Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Curr Osteoporos Rep ; 21(6): 825-841, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37707757

RESUMEN

PURPOSE OF REVIEW: To summarise the evidence regarding the effects of gender-affirming hormone therapy (GAHT) on bone health in transgender people, to identify key knowledge gaps and how these gaps can be addressed using preclinical rodent models. RECENT FINDINGS: Sex hormones play a critical role in bone physiology, yet there is a paucity of research regarding the effects of GAHT on bone microstructure and fracture risk in transgender individuals. The controlled clinical studies required to yield fracture data are unethical to conduct making clinically translatable preclinical research of the utmost importance. Novel genetic and surgical preclinical models have yielded significant mechanistic insight into the roles of sex steroids on skeletal integrity. Preclinical models of GAHT have the potential inform clinical approaches to preserve skeletal integrity and prevent fractures in transgender people undergoing GAHT. This review highlights the key considerations required to ensure the information gained from preclinical models of GAHT are informative.


Asunto(s)
Fracturas Óseas , Personas Transgénero , Humanos , Densidad Ósea , Hormonas
2.
Bone Res ; 12(1): 1, 2024 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-38212599

RESUMEN

The effects of gender-affirming hormone therapy on the skeletal integrity and fracture risk in transitioning adolescent trans girls are unknown. To address this knowledge gap, we developed a mouse model to simulate male-to-female transition in human adolescents in whom puberty is first arrested by using gonadotrophin-releasing hormone analogs with subsequent estradiol treatment. Puberty was suppressed by orchidectomy in male mice at 5 weeks of age. At 3 weeks post-surgery, male-to-female mice were treated with a high dose of estradiol (~0.85 mg) by intraperitoneal silastic implantation for 12 weeks. Controls included intact and orchidectomized males at 3 weeks post-surgery, vehicle-treated intact males, intact females and orchidectomized males at 12 weeks post-treatment. Compared to male controls, orchidectomized males exhibited decreased peak bone mass accrual and a decreased maximal force the bone could withstand prior to fracture. Estradiol treatment in orchidectomized male-to-female mice compared to mice in all control groups was associated with an increased cortical thickness in the mid-diaphysis, while the periosteal circumference increased to a level that was intermediate between intact male and female controls, resulting in increased maximal force and stiffness. In trabecular bone, estradiol treatment increased newly formed trabeculae arising from the growth plate as well as mineralizing surface/bone surface and bone formation rate, consistent with the anabolic action of estradiol on osteoblast proliferation. These data support the concept that skeletal integrity can be preserved and that long-term fractures may be prevented in trans girls treated with GnRHa and a sufficiently high dose of GAHT. Further study is needed to identify an optimal dose of estradiol that protects the bone without adverse side effects.


Asunto(s)
Hueso Esponjoso , Estradiol , Adolescente , Masculino , Humanos , Femenino , Ratones , Animales , Estradiol/farmacología , Huesos , Identidad de Género , Modelos Animales de Enfermedad
3.
Bone ; 186: 117143, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38866125

RESUMEN

The effects of gender affirming hormone therapy (GAHT) on bone microarchitecture and fracture risk in adult transgender women is unclear. To investigate the concept that skeletal integrity and strength in trans women may be improved by treatment with a higher dose of GAHT than commonly prescribed, we treated adult male mice with a sustained, high dose of estradiol. Adult male mice at 16 weeks of age were administered ~1.3 mg estradiol by silastic implant, implanted intraperitoneally, for 12 weeks. Controls included vehicle treated intact females and males. High-dose estradiol treatment in males stimulated the endocortical deposition of bone at the femoral mid-diaphysis, increasing cortical thickness and bone area. This led to higher stiffness, maximum force, and the work required to fracture the bone compared to male controls, while post-yield displacement was unaffected. Assessment of the material properties of the bone showed an increase in both elastic modulus and ultimate stress in the estradiol treated males. Treatment of male mice with high dose estradiol was also anabolic for trabecular bone, markedly increasing trabecular bone volume, number and thickness in the distal metaphysis which was accompanied by an increase in the histomorphometric markers of bone remodelling, mineralizing surface/bone surface, bone formation rate and osteoclast number. In conclusion, a high dose of estradiol is anabolic for cortical and trabecular bone in a male to female transgender mouse model, increasing both stiffness and strength. These findings suggest that increasing the current dose of GAHT administered to trans women, while considering other potential adverse effects, may be beneficial to preserving their bone microstructure and strength.


Asunto(s)
Estradiol , Animales , Masculino , Estradiol/farmacología , Estradiol/sangre , Femenino , Ratones , Huesos/efectos de los fármacos , Huesos/diagnóstico por imagen , Densidad Ósea/efectos de los fármacos , Anabolizantes/farmacología , Tamaño de los Órganos/efectos de los fármacos , Ratones Endogámicos C57BL , Humanos , Modelos Animales , Microtomografía por Rayos X
4.
Obes Rev ; 23(6): e13429, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35083843

RESUMEN

Obesity is associated with hypothalamic-pituitary-testicular axis dysregulation in males. Here, we summarize recent evidence derived from clinical trials and studies in preclinical animal models regarding the role of androgen receptor (AR) signaling in the pathophysiology of males with obesity. We also discuss therapeutic strategies targeting the AR for the treatment of obesity and their limitations and provide insight into the future research necessary to advance this field.


Asunto(s)
Receptores Androgénicos , Testosterona , Animales , Humanos , Masculino , Obesidad/etiología , Testosterona/uso terapéutico
5.
J Mol Endocrinol ; 69(1): 269-283, 2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35388795

RESUMEN

We previously identified a novel pathway of testosterone action via the androgen receptor (AR) in bone marrow mesenchymal precursor cells (BM-PCs) to negatively regulate fat mass and improve metabolic function in male mice. This was achieved using our PC-AR Gene Replacement mouse model in which the AR is only expressed in BM-PCs and deleted in all other tissues. We hypothesise that the markedly reduced fat mass and increased insulin sensitivity of PC-AR Gene Replacements will confer protection from diet-induced overweight and obesity. To test this, 6-week-old male PC-AR Gene Replacements and controls (WT, global-AR knockouts (KOs)) were fed a chow or high-caloric diet (HCD) for 8 or 18 weeks. Following 8 weeks (short-term) of HCD, WT and Global-ARKOs had markedly increased subcutaneous white adipose tissue (WAT) and retroperitoneal visceral adipose tissue (VAT) mass compared to chow-fed controls. In contrast, PC-AR Gene Replacements were resistant to WAT and VAT accumulation following short-term HCD feeding accompanied by fewer large adipocytes and upregulation of expression of the metabolic genes Acaca and Pnlpa2. Following long-term HCD feeding for 18 weeks, the PC-AR Gene Replacements were no longer resistant to increased WAT and VAT adiposity, however, maintained their improved whole-body insulin sensitivity with an increased rate of glucose disappearance and increased glucose uptake into subcutaneous WAT. In conclusion, the action of testosterone via the AR in BM-PCs to negatively regulate fat mass and improve metabolism confers resistance from short-term diet-induced weight gain and partial protection from long-term diet-induced obesity in male mice.


Asunto(s)
Resistencia a la Insulina , Animales , Médula Ósea/metabolismo , Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/metabolismo , Sobrepeso , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Células Madre/metabolismo , Testosterona , Aumento de Peso
6.
J Mol Neurosci ; 71(11): 2210-2218, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33403594

RESUMEN

Dysregulation of the serotonergic system has been reported to have a significant role in several neurological disorders including depression, autism and substance abuse disorders. Changes in the expression of the serotonin transporter (SERT) through polymorphisms in the regulatory regions of the SERT gene have been associated, but not yet been conclusively linked to, neuropsychiatric disorders. In turn, dendritic spine structure and function are critical for neuronal function and the disruption of dendritic spine formation at glutamatergic synapses is a hallmark of several neuropsychiatric disorders. To understand the effect of SERT depletion on dendritic spine formation, neuronal cultures were established from the cortex of postnatal day 0-1 SERT knockout (KO) rats. Cortical neurons were subsequently allowed to mature to 21 days in vitro, and dendritic spine density was assessed using immunocytochemical co-labelling of drebrin and microtubule associated protein 2. Genetic knockout of the SERT had a gene-dose effect on dendritic spine densities of cortical neurons. The results of this paper implicate SERT function with the formation of dendritic spines at glutamatergic synapses, thereby offering insight into the aetiology of several neuropathologies.


Asunto(s)
Espinas Dendríticas/metabolismo , Proteínas de Unión al ARN/genética , Animales , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Espinas Dendríticas/fisiología , Eliminación de Gen , Proyección Neuronal , Proteínas de Unión al ARN/metabolismo , Ratas
7.
Sci Rep ; 11(1): 13766, 2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-34215813

RESUMEN

Sexual dimorphism in gene regulation, including DNA methylation, is the main driver of sexual dimorphism in phenotypes. However, the questions of how and when sex shapes DNA methylation remain unresolved. Recently, using mice with different combinations of genetic and phenotypic sex, we identified sex-associated differentially methylated regions (sDMRs) that depended on the sex phenotype. Focusing on a panel of validated sex-phenotype dependent male- and female-biased sDMRs, we tested the developmental dynamics of sex bias in liver methylation and the impacts of mutations in the androgen receptor, estrogen receptor alpha, or the transcriptional repressor Bcl6 gene. True hermaphrodites that carry both unilateral ovaries and contralateral testes were also tested. Our data show that sex bias in methylation either coincides with or follows sex bias in the expression of sDMR-proximal genes, suggesting that sex bias in gene expression may be required for demethylation at certain sDMRs. Global ablation of AR, ESR1, or a liver-specific loss of BCL6, all alter sDMR methylation, whereas presence of both an ovary and a testis delays the establishment of male-type methylation levels in hermaphrodites. Moreover, the Bcl6-LKO shows dissociation between expression and methylation, suggesting a distinct role of BCL6 in demethylation of intragenic sDMRs.


Asunto(s)
Metilación de ADN/genética , Receptor alfa de Estrógeno/genética , Proteínas Proto-Oncogénicas c-bcl-6/genética , Receptores Androgénicos/genética , Animales , Trastornos del Desarrollo Sexual/genética , Epigénesis Genética , Femenino , Regulación de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/genética , Hígado/crecimiento & desarrollo , Hígado/metabolismo , Masculino , Ratones , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Caracteres Sexuales , Sexismo , Testículo/crecimiento & desarrollo , Testículo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA