabecular bone volume fraction was unchanged. In conclusion, inactivation of IGF-I in cortical bone with unchanged liver-derived IGF-I in older male mice resulted in reduced radial growth of cortical bone. This suggests that not only circulating IGF-I but also locally derived IGF-I regulates the cortical bone phenotype in older mice., (© The Author(s) 2023. Published by Oxford University Press on behalf of the Endocrine Society.)
- Published
- 2023
- Full Text
- View/download PDF
12. Dehydroepiandrosterone Supplementation Results in Varying Tissue-specific Levels of Dihydrotestosterone in Male Mice.
- Author
-
Colldén H, Nilsson ME, Norlén AK, Landin A, Windahl SH, Wu J, Horkeby K, Lagerquist MK, Ryberg H, Poutanen M, Vandenput L, and Ohlsson C
- Subjects
- Male, Mice, Animals, Dehydroepiandrosterone pharmacology, Dehydroepiandrosterone metabolism, Testosterone, Dietary Supplements, Dihydrotestosterone pharmacology, Androgens pharmacology
- Abstract
Dehydroepiandrosterone (DHEA), an adrenal androgen precursor, can be metabolized in target tissues into active sex steroids. It has been proposed that DHEA supplementation might result in restoration of physiological local sex steroid levels, but knowledge on the effect of DHEA treatment on local sex steroid levels in multiple tissues is lacking. To determine the effects of DHEA on tissue-specific levels of sex steroids, we treated orchiectomized (ORX) male mice with DHEA for 3 weeks and compared them with vehicle-treated ORX mice and gonadal intact mice. Intra-tissue levels of sex steroids were analyzed in reproductive organs (seminal vesicles, prostate, m. levator ani), major body compartments (white adipose tissue, skeletal muscle, and brain), adrenals, liver, and serum using a sensitive and validated gas chromatography-mass spectrometry method. DHEA treatment restored levels of both testosterone (T) and dihydrotestosterone (DHT) to approximately physiological levels in male reproductive organs. In contrast, this treatment did not increase DHT levels in skeletal muscle or brain. In the liver, DHEA treatment substantially increased levels of T (at least 4-fold) and DHT (+536%, P < 0.01) compared with vehicle-treated ORX mice. In conclusion, we provide a comprehensive map of the effect of DHEA treatment on intra-tissue sex steroid levels in ORX mice with a restoration of physiological levels of androgens in male reproductive organs while DHT levels were not restored in the skeletal muscle or brain. This, and the unexpected supraphysiological androgen levels in the liver, may be a cause for concern considering the uncontrolled use of DHEA., (© The Author(s) 2022. Published by Oxford University Press on behalf of the Endocrine Society.)
- Published
- 2022
- Full Text
- View/download PDF
13. Inactivation of AR or ERα in Extrahypothalamic Neurons Does not Affect Osteogenic Response to Loading in Male Mice.
- Author
-
Kim NR, David K, Sommers V, Schollaert D, Deboel L, Ohlsson C, Gustafsson JÅ, Antonio L, Decallonne B, Claessens F, Vanderschueren D, and Dubois V
- Subjects
- Animals, Cross-Sectional Studies, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Neurons metabolism, Tibia, Estrogen Receptor alpha genetics, Estrogen Receptor alpha metabolism, Receptors, Androgen genetics, Receptors, Androgen metabolism
- Abstract
Failure of bone mass maintenance in spite of functional loading is an important contributor to osteoporosis and related fractures. While the link between sex steroids and the osteogenic response to loading is well established, the underlying mechanisms are unknown, hampering clinical relevance. Androgens inhibit mechanoresponsiveness in male mice, but the cell type mediating this effect remains unidentified. To evaluate the role of neuronal sex steroid receptor signaling in the male bone's adaptive capacity, we subjected adult male mice with an extrahypothalamic neuron-specific knockout of the androgen receptor (N-ARKO) or the estrogen receptor alpha (N-ERαKO) to in vivo mechanical stimulation of the tibia. Loading increased cortical thickness in the control animals mainly through periosteal expansion, as total cross-sectional tissue area and cortical bone area but not medullary area were higher in the loaded than the unloaded tibia. Trabecular bone volume fraction also increased upon loading in the control group, mostly due to trabecular thickening. N-ARKO and N-ERαKO males displayed a loading response at both the cortical and trabecular bone compartments that was not different from their control littermates. In conclusion, we show that the presence of androgen receptor or estrogen receptor alpha in extrahypothalamic neurons is dispensable for the osteogenic response to mechanical loading in male mice., (© The Author(s) 2022. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
14. Comprehensive Sex Steroid Profiling in Multiple Tissues Reveals Novel Insights in Sex Steroid Distribution in Male Mice.
- Author
-
Colldén H, Nilsson ME, Norlén AK, Landin A, Windahl SH, Wu J, Gustafsson KL, Poutanen M, Ryberg H, Vandenput L, and Ohlsson C
- Subjects
- Adipose Tissue, White chemistry, Androstenedione analysis, Animals, Dihydrotestosterone analysis, Estradiol analysis, Estrone analysis, Gas Chromatography-Mass Spectrometry methods, Gonadal Steroid Hormones blood, Gonadal Steroid Hormones pharmacokinetics, Male, Mice, Mice, Inbred C57BL, Orchiectomy, Progesterone analysis, Sensitivity and Specificity, Tandem Mass Spectrometry methods, Testosterone analysis, Tissue Distribution, Gonadal Steroid Hormones analysis
- Abstract
A comprehensive atlas of sex steroid distribution in multiple tissues is currently lacking, and how circulating and tissue sex steroid levels correlate remains unknown. Here, we adapted and validated a gas chromatography tandem mass spectrometry method for simultaneous measurement of testosterone (T), dihydrotestosterone (DHT), androstenedione, progesterone (Prog), estradiol, and estrone in mouse tissues. We then mapped the sex steroid pattern in 10 different endocrine, reproductive, and major body compartment tissues and serum of gonadal intact and orchiectomized (ORX) male mice. In gonadal intact males, high levels of DHT were observed in reproductive tissues, but also in white adipose tissue (WAT). A major part of the total body reservoir of androgens (T and DHT) and Prog was found in WAT. Serum levels of androgens and Prog were strongly correlated with corresponding levels in the brain while only modestly correlated with corresponding levels in WAT. After orchiectomy, the levels of the active androgens T and DHT decreased markedly while Prog levels in male reproductive tissues increased slightly. In ORX mice, Prog was by far the most abundant sex steroid, and, again, WAT constituted the major reservoir of Prog in the body. In conclusion, we present a comprehensive atlas of tissue and serum concentrations of sex hormones in male mice, revealing novel insights in sex steroid distribution. Brain sex steroid levels are well reflected by serum levels and WAT constitutes a large reservoir of sex steroids in male mice. In addition, Prog is the most abundant sex hormone in ORX mice., (© The Author(s) 2022. Published by Oxford University Press on behalf of the Endocrine Society.)
- Published
- 2022
- Full Text
- View/download PDF
15. Testosterone Reduces Body Fat in Male Mice by Stimulation of Physical Activity Via Extrahypothalamic ERα Signaling.
- Author
-
Kim NR, David K, Corbeels K, Khalil R, Antonio L, Schollaert D, Deboel L, Ohlsson C, Gustafsson JÅ, Vangoitsenhoven R, Van der Schueren B, Decallonne B, Claessens F, Vanderschueren D, and Dubois V
- Subjects
- Adipose Tissue metabolism, Animals, Dihydrotestosterone pharmacology, Energy Metabolism drug effects, Energy Metabolism genetics, Estrogen Receptor alpha genetics, Estrogen Receptor alpha metabolism, Hypogonadism genetics, Hypogonadism metabolism, Hypothalamus metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Obese, Motor Activity drug effects, Obesity genetics, Obesity metabolism, Physical Conditioning, Animal physiology, Signal Transduction drug effects, Signal Transduction genetics, Testosterone Congeners pharmacology, Adipose Tissue drug effects, Estrogen Receptor alpha physiology, Motor Activity physiology, Testosterone pharmacology
- Abstract
Testosterone (T) reduces male fat mass, but the underlying mechanisms remain elusive, limiting its clinical relevance in hypogonadism-associated obesity. Here, we subjected chemically castrated high-fat diet-induced adult obese male mice to supplementation with T or the nonaromatizable androgen dihydrotestosterone (DHT) for 20 weeks. Both hormones increased lean mass, thereby indirectly increasing oxygen consumption and energy expenditure. In addition, T but not DHT decreased fat mass and increased ambulatory activity, indicating a role for aromatization into estrogens. Investigation of the pattern of aromatase expression in various murine tissues revealed the absence of Cyp19a1 expression in adipose tissue while high levels were observed in brain and gonads. In obese hypogonadal male mice with extrahypothalamic neuronal estrogen receptor alpha deletion (N-ERαKO), T still increased lean mass but was unable to decrease fat mass. The stimulatory effect of T on ambulatory activity was also abolished in N-ERαKO males. In conclusion, our work demonstrates that the fat-burning action of T is dependent on aromatization into estrogens and is at least partially mediated by the stimulation of physical activity via extrahypothalamic ERα signaling. In contrast, the increase in lean mass upon T supplementation is mediated through the androgen receptor and indirectly leads to an increase in energy expenditure, which might also contribute to the fat-burning effects of T., (© The Author(s) 2021. Published by Oxford University Press on behalf of the Endocrine Society.)
- Published
- 2021
- Full Text
- View/download PDF
16. A Body Weight Sensor Regulates Prepubertal Growth via the Somatotropic Axis in Male Rats.
- Author
-
Jansson JO, Dalmau Gasull A, Schéle E, Dickson SL, Palsdottir V, Palmquist A, Gironès FF, Bellman J, Anesten F, Hägg D, and Ohlsson C
- Subjects
- Animals, Body Weight drug effects, Growth Hormone metabolism, Growth Hormone-Releasing Hormone metabolism, Homeostasis drug effects, Locomotion physiology, Male, Rats, Rats, Sprague-Dawley, Receptors, Somatotropin drug effects, Receptors, Somatotropin metabolism, Receptors, Somatotropin physiology, Sexual Maturation drug effects, Signal Transduction drug effects, Body Weight physiology, Growth Hormone pharmacology, Growth and Development drug effects
- Abstract
In healthy conditions, prepubertal growth follows an individual specific growth channel. Growth hormone (GH) is undoubtedly the major regulator of growth. However, the homeostatic regulation to maintain the individual specific growth channel during growth is unclear. We recently hypothesized a body weight sensing homeostatic regulation of body weight during adulthood, the gravitostat. We now investigated if sensing of body weight also contributes to the strict homeostatic regulation to maintain the individual specific growth channel during prepubertal growth. To evaluate the effect of increased artificial loading on prepubertal growth, we implanted heavy (20% of body weight) or light (2% of the body weight) capsules into the abdomen of 26-day-old male rats. The body growth, as determined by change in biological body weight and growth of the long bones and the axial skeleton, was reduced in rats bearing a heavy load compared with light load. Removal of the increased load resulted in a catch-up growth and a normalization of body weight. Loading decreased hypothalamic growth hormone releasing hormone mRNA, liver insulin-like growth factor (IGF)-1 mRNA, and serum IGF-1, suggesting that the reduced body growth was caused by a negative feedback regulation on the somatotropic axis and this notion was supported by the fact that increased loading did not reduce body growth in GH-treated rats. Based on these data, we propose the gravitostat hypothesis for the regulation of prepubertal growth. This states that there is a homeostatic regulation to maintain the individual specific growth channel via body weight sensing, regulating the somatotropic axis and explaining catch-up growth., (© The Author(s) 2021. Published by Oxford University Press on behalf of the Endocrine Society.)
- Published
- 2021
- Full Text
- View/download PDF
17. Interactions Between the Gravitostat and the Fibroblast Growth Factor System for the Regulation of Body Weight.
- Author
-
Palsdottir V, Windahl SH, Hägg DA, Keantar H, Bellman J, Buchanan A, Vaughan TJ, Lindén D, Jansson JO, and Ohlsson C
- Subjects
- Adipose Tissue drug effects, Adipose Tissue metabolism, Animals, Antibodies, Monoclonal immunology, Antibodies, Monoclonal pharmacology, Body Weight drug effects, Body Weight genetics, Fibroblast Growth Factors blood, Fibroblast Growth Factors genetics, Gene Expression drug effects, Leptin pharmacology, Liver drug effects, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Obesity metabolism, Receptor, Fibroblast Growth Factor, Type 1 genetics, Receptor, Fibroblast Growth Factor, Type 1 immunology, Receptor, Fibroblast Growth Factor, Type 1 metabolism, Body Weight physiology, Fibroblast Growth Factors metabolism, Liver metabolism
- Abstract
Both fibroblast growth factors (FGFs), by binding to FGF receptors (FGFRs), and activation of the gravitostat, by artificial loading, decrease the body weight (BW). Previous studies demonstrate that both the FGF system and loading have the capacity to regulate BW independently of leptin. The aim of the current study was to determine the possible interactions between the effect of increased loading and the FGF system for the regulation of BW. We observed that the BW-reducing effect of increased loading was abolished in mice treated with a monoclonal antibody directed against FGFR1c, suggesting interactions between the two systems. As serum levels of endocrine FGF21 and hepatic FGF21 mRNA were increased in the loaded mice compared with the control mice, we first evaluated the loading response in FGF21 over expressing mice with constant high FGF21 levels. Leptin treatment, but not increased loading, decreased the BW in the FGF21-overexpressing mice, demonstrating that specifically the loading effect is attenuated in the presence of high activity in the FGF system. However, as FGF21 knockout mice displayed a normal loading response on BW, FGF21 is neither mediating nor essential for the loading response. In conclusion, the BW-reducing effect of increased loading but not of leptin treatment is blocked by high activity in the FGF system. We propose that both the gravitostat and the FGF system regulate BW independently of leptin and that pharmacologically enhanced activity in the FGF system reduces the sensitivity of the gravitostat., (Copyright © 2019 Endocrine Society.)
- Published
- 2019
- Full Text
- View/download PDF
18. The Gravitostat Regulates Fat Mass in Obese Male Mice While Leptin Regulates Fat Mass in Lean Male Mice.
- Author
-
Ohlsson C, Hägg DA, Hammarhjelm F, Dalmau Gasull A, Bellman J, Windahl SH, Palsdottir V, and Jansson JO
- Subjects
- Animals, Leptin blood, Lipid Metabolism drug effects, Male, Mice, Mice, Inbred C57BL, Obesity metabolism, Overweight metabolism, Thinness metabolism, Adipose Tissue drug effects, Body Weight drug effects, Leptin pharmacology
- Abstract
Leptin has been the only known homeostatic regulator of fat mass, but we recently found evidence for a second one, named the gravitostat. In the current study, we compared the effects of leptin and increased loading (gravitostat stimulation) on fat mass in mice with different levels of body weight (lean, overweight, and obese). Leptin infusion suppressed body weight and fat mass in lean mice given normal chow but not in overweight or obese mice given a high-fat diet for 4 and 8 weeks, respectively. The maximum effect of leptin on body weight and fat mass was obtained already at <44 ng/mL of serum leptin. Increased loading using intraperitoneal capsules with different weights decreased body weight in overweight and obese mice. Although the implantation of an empty capsule reduced the body weight in lean mice, only a nonsignificant tendency of a specific effect of increased loading was observed in the lean mice. These findings demonstrate that the gravitostat regulates fat mass in obese mice, whereas leptin regulates fat mass only in lean mice with low endogenous serum leptin levels. We propose that activation of the gravitostat primarily protects against obesity, whereas low levels of leptin protect against undernutrition.
- Published
- 2018
- Full Text
- View/download PDF
19. The Bone Sparing Effects of 2-Methoxyestradiol Are Mediated via Estrogen Receptor-α in Male Mice.
- Author
-
Eriksson AL, Wilhelmson AS, Fagman JB, Ryberg H, Koskela A, Tuukkanen J, Tivesten Å, and Ohlsson C
- Subjects
- 2-Methoxyestradiol, Animals, Body Weight drug effects, Bone Density drug effects, Cancellous Bone drug effects, Cancellous Bone metabolism, Cortical Bone drug effects, Cortical Bone metabolism, Estradiol blood, Estradiol metabolism, Estradiol pharmacology, Female, Male, Mice, Mice, Knockout, Orchiectomy, Organ Size drug effects, Receptors, Estrogen genetics, Testosterone blood, Tomography, X-Ray Computed, X-Ray Microtomography, Bone and Bones drug effects, Bone and Bones metabolism, Estradiol analogs & derivatives, Receptors, Estrogen metabolism
- Abstract
2-Methoxyestradiol (2ME2), a metabolite of 17β-estradiol (E2), exerts bone sparing effects in animal models. We hypothesized that the underlying mechanism is back conversion of 2ME2 to E2, which subsequently acts via estrogen receptor (ER)α. We measured serum E2 levels in orchidectomized wild-type (WT) mice treated with 2ME2 66.6 μg/d or placebo. In placebo-treated animals, E2 was below the detection limit. In 2ME2-treated mice, the serum E2 level was 4.97 ± 0.68 pg/mL. This corresponds to the level found in diesterus in cycling female mice. Next, we investigated bone parameters in orchidectomized WT and ERα knockout mice treated with 2ME2 or placebo for 35 days. 2ME2 (6.66 μg/d) preserved trabecular and cortical bone in WT mice. Trabecular volumetric-bone mineral density was 64 ± 20%, and trabecular bone volume/total volume was 60 ± 20% higher in the metaphyseal region of the femur in the 2ME2 group, compared with placebo (P < .01). Both trabecular number and trabecular thickness were increased (P < .01). Cortical bone mineral content in the diaphyseal region of the femur was 31 ± 3% higher in the 2ME2 group, compared with placebo (P < .001). This was due to larger cortical area (P < .001). Three-point bending showed an increased bone strength in WT 2ME2-treated animals compared with placebo (maximum load [Fmax] +19±5% in the 2ME2 group, P < .05). Importantly, no bone parameter was affected by 2ME2 treatment in ERα knockout mice. In conclusion, 2ME2 treatment of orchidectomized mice results in increased serum E2. ERα mediates the bone sparing effects of 2ME2. The likely mediator of this effect is E2 resulting from back conversion of 2ME2.
- Published
- 2016
- Full Text
- View/download PDF
20. The Hydroxysteroid (17β) Dehydrogenase Family Gene HSD17B12 Is Involved in the Prostaglandin Synthesis Pathway, the Ovarian Function, and Regulation of Fertility.
- Author
-
Kemiläinen H, Adam M, Mäki-Jouppila J, Damdimopoulou P, Damdimopoulos AE, Kere J, Hovatta O, Laajala TD, Aittokallio T, Adamski J, Ryberg H, Ohlsson C, Strauss L, and Poutanen M
- Subjects
- 17-Hydroxysteroid Dehydrogenases genetics, Animals, Arachidonic Acid metabolism, Estrous Cycle, Female, Gonadal Steroid Hormones metabolism, Humans, Male, Meiosis, Mice, Inbred C57BL, Oogenesis, Ovulation, Random Allocation, 17-Hydroxysteroid Dehydrogenases metabolism, Fertility, Ovary physiology, Prostaglandins biosynthesis
- Abstract
The hydroxysteroid (17beta) dehydrogenase (HSD17B)12 gene belongs to the hydroxysteroid (17β) dehydrogenase superfamily, and it has been implicated in the conversion of estrone to estradiol as well as in the synthesis of arachidonic acid (AA). AA is a precursor of prostaglandins, which are involved in the regulation of female reproduction, prompting us to study the role of HSD17B12 enzyme in the ovarian function. We found a broad expression of HSD17B12 enzyme in both human and mouse ovaries. The enzyme was localized in the theca interna, corpus luteum, granulosa cells, oocytes, and surface epithelium. Interestingly, haploinsufficiency of the HSD17B12 gene in female mice resulted in subfertility, indicating an important role for HSD17B12 enzyme in the ovarian function. In line with significantly increased length of the diestrous phase, the HSD17B
+/- females gave birth less frequently than wild-type females, and the litter size of HSD17B12+/- females was significantly reduced. Interestingly, we observed meiotic spindle formation in immature follicles, suggesting defective meiotic arrest in HSD17B12+/- ovaries. The finding was further supported by transcriptome analysis showing differential expression of several genes related to the meiosis. In addition, polyovular follicles and oocytes trapped inside the corpus luteum were observed, indicating a failure in the oogenesis and ovulation, respectively. Intraovarian concentrations of steroid hormones were normal in HSD17B12+/- females, whereas the levels of AA and its metabolites (6-keto prostaglandin F1alpha, prostaglandin D2 , prostaglandin E2 , prostaglandin F2α , and thromboxane B2 ) were decreased. In conclusion, our study demonstrates that HSD17B12 enzyme plays an important role in female fertility through its role in AA metabolism.- Published
- 2016
- Full Text
- View/download PDF
21. Suppression of Experimental Arthritis and Associated Bone Loss by a Tissue-Selective Estrogen Complex.
- Author
-
Andersson A, Bernardi AI, Nurkkala-Karlsson M, Stubelius A, Grahnemo L, Ohlsson C, Carlsten H, and Islander U
- Subjects
- Animals, Arthritis, Experimental immunology, Arthritis, Rheumatoid immunology, Autoantibodies immunology, Bone Diseases, Metabolic immunology, Cytokines immunology, Female, Femur diagnostic imaging, Femur drug effects, Immunoglobulin G drug effects, Immunoglobulin G immunology, Interleukin-6 immunology, Mice, Ovariectomy, Radiography, Tibia diagnostic imaging, Tibia drug effects, Arthritis, Experimental diagnostic imaging, Arthritis, Rheumatoid diagnostic imaging, Autoantibodies drug effects, Bone Density drug effects, Bone Diseases, Metabolic diagnostic imaging, Cytokines drug effects, Estradiol pharmacology, Estrogens pharmacology, Indoles pharmacology, Selective Estrogen Receptor Modulators pharmacology
- Abstract
In addition to the systemic inflammation present in rheumatoid arthritis (RA), decreased estradiol levels in postmenopausal RA patients further accelerate bone loss in these patients. The tissue-selective estrogen complex (TSEC), an estrogen combined with a selective estrogen receptor modulator, is a new hormone replacement therapy option. The first approved TSEC, containing conjugated estrogens and bazedoxifene (BZA), reduces menopausal symptoms and prevents osteoporosis with an improved safety profile compared with conventional hormone replacement therapy. Previous studies have shown that estrogens strongly inhibit experimental arthritis whereas BZA is mildly suppressive. In this study the antiarthritic potential of combined BZA and estradiol is explored for the first time. Female ovariectomized DBA/1 mice were subjected to collagen-induced arthritis, an experimental postmenopausal RA model, and treated with BZA, 17β-estradiol (E2), combined BZA and E2 (BZA/E2), or vehicle. BZA/E2 suppressed arthritis severity and frequency, synovitis, and joint destruction, equally efficient as E2 alone. Unwanted estrogenic proliferative effects on the endometrium were blocked by the addition of BZA, determined by collecting uterine weights. Bone mineral density was measured by peripheral quantitative computed tomography, and all treatments protected collagen-induced arthritis mice from both trabecular and cortical bone loss. Moreover, BZA/E2, but not E2 alone, inhibited preosteoclast formation and reduced serum anticollagen type II antibodies. In conclusion, a TSEC, herein combined BZA/E2, suppresses experimental arthritis and prevents associated bone loss as efficiently as E2 alone but with minimal uterine effects, highlighting the need for clinical trials that evaluate the addition of a TSEC to conventional postmenopausal RA treatment.
- Published
- 2016
- Full Text
- View/download PDF
22. Enzalutamide Reduces the Bone Mass in the Axial But Not the Appendicular Skeleton in Male Mice.
- Author
-
Wu J, Movérare-Skrtic S, Börjesson AE, Lagerquist MK, Sjögren K, Windahl SH, Koskela A, Grahnemo L, Islander U, Wilhelmson AS, Tivesten Å, Tuukkanen J, and Ohlsson C
- Subjects
- Absorptiometry, Photon, Animals, Benzamides, Femur diagnostic imaging, Lumbar Vertebrae diagnostic imaging, Male, Mice, Nitriles, Orchiectomy, Organ Size drug effects, Phenylthiohydantoin pharmacology, Spine diagnostic imaging, Spine drug effects, Tibia diagnostic imaging, X-Ray Microtomography, Androgen Receptor Antagonists pharmacology, Antineoplastic Agents, Hormonal pharmacology, Bone Density drug effects, Bone Remodeling drug effects, Femur drug effects, Lumbar Vertebrae drug effects, Phenylthiohydantoin analogs & derivatives, Tibia drug effects
- Abstract
Testosterone is a crucial regulator of the skeleton, but the role of the androgen receptor (AR) for the maintenance of the adult male skeleton is unclear. In the present study, the role of the AR for bone metabolism and skeletal growth after sexual maturation was evaluated by means of the drug enzalutamide, which is a new AR antagonist used in the treatment of prostate cancer patients. Nine-week-old male mice were treated with 10, 30, or 100 mg/kg·d of enzalutamide for 21 days or were surgically castrated and were compared with vehicle-treated gonadal intact mice. Although orchidectomy reduced the cortical bone thickness and trabecular bone volume fraction in the appendicular skeleton, these parameters were unaffected by enzalutamide. In contrast, both enzalutamide and orchidectomy reduced the bone mass in the axial skeleton as demonstrated by a reduced lumbar spine areal bone mineral density (P < .001) and trabecular bone volume fraction in L5 vertebrae (P < .001) compared with vehicle-treated gonadal intact mice. A compression test of the L5 vertebrae revealed that the mechanical strength in the axial skeleton was significantly reduced by enzalutamide (maximal load at failure -15.3% ± 3.5%; P < .01). The effects of enzalutamide in the axial skeleton were associated with a high bone turnover. In conclusion, enzalutamide reduces the bone mass in the axial but not the appendicular skeleton in male mice after sexual maturation. We propose that the effect of testosterone on the axial skeleton in male mice is mainly mediated via the AR.
- Published
- 2016
- Full Text
- View/download PDF
23. Estrogen Therapy Delays Autoimmune Diabetes and Promotes the Protective Efficiency of Natural Killer T-Cell Activation in Female Nonobese Diabetic Mice.
- Author
-
Gourdy P, Bourgeois EA, Levescot A, Pham L, Riant E, Ahui ML, Damotte D, Gombert JM, Bayard F, Ohlsson C, Arnal JF, and Herbelin A
- Subjects
- Animals, Autoimmune Diseases drug therapy, Autoimmune Diseases immunology, Autoimmune Diseases metabolism, Cytokines blood, Cytokines metabolism, Diabetes Mellitus, Type 1 drug therapy, Diabetes Mellitus, Type 1 immunology, Diabetes Mellitus, Type 1 metabolism, Drug Implants, Estradiol administration & dosage, Estrogen Replacement Therapy, Estrogens administration & dosage, Female, Galactosylceramides agonists, Galactosylceramides pharmacology, Galactosylceramides therapeutic use, Immune Tolerance drug effects, Killer Cells, Natural immunology, Killer Cells, Natural metabolism, Lymphocyte Depletion adverse effects, Mice, Inbred C57BL, Mice, Inbred NOD, Mice, Mutant Strains, Ovariectomy adverse effects, Prediabetic State drug therapy, Prediabetic State immunology, Prediabetic State metabolism, T-Lymphocytes, Regulatory drug effects, T-Lymphocytes, Regulatory immunology, T-Lymphocytes, Regulatory metabolism, Autoimmune Diseases prevention & control, Diabetes Mellitus, Type 1 prevention & control, Estradiol therapeutic use, Estrogens therapeutic use, Killer Cells, Natural drug effects, Lymphocyte Activation drug effects, Prediabetic State prevention & control
- Abstract
Therapeutic strategies focused on restoring immune tolerance remain the main avenue to prevent type 1 diabetes (T1D). Because estrogens potentiate FoxP3+ regulatory T cells (Treg) and invariant natural killer T (iNKT) cells, two regulatory lymphocyte populations that are functionally deficient in nonobese diabetic (NOD) mice, we investigated whether estradiol (E2) therapy influences the course of T1D in this model. To this end, female NOD mice were sc implanted with E2- or placebo-delivering pellets to explore the course of spontaneous and cyclophosphamide-induced diabetes. Treg-depleted and iNKT-cell-deficient (Jα18(-/-)) NOD mice were used to assess the respective involvement of these lymphocyte populations in E2 effects. Early E2 administration (from 4 wk of age) was found to preserve NOD mice from both spontaneous and cyclophosphamide-induced diabetes, and a complete protection was also observed throughout treatment when E2 treatment was initiated after the onset of insulitis (from 12 wk of age). This delayed E2 treatment remained fully effective in Treg-depleted mice but failed to entirely protect Jα18(-/-) mice. Accordingly, E2 administration was shown to restore the cytokine production of iNKT cells in response to in vivo challenge with the cognate ligand α-galactosylceramide. Finally, transient E2 administration potentiated the previously described protective action of α-galactosylceramide treatment in NOD females. This study provides original evidence that E2 therapy strongly protects NOD mice from T1D and reveals the estrogen/iNKT cell axis as a new effective target to counteract diabetes onset at the stage of insulitis. Estrogen-based therapy should thus be considered for T1D prevention.
- Published
- 2016
- Full Text
- View/download PDF
24. Measurement of a Comprehensive Sex Steroid Profile in Rodent Serum by High-Sensitive Gas Chromatography-Tandem Mass Spectrometry.
- Author
-
Nilsson ME, Vandenput L, Tivesten Å, Norlén AK, Lagerquist MK, Windahl SH, Börjesson AE, Farman HH, Poutanen M, Benrick A, Maliqueo M, Stener-Victorin E, Ryberg H, and Ohlsson C
- Subjects
- Androstenedione analysis, Androstenedione blood, Animals, Dehydroepiandrosterone analysis, Dehydroepiandrosterone blood, Dihydrotestosterone analysis, Dihydrotestosterone blood, Estradiol analysis, Estradiol blood, Estrone analysis, Estrone blood, Female, Gonadal Steroid Hormones analysis, Mice, Progesterone analysis, Progesterone blood, Rats, Testosterone analysis, Testosterone blood, Gas Chromatography-Mass Spectrometry methods, Gonadal Steroid Hormones blood, Tandem Mass Spectrometry methods
- Abstract
Accurate measurement of sex steroid concentrations in rodent serum is essential to evaluate mouse and rat models for sex steroid-related disorders. The aim of the present study was to develop a sensitive and specific gas chromatography-tandem mass spectrometry (GC-MS/MS) method to assess a comprehensive sex steroid profile in rodent serum. A major effort was invested in reaching an exceptionally high sensitivity for measuring serum estradiol concentrations. We established a GC-MS/MS assay with a lower limit of detection for estradiol, estrone, T, DHT, progesterone, androstenedione, and dehydroepiandrosterone of 0.3, 0.5, 4.0, 1.6, 8, 4.0, and 50 pg/mL, respectively, whereas the corresponding values for the lower limit of quantification were 0.5, 0.5, 8, 2.5, 74, 12, and 400 pg/mL, respectively. Calibration curves were linear, intra- and interassay coefficients of variation were low, and accuracy was excellent for all analytes. The established assay was used to accurately measure a comprehensive sex steroid profile in female rats and mice according to estrous cycle phase. In addition, we characterized the impact of age, sex, gonadectomy, and estradiol treatment on serum concentrations of these sex hormones in mice. In conclusion, we have established a highly sensitive and specific GC-MS/MS method to assess a comprehensive sex steroid profile in rodent serum in a single run. This GC-MS/MS assay has, to the best of our knowledge, the best detectability reported for estradiol. Our method therefore represents an ideal tool to characterize sex steroid metabolism in a variety of sex steroid-related rodent models and in human samples with low estradiol levels.
- Published
- 2015
- Full Text
- View/download PDF
25. Androgens regulate bone marrow B lymphopoiesis in male mice by targeting osteoblast-lineage cells.
- Author
-
Wilhelmson AS, Stubelius A, Börjesson AE, Wu J, Stern A, Malin S, Mårtensson IL, Ohlsson C, Carlsten H, and Tivesten Å
- Subjects
- Animals, B-Lymphocytes cytology, Biomarkers blood, Bone Marrow metabolism, Cell Lineage, Collagen Type I blood, Male, Mice, Mice, Knockout, Osteocalcin blood, Peptides blood, Receptors, Androgen genetics, Spleen metabolism, Androgens metabolism, B-Lymphocytes metabolism, Lymphopoiesis physiology, Osteoblasts metabolism, Receptors, Androgen metabolism
- Abstract
Testosterone has profound immune-modulatory actions, which may be important for the sexual dimorphism in immune-related disorders, such as autoimmune diseases. A well-known effect of androgens is inhibition of bone marrow B lymphopoiesis; however, a plausible target cell for this effect has not yet been presented. The aim of this study was to determine the target cell for androgen-mediated regulation of bone marrow B lymphopoiesis in males. We confirm higher number of bone marrow B cells in male mice with global inactivation of the androgen receptor (AR) and these global AR knockout (G-ARKO) mice had increased number of B cell precursors from the pro-B stage. Because osteoblast-lineage cells are known to support B lymphopoiesis at the pro-B stage, we investigated the effect on B lymphopoiesis in osteoblast-lineage cell-specific ARKO (O-ARKO) mice; O-ARKO mice had increased number of B cells in the bone marrow, and the number of B cell precursors was increased from the pro-B stage, demonstrating that O-ARKO mimics the bone marrow B lymphopoiesis pattern of G-ARKO mice. By contrast, O-ARKO mice displayed only minor changes in B cell numbers in the splenic compartment compared with G-ARKO. Further, O-ARKO mice had moderately reduced number of bone trabeculae in the vertebrae, whereas cortical bone was unaffected. In conclusion, androgens exert inhibitory effects on bone marrow B lymphopoiesis in males by targeting the AR in osteoblast-lineage cells. The identification of the likely target cell for androgen-mediated regulation of bone marrow B lymphopoiesis will contribute to elucidation of the mechanisms by which androgens modulate immune-related disorders.
- Published
- 2015
- Full Text
- View/download PDF
26. Combined treatment with GH and IGF-I: additive effect on cortical bone mass but not on linear bone growth in female rats.
- Author
-
Sundström K, Cedervall T, Ohlsson C, Camacho-Hübner C, and Sävendahl L
- Subjects
- Animals, Body Weight drug effects, Bone and Bones metabolism, Drug Therapy, Combination, Dwarfism, Pituitary drug therapy, Female, Glucose metabolism, Growth Hormone blood, Human Growth Hormone therapeutic use, Insulin-Like Growth Factor Binding Protein 3 blood, Insulin-Like Growth Factor I metabolism, Insulin-Like Growth Factor I therapeutic use, Pituitary Gland drug effects, Rats, Sprague-Dawley, Bone Development drug effects, Bone and Bones drug effects, Human Growth Hormone pharmacology, Insulin-Like Growth Factor I pharmacology
- Abstract
The growth-promoting effect of combined therapy with GH and IGF-I in normal rats is not known. We therefore investigated the efficacy of treatment with recombinant human (rh)GH and/or rhIGF-I on longitudinal bone growth and bone mass in intact, prepubertal, female Sprague-Dawley rats. rhGH was injected twice daily sc (5 mg/kg·d) and rhIGF-I continuously infused sc (2.2 or 4.4 mg/kg·d) for 28 days. Longitudinal bone growth was monitored by weekly x-rays of tibiae and nose-anus length measurements, and tibial growth plate histomorphology was analyzed. Bone mass was evaluated by peripheral quantitative computed tomography. In addition, serum levels of IGF-I, rat GH, acid labile subunit, IGF binding protein-3, 150-kDa ternary complex formation, and markers of bone formation and degradation were measured. Monotherapy with rhGH was more effective than rhIGF-I (4.4 mg/kg·d) to increase tibia and nose-anus length, whereas combined therapy did not further increase tibia, or nose-anus, lengths or growth plate height. In contrast, combined rhGH and rhIGF-I (4.4 mg/kg·d) therapy had an additive stimulatory effect on cortical bone mass vs rhGH alone. Combined treatment with rhGH and rhIGF-I resulted in markedly higher serum IGF-I concentrations vs rhGH alone but did not compromise the endogenous secretion of GH. We conclude that rhIGF-I treatment augments cortical bone mass but does not further improve bone growth in rhGH-treated young, intact, female rats.
- Published
- 2014
- Full Text
- View/download PDF
27. Role of androgen and estrogen receptors for the action of dehydroepiandrosterone (DHEA).
- Author
-
Engdahl C, Lagerquist MK, Stubelius A, Andersson A, Studer E, Ohlsson C, Westberg L, Carlsten H, and Forsblad-d'Elia H
- Subjects
- Adjuvants, Immunologic chemistry, Androgens metabolism, Animals, Bone Density, Bone and Bones metabolism, Dehydroepiandrosterone pharmacology, Dihydrotestosterone metabolism, Estrogens metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Phenotype, Seminal Vesicles metabolism, Submandibular Gland metabolism, Thymus Gland metabolism, Dehydroepiandrosterone physiology, Gene Expression Regulation, Receptors, Androgen metabolism, Receptors, Estrogen metabolism, Signal Transduction
- Abstract
Dehydroepiandrosterone (DHEA) is an abundant steroid hormone, and its mechanism of action is yet to be determined. The aim of this study was to elucidate the importance of androgen receptors (ARs) and estrogen receptors (ERs) for DHEA function. Orchidectomized C57BL/6 mice were treated with DHEA, DHT, 17β-estradiol-3-benzoate (E2), or vehicle. Orchidectomized AR-deficient (ARKO) mice and wild-type (WT) littermates were treated with DHEA or vehicle for 2.5 weeks. At termination, bone mineral density (BMD) was evaluated, thymus and seminal vesicles were weighted, and submandibular glands (SMGs) were histologically examined. To evaluate the in vivo ER activation of the classical estrogen signaling pathway, estrogen response element reporter mice were treated with DHEA, DHT, E2, or vehicle, and a reporter gene was investigated in different sex steroid-sensitive organs after 24 hours. DHEA treatment increased trabecular BMD and thymic atrophy in both WT and ARKO mice. In WT mice, DHEA induced enlargement of glands in the SMGs, whereas this effect was absent in ARKO mice. Furthermore, DHEA was able to induce activation of classical estrogen signaling in bone, thymus, and seminal vesicles but not in the SMGs. In summary, the DHEA effects on trabecular BMD and thymus do not require signaling via AR and DHEA can activate the classical estrogen signaling in these organs. In contrast, DHEA induction of gland size in the SMGs is dependent on AR and does not involve classical estrogen signaling. Thus, both ERs and ARs are involved in mediating the effects of DHEA in an organ-dependent manner.
- Published
- 2014
- Full Text
- View/download PDF
28. Androgen receptor-dependent and independent atheroprotection by testosterone in male mice.
- Author
-
Bourghardt J, Wilhelmson AS, Alexanderson C, De Gendt K, Verhoeven G, Krettek A, Ohlsson C, and Tivesten A
- Subjects
- Animals, Aorta drug effects, Aortic Diseases genetics, Aortic Diseases prevention & control, Apolipoproteins E genetics, Apolipoproteins E metabolism, Atherosclerosis genetics, Atherosclerosis prevention & control, Blood Pressure physiology, Cytokines blood, Lipids blood, Male, Mice, Mice, Knockout, Orchiectomy, Receptors, Androgen genetics, Reverse Transcriptase Polymerase Chain Reaction, Statistics, Nonparametric, Testosterone pharmacology, Aorta metabolism, Aortic Diseases metabolism, Atherosclerosis metabolism, Receptors, Androgen metabolism, Testosterone metabolism
- Abstract
The atheroprotective effect of testosterone is thought to require aromatization of testosterone to estradiol, but no study has adequately addressed the role of the androgen receptor (AR), the major pathway for the physiological effects of testosterone. We used AR knockout (ARKO) mice on apolipoprotein E-deficient background to study the role of the AR in testosterone atheroprotection in male mice. Because ARKO mice are testosterone deficient, we sham operated or orchiectomized (Orx) the mice before puberty, and Orx mice were supplemented with placebo or a physiological testosterone dose. From 8 to 16 wk of age, the mice consumed a high-fat diet. In the aortic root, ARKO mice showed increased atherosclerotic lesion area (+80%, P < 0.05). Compared with placebo, testosterone reduced lesion area both in Orx wild-type (WT) mice (by 50%, P < 0.001) and ARKO mice (by 24%, P < 0.05). However, lesion area was larger in testosterone-supplemented ARKO compared with testosterone-supplemented WT mice (+57%, P < 0.05). In WT mice, testosterone reduced the presence of a necrotic core in the plaque (80% among placebo-treated vs. 12% among testosterone-treated mice; P < 0.05), whereas there was no significant effect in ARKO mice (P = 0.20). In conclusion, ARKO mice on apolipoprotein E-deficient background display accelerated atherosclerosis. Testosterone treatment reduced atherosclerosis in both WT and ARKO mice. However, the effect on lesion area and complexity was more pronounced in WT than in ARKO mice, and lesion area was larger in ARKO mice even after testosterone supplementation. These results are consistent with an AR-dependent as well as an AR-independent component of testosterone atheroprotection in male mice.
- Published
- 2010
- Full Text
- View/download PDF
29. Deletion of the G protein-coupled receptor 30 impairs glucose tolerance, reduces bone growth, increases blood pressure, and eliminates estradiol-stimulated insulin release in female mice.
- Author
-
Mårtensson UE, Salehi SA, Windahl S, Gomez MF, Swärd K, Daszkiewicz-Nilsson J, Wendt A, Andersson N, Hellstrand P, Grände PO, Owman C, Rosen CJ, Adamo ML, Lundquist I, Rorsman P, Nilsson BO, Ohlsson C, Olde B, and Leeb-Lundberg LM
- Subjects
- Animals, Female, Gene Deletion, Glucose Tolerance Test veterinary, Insulin Secretion, Islets of Langerhans metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Pregnancy, RNA, Messenger metabolism, Receptors, Estrogen, Receptors, G-Protein-Coupled metabolism, Receptors, G-Protein-Coupled physiology, Sex Characteristics, Tissue Distribution, Blood Pressure genetics, Bone Development genetics, Estradiol pharmacology, Glucose Intolerance genetics, Insulin metabolism, Receptors, G-Protein-Coupled genetics
- Abstract
In vitro studies suggest that the G protein-coupled receptor (GPR) 30 is a functional estrogen receptor. However, the physiological role of GPR30 in vivo is unknown, and it remains to be determined whether GPR30 is an estrogen receptor also in vivo. To this end, we studied the effects of disrupting the GPR30 gene in female and male mice. Female GPR30((-/-)) mice had hyperglycemia and impaired glucose tolerance, reduced body growth, increased blood pressure, and reduced serum IGF-I levels. The reduced growth correlated with a proportional decrease in skeletal development. The elevated blood pressure was associated with an increased vascular resistance manifested as an increased media to lumen ratio of the resistance arteries. The hyperglycemia and impaired glucose tolerance in vivo were associated with decreased insulin expression and release in vivo and in vitro in isolated pancreatic islets. GPR30 is expressed in islets, and GPR30 deletion abolished estradiol-stimulated insulin release both in vivo in ovariectomized adult mice and in vitro in isolated islets. Our findings show that GPR30 is important for several metabolic functions in female mice, including estradiol-stimulated insulin release.
- Published
- 2009
- Full Text
- View/download PDF
30. Estrogen up-regulates hepatic expression of suppressors of cytokine signaling-2 and -3 in vivo and in vitro.
- Author
-
Leong GM, Moverare S, Brce J, Doyle N, Sjögren K, Dahlman-Wright K, Gustafsson JA, Ho KK, Ohlsson C, and Leung KC
- Subjects
- Animals, Carcinoma, Hepatocellular, Cell Line, Tumor, Estrogen Receptor alpha genetics, Estrogen Receptor beta genetics, Female, Growth Hormone pharmacology, Humans, In Vitro Techniques, Liver Neoplasms, Mice, Mice, Inbred C57BL, Mice, Knockout, Promoter Regions, Genetic physiology, Signal Transduction physiology, Suppressor of Cytokine Signaling 3 Protein, Suppressor of Cytokine Signaling Proteins, Transcription Factors genetics, Up-Regulation drug effects, DNA-Binding Proteins genetics, Estradiol pharmacology, Liver physiology, Repressor Proteins genetics, Signal Transduction drug effects, Trans-Activators genetics
- Abstract
Suppressors of cytokine signaling (SOCS) are important negative regulators of cytokine action. We recently reported that estrogen stimulates SOCS-2 expression and inhibits GH signaling in kidney cells. The effects of estrogen on SOCS expression in other tissues are unclear. The aim of this study was to investigate in vivo and in vitro whether estrogen affected SOCS expression in the liver, a major target organ of GH. The in vivo hepatic effects of estrogen on ovariectomized mice lacking estrogen receptor (ER)-alpha, ERbeta, or both and their wild-type littermates were examined by DNA microarray analysis. In vitro, the effects of estrogen on SOCS expression in human hepatoma cells were examined by reverse transcription quantitative PCR. Long-term (3 wk) estrogen treatment induced a 2- to 3-fold increase in hepatic expression of SOCS-2 and -3 in wild-type and ERbeta knockout mice but not in those lacking ERalpha or both ER subtypes. Short-term treatment (at 24 h) increased the mRNA level of SOCS-3 but not SOCS-2. In cultured hepatoma cells, estrogen increased SOCS-2 and -3 mRNA levels by 2-fold in a time- and dose-dependent manner (P < 0.05). Estrogen induced murine SOCS-3 promoter activity by 2-fold (P < 0.05) in constructs containing a region between nucleotides -1862 and -855. Moreover, estrogen and GH had additive effects on the SOCS-3 promoter activity. In summary, estrogen, via ERalpha, up-regulated hepatic expression of SOCS-2 and -3, probably through transcriptional activation. This indicates a novel mechanism of estrogen regulation of cytokine action.
- Published
- 2004
- Full Text
- View/download PDF
31. Transgenic mice expressing fibroblast growth factor 23 under the control of the alpha1(I) collagen promoter exhibit growth retardation, osteomalacia, and disturbed phosphate homeostasis.
- Author
-
Larsson T, Marsell R, Schipani E, Ohlsson C, Ljunggren O, Tenenhouse HS, Jüppner H, and Jonsson KB
- Subjects
- Animals, Bone and Bones pathology, Bone and Bones physiology, Calcification, Physiologic, Calcium blood, Calcium urine, Fibroblast Growth Factor-23, Growth Disorders pathology, Growth Disorders physiopathology, Homeostasis physiology, Humans, Kidney cytology, Kidney physiology, Mice, Mice, Inbred C57BL, Mice, Inbred CBA, Mice, Transgenic, Osteomalacia pathology, Osteomalacia physiopathology, Parathyroid Glands pathology, Parathyroid Glands physiology, Phenotype, Phosphates urine, Promoter Regions, Genetic, Vitamin D metabolism, Collagen Type I genetics, Fibroblast Growth Factors genetics, Fibroblast Growth Factors metabolism, Growth Disorders metabolism, Osteomalacia metabolism, Phosphates blood
- Abstract
Mutations in the fibroblast growth factor 23 gene, FGF23, cause autosomal dominant hypophosphatemic rickets (ADHR). The gene product, FGF-23, is produced by tumors from patients with oncogenic osteomalacia (OOM), circulates at increased levels in most patients with X-linked hypophosphatemia (XLH) and is phosphaturic when injected into rats or mice, suggesting involvement in the regulation of phosphate (Pi) homeostasis. To better define the precise role of FGF-23 in maintaining Pi balance and bone mineralization, we generated transgenic mice that express wild-type human FGF-23, under the control of the alpha1(I) collagen promoter, in cells of the osteoblastic lineage. At 8 wk of age, transgenic mice were smaller (body weight = 17.5 +/- 0.57 vs. 24.3 +/- 0.37 g), exhibited decreased serum Pi concentrations (1.91 +/- 0.27 vs. 2.75 +/- 0.22 mmol/liter) and increased urinary Pi excretion when compared with wild-type littermates. The serum concentrations of human FGF-23 (undetectable in wild-type mice) was markedly elevated in transgenic mice (>7800 reference units/ml). Serum PTH levels were increased in transgenic mice (231 +/- 62 vs. 139 +/- 44 pg/ml), whereas differences in calcium and 1,25-dihydroxyvitamin D were not apparent. Expression of Npt2a, the major renal Na(+)/Pi cotransporter, as well as Npt1 and Npt2c mRNAs, was significantly decreased in the kidneys of transgenic mice. Histology of tibiae displayed a disorganized and widened growth plate and peripheral quantitative computerized tomography analysis revealed reduced bone mineral density in transgenic mice. The data indicate that FGF-23 induces phenotypic changes in mice resembling those of patients with ADHR, OOM, and XLH and that FGF-23 is an important determinant of Pi homeostasis and bone mineralization.
- Published
- 2004
- Full Text
- View/download PDF
32. Liver-derived insulin-like growth factor-I is involved in the regulation of blood pressure in mice.
- Author
-
Tivesten A, Bollano E, Andersson I, Fitzgerald S, Caidahl K, Sjögren K, Skøtt O, Liu JL, Mobini R, Isaksson OG, Jansson JO, Ohlsson C, Bergström G, and Isgaard J
- Subjects
- Acetylcholine pharmacology, Animals, Body Weight, Cardiac Output, Creatinine blood, Creatinine urine, Echocardiography, Endothelin-1 genetics, Insulin-Like Growth Factor I deficiency, Insulin-Like Growth Factor I genetics, Mesenteric Arteries drug effects, Mice, Mice, Inbred C57BL, Mice, Inbred CBA, Mice, Knockout, Myocardium chemistry, Nitric Oxide Synthase antagonists & inhibitors, Nitric Oxide Synthase genetics, Nitric Oxide Synthase Type II, Nitric Oxide Synthase Type III, Norepinephrine analysis, Organ Size, RNA, Messenger analysis, Renin blood, Vasodilation drug effects, Blood Pressure physiology, Insulin-Like Growth Factor I physiology, Liver chemistry
- Abstract
IGF-I has been suggested to be of importance for cardiovascular structure and function, but the relative role of locally produced and liver-derived endocrine IGF-I remains unclear. Using the Cre-LoxP recombination system, we have previously created transgenic mice with a liver-specific, inducible IGF-I knockout (LI-IGF-I-/-). To examine the role of liver-derived IGF-I in cardiovascular physiology, liver-derived IGF-I was inactivated at 4 wk of age, resulting in a 79% reduction of serum IGF-I levels. At 4 months of age, systolic blood pressure (BP) was increased in LI-IGF-I-/- mice. Echocardiography showed increased posterior wall thickness in combination with decreased stroke volume and cardiac output, whereas other systolic variables were unchanged, suggesting that these cardiac effects were secondary to increased peripheral resistance. Acute nitric oxide-synthase inhibition increased systolic BP more in LI-IGF-I-/- mice than in control mice. LI-IGF-I-/- mice showed impaired acetylcholine-induced vasorelaxation in mesenteric resistance vessels and increased levels of endothelin-1 mRNA in aorta. Thus, the increased peripheral resistance in LI-IGF-I-/- mice might be attributable to endothelial dysfunction associated with increased expression of endothelin-1 and impaired vasorelaxation of resistance vessels. In conclusion, our findings suggest that liver-derived IGF-I is involved in the regulation of BP in mice.
- Published
- 2002
- Full Text
- View/download PDF
33. Liver-derived IGF-I regulates GH secretion at the pituitary level in mice.
- Author
-
Wallenius K, Sjögren K, Peng XD, Park S, Wallenius V, Liu JL, Umaerus M, Wennbo H, Isaksson O, Frohman L, Kineman R, Ohlsson C, and Jansson JO
- Subjects
- Animals, Female, Growth Hormone blood, Growth Hormone genetics, Growth Hormone-Releasing Hormone pharmacology, Hypothalamus metabolism, Insulin-Like Growth Factor I analysis, Insulin-Like Growth Factor I genetics, Liver anatomy & histology, Male, Mice, Mice, Knockout genetics, Neuropeptides physiology, Organ Size, Proteins genetics, RNA, Messenger metabolism, Receptors, Cell Surface physiology, Receptors, Prolactin genetics, Growth Hormone metabolism, Insulin-Like Growth Factor I physiology, Liver metabolism, Pituitary Gland metabolism
- Abstract
We have reported that liver-specific deletion of IGF-I in mice (LI-IGF-I-/-) results in decreased circulating IGF-I and increased GH levels. In the present study, we determined how elimination of hepatic IGF-I modifies the hypothalamic-pituitary GH axis to enhance GH secretion. The pituitary mRNA levels of GH releasing factor (GHRF) receptor and GH secretagogue (GHS) receptor were increased in LI-IGF-I-/- mice, and in line with this, their GH response to ip injections of GHRF and GHS was increased. Expression of mRNA for pituitary somatostatin receptors, hypothalamic GHRF, somatostatin, and neuropeptide Y was not altered in LI-IGF-I-/- mice, whereas hypothalamic IGF-I expression was increased. Changes in hepatic expression of major urinary protein and the PRL receptor in male LI-IGF-I-/- mice indicated an altered GH release pattern most consistent with enhanced GH trough levels. Liver weight was enhanced in LI-IGF-I-/- mice of both genders. In conclusion, loss of liver-derived IGF-I enhances GH release by increasing expression of pituitary GHRF and GHS receptors. The enhanced GH release in turn affects several liver parameters, in line with the existence of a pituitary-liver axis.
- Published
- 2001
- Full Text
- View/download PDF
34. Enhanced spontaneous locomotor activity in bovine GH transgenic mice involves peripheral mechanisms.
- Author
-
Bohlooly-Y M, Olsson B, Gritli-Linde A, Brusehed O, Isaksson OG, Ohlsson C, Söderpalm B, and Törnell J
- Subjects
- Animals, Cattle, Mice, Mice, Transgenic, Growth Hormone physiology, Motor Activity physiology
- Abstract
Clinical and experimental studies indicate a role for GH in mechanisms related to anhedonia/hedonia, psychic energy, and reward. Recently we showed that transgenic mice with general overexpression of bovine GH display increased spontaneous locomotor activity. In the present study, we investigated whether this behavioral change is owing to a direct action of GH in the central nervous system or to peripheral GH actions. A transgenic construct, containing the glial fibrillary acidic protein promoter directing specific expression of bovine GH to the central nervous system, was designed. The central nervous system-specific expression of bovine GH in the glial fibrillary acidic protein-bovine GH transgenic mice was confirmed, but no effect on spontaneous locomotor activity was observed. Serum bovine GH levels were increased in glial fibrillary acidic protein-bovine GH transgenic mice but clearly lower than in transgenic mice with general overexpression of bovine GH. In contrast to the transgenic mice with general overexpression of bovine GH, glial fibrillary acidic protein-bovine GH mice did not display any difference in serum IGF-I levels. The levels of free T(3) and the conversion of the free T(4) to free T(3) were only increased in transgenic mice with general overexpression of bovine GH, but serum corticosterone levels were similarly increased in both transgenic models. These results suggest that free T(3) and/or IGF-I, affecting dopamine and serotonin systems in the central nervous system, may mediate the enhanced locomotor activity observed in transgenic mice with general overexpression of bovine GH.
- Published
- 2001
- Full Text
- View/download PDF
35. Retarded liver growth in interleukin-6-deficient and tumor necrosis factor receptor-1-deficient mice.
- Author
-
Wallenius V, Wallenius K, Hisaoka M, Sandstedt J, Ohlsson C, Kopf M, and Jansson JO
- Subjects
- Aging physiology, Animals, Antigens, CD genetics, DNA metabolism, Growth Hormone pharmacology, Humans, Interleukin-6 genetics, Interleukin-6 pharmacology, Liver anatomy & histology, Liver drug effects, Mice, Mice, Knockout genetics, Organ Size physiology, Protein Isoforms deficiency, Protein Isoforms genetics, Proteins metabolism, Receptors, Tumor Necrosis Factor genetics, Receptors, Tumor Necrosis Factor, Type I, Reference Values, Tumor Necrosis Factor-alpha pharmacology, Interleukin-6 deficiency, Liver growth & development, Receptors, Tumor Necrosis Factor deficiency
- Abstract
The liver size in adult mammals is tightly regulated in relation to body weight, but the hormonal control of this is largely unknown. We investigated the roles of interleukin-6 (IL-6) and tumor necrosis factor (TNF) receptor-1 in the regulation of intact liver weight in adult mice. The relative liver wet and dry weights of older adult (5- to 10-month-old) IL-6 knockout (IL-6(-/-)) mice were decreased by 22-28%, and total contents of DNA and protein were decreased compared with those in age-matched wild-type mice. Weights of other visceral organs were unaffected. Older adult (6- to 8-month-old) TNF receptor-1 knockout (TNFR1(-/-)) mice displayed decreased relative liver weight. Treatment with a single injection of IL-6 increased liver wet and dry weights in IL-6(-/-) and wild-type mice, but not TNFR1(-/-) mice. Treatment with TNFalpha enhanced liver weight and DNA synthesis of nonparenchymal liver cells at 24 h in wild-type, but not IL-6(-/-), mice. At 48 h, TNFalpha induced DNA synthesis in nonparenchymal cells and hepatocytes of both wild-type and IL-6(-/-) mice. In conclusion, TNF receptor-1 stimulation and IL-6 production are both necessary for normal liver weight gain in older adult mice. The results of TNFalpha and IL-6 treatment further indicate that the effects of TNF receptor-1 and IL-6 depend on each other for full stimulation of liver growth.
- Published
- 2001
- Full Text
- View/download PDF
36. Dexamethasone impairs growth hormone (GH)-stimulated growth by suppression of local insulin-like growth factor (IGF)-I production and expression of GH- and IGF-I-receptor in cultured rat chondrocytes.
- Author
-
Jux C, Leiber K, Hügel U, Blum W, Ohlsson C, Klaus G, and Mehls O
- Subjects
- Animals, Cartilage cytology, Cell Division drug effects, Cells, Cultured, DNA biosynthesis, Epiphyses cytology, Epiphyses metabolism, Humans, Insulin-Like Growth Factor I metabolism, Rats, Rats, Sprague-Dawley, Receptors, Somatomedin metabolism, Receptors, Somatotropin genetics, Receptors, Somatotropin metabolism, Transcription, Genetic physiology, Cartilage metabolism, Dexamethasone pharmacology, Glucocorticoids pharmacology, Human Growth Hormone pharmacology, Insulin-Like Growth Factor I antagonists & inhibitors, Receptors, Somatomedin antagonists & inhibitors, Receptors, Somatotropin antagonists & inhibitors
- Abstract
Growth depression as a side effect of glucocorticoid therapy in childhood is partially mediated by alterations of the somatotropic hormone axis. The mechanisms of interaction between glucocorticoids and somatotropic hormones on the cellular and molecular level are poorly understood. In an experimental model of primary cultured rat growth plate chondrocytes, basal as well as GH (40 ng/ml) or insulin-like growth factor (IGF)-I (60 ng/ml)-stimulated growth was suppressed dose dependently (10(-l2)-10(-7)M) by dexamethasone (Dexa). An IGF-I antibody specifically and dose dependently inhibited the GH- but not the basic fibroblast growth factor (bFGF)-stimulated cell proliferation. GH increased the IGF-I concentration in conditioned serum-free culture medium; this was reversed by concomitant Dexa. Dexa time dependently suppressed the transcription of GH receptor (GHR) messenger RNA (mRNA) and down-regulated the basal and GH-stimulated expression of GHR. Whereas no suppressive effect on basal type I IGF-receptor (IGFR) was observed, Dexa blocked the IGF-I induced increase of IGF binding. These results were confirmed by GHR and IGFR immunostaining. We conclude that Dexa impairs the GH-induced stimulation of local secretion and paracrine action of IGF-I and reduces the homologous increase of IGFR and GHR expression. The above experiments give further insight on the interaction between GH and glucocorticoids on the cellular and molecular level of growth plate chondrocytes.
- Published
- 1998
- Full Text
- View/download PDF
37. p53 regulates insulin-like growth factor-I (IGF-I) receptor expression and IGF-I-induced tyrosine phosphorylation in an osteosarcoma cell line: interaction between p53 and Sp1.
- Author
-
Ohlsson C, Kley N, Werner H, and LeRoith D
- Subjects
- Humans, Mutation, Phosphorylation, Promoter Regions, Genetic, Receptor, IGF Type 1 genetics, Tumor Cells, Cultured, Tumor Suppressor Protein p53 physiology, Insulin-Like Growth Factor I pharmacology, Osteosarcoma metabolism, Receptor, IGF Type 1 analysis, Sp1 Transcription Factor physiology, Tyrosine metabolism
- Abstract
The insulin-like growth factor-I receptor (IGF-IR) is involved in tumorigenesis. The aim of the present study was to investigate whether the IGF-IR is a physiological target for p53 in osteosarcoma cells. The p53-induced regulation of IGF-IR levels was studied in a tetracycline-regulated expression system. When expressed in Saos-2, osteosarcoma cells that lack p53, wild-type p53 decreased, whereas mutated p53 increased IGF-IR expression, and IGF-I-induced tyrosine phosphorylation of the IGF-IR. Similarly, wild-type p53 decreased IGF-I-induced tyrosine phosphorylation of IRS-1. A functional and physical interaction between p53 and Sp1, in the regulation of the IGF-R, was studied in osteosarcoma cells. Expression of p53 decreased IGF-IR promoter activity, whereas no effect on promoter activity was seen by Sp1 expressed alone. However, Sp1 counteracted the inhibitory effect of p53 on IGF-IR promoter activity in a dose-dependent manner. Furthermore, wild-type and mutated p53 were coimmunoprecipitated with Sp1, indicating a physical interaction between p53 and Sp1. In conclusion, p53 regulates IGF-IR expression, as reflected by a reduction in IGF-IR protein and a parallel reduction in IGF-I-induced tyrosine phosphorylation of the IGF-IR and IRS-1 in an osteosarcoma cell line. These data indicate that the IGF-I receptor is a physiological target for p53 in osteosarcoma cells. Furthermore, data supporting an interaction between p53 and Sp1 in the regulation of the promoter activity of IGF-IR are presented.
- Published
- 1998
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.