Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Endocrinol ; 261(3)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38579776

ABSTRACT

Androgens can modulate immune cell function and may contribute to differences in the prevalence and severity of common inflammatory conditions. Although most immune cells are androgen targets, our understanding of how changes in androgen bioavailability can affect immune responses is incomplete. Androgens alter immune cell composition, phenotype, and activation by modulating the expression and secretion of inflammatory mediators or by altering the development and maturation of immune cell precursors. Androgens are generally associated with having suppressive effects on the immune system, but their impacts are cell and tissue context-dependent and can be highly nuanced even within immune cell subsets. In response to androgens, innate immune cells such as neutrophils, monocytes, and macrophages increase the production of the anti-inflammatory cytokine IL-10 and decrease nitric oxide production. Androgens promote the differentiation of T cell subsets and reduce the production of inflammatory mediators, such as IFNG, IL-4 and IL-5. Additionally, androgens/androgen receptor can promote the maturation of B cells. Thus, androgens can be considered as immunomodulatory agents, but further work is required to understand the precise molecular pathways that are regulated at the intersection between endocrine and inflammatory signals. This narrative review focusses on summarising our current understanding of how androgens can alter immune cell function and how this might affect inflammatory responses in health and disease.


Subject(s)
Androgens , Humans , Androgens/metabolism , Androgens/physiology , Animals , Inflammation/immunology , Inflammation/metabolism , Immune System/metabolism , Immune System/physiology , Receptors, Androgen/metabolism
2.
Pain ; 162(9): 2349-2365, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34448751

ABSTRACT

ABSTRACT: Endometriosis (ENDO) and interstitial cystitis/bladder pain syndrome (IC/BPS) are chronic pain conditions for which better treatments are urgently needed. Development of new therapies with proven clinical benefit has been slow. We have conducted a review of existing preclinical in vivo models for ENDO and IC/BPS in rodents, discussed to what extent they replicate the phenotype and pain experience of patients, as well as their relevance for translational research. In 1009 publications detailing ENDO models, 41% used autologous, 26% syngeneic, 18% xenograft, and 11% allogeneic tissue in transplantation models. Intraperitoneal injection of endometrial tissue was the subcategory with the highest construct validity score for translational research. From 1055 IC/BPS publications, most interventions were bladder centric (85%), followed by complex mechanisms (8%) and stress-induced models (7%). Within these categories, the most frequently used models were instillation of irritants (92%), autoimmune (43%), and water avoidance stress (39%), respectively. Notably, although pelvic pain is a hallmark of both conditions and a key endpoint for development of novel therapies, only a small proportion of the studies (models of ENDO: 0.5%-12% and models of IC/BPS: 20%-44%) examined endpoints associated with pain. Moreover, only 2% and 3% of publications using models of ENDO and IC/BPS investigated nonevoked pain endpoints. This analysis highlights the wide variety of models used, limiting reproducibility and translation of results. We recommend refining models so that they better reflect clinical reality, sharing protocols, and using standardized endpoints to improve reproducibility. We are addressing this in our project Innovative Medicines Initiative-PainCare/Translational Research in Pelvic Pain.


Subject(s)
Cystitis, Interstitial , Endometriosis , Cystitis, Interstitial/therapy , Female , Humans , Pelvic Pain/therapy , Reproducibility of Results , Translational Research, Biomedical
3.
J Endocrinol ; 246(3): R75-R93, 2020 09.
Article in English | MEDLINE | ID: mdl-32544881

ABSTRACT

The endometrium is a complex multicellular tissue that is exquisitely sensitive to the actions of sex steroids synthesised in the ovary (endocrine system). Recent studies have highlighted a previously under-appreciated role for local (intracrine) metabolism in fine-tuning tissue function in both health and disease. In this review we have focused on the impact of oestrogens and androgens on endometrial function summarising data from studies on normal endometrial physiology and disorders including infertility, endometriosis and cancer. We consider the evidence that expression of enzymes including aromatase, sulphatase and AKR1C3 by endometrial cells plays an important role in tissue function and malfunction and discuss results from studies using drugs targeting intracrine pathways to treat endometrial disorders. We summarise studies exploring the spatial and temporal expression of oestrogen receptors (ERalpha/ESR1, ERbeta/ESR2 and GPER) and their role in mediating the impact of endogenous and synthetic ligands on cross-talk between vascular, immune, epithelial and stromal cells. There is a single androgen receptor gene and androgens play a key role in stromal-epithelial cross-talk, scar-free healing of endometrium during menstruation and regulation of cell proliferation. The development of new receptor-selective drugs (SERMs, SARMs, SARDs) has reinvigorated interest in targeting receptor subtypes in treatment of disorders including endometriosis and endometrial cancer and some show promise as novel therapies. In summary, understanding the mechanisms regulated by sex steroids provides the platform for improved personalised treatment of endometrial disorders as well as novel insights into the impact of steroids on processes such as tissue repair and regeneration.


Subject(s)
Androgens/metabolism , Endometrium/metabolism , Estrogens/metabolism , Endometriosis/metabolism , Estrogen Receptor alpha/metabolism , Female , Humans
4.
Mol Cell Endocrinol ; 514: 110891, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32533994

ABSTRACT

Renal calcium and phosphate handling is an important contributor to mineral homeostasis and bone health and the androgen receptor (AR) is highly expressed in the kidney. We investigated the short term effects of androgen deprivation on renal calcium and phosphate reabsorption, independent of their effects on bone. Two weeks following orchidectomy (ORX) of adult mice, bone loss occurred along with hypercalciuria, which was similarly prevented by testosterone and dihydrotestosterone supplementation. Treatment with bisphosphonates prior to ORX also inhibited hypercalciuria, indicating that the calcium flux originated from the bone. Renal calcium and phosphate transporter expression was increased post-ORX, independent of bisphosphonates. Furthermore, androgen deprivation appeared to stimulate local synthesis of 1,25(OH)2D3. When bisphosphonate-treated mice were fed a low calcium diet, bone resorption was no longer blocked and secondary hyperparathyroidism developed, which was more pronounced in ORX mice than sham-operated mice. In conclusion, this study shows that androgen deprivation increased renal calcium and phosphate transporter expression, independent of bone, and underlines the importance of adequate intestinal calcium supply in circumstances of androgen deprivation and bisphosphonate treatment.


Subject(s)
Androgens/pharmacology , Calcium, Dietary/pharmacology , Calcium/metabolism , Diphosphonates/pharmacology , Kidney/drug effects , Phosphates/metabolism , Animals , Bone Density/drug effects , Bone Resorption/metabolism , Bone Resorption/pathology , Bone and Bones/drug effects , Bone and Bones/metabolism , Diet , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Orchiectomy , Urinalysis
5.
Proc Natl Acad Sci U S A ; 116(51): 25389-25391, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31792175

ABSTRACT

Endometriosis is a chronic pain condition affecting ∼176 million women worldwide. It is defined by the presence of endometrium-like tissue (lesions) outside the uterus, most commonly on the pelvic peritoneum. There is no cure for endometriosis. All endometriosis drug approvals to date have been contraceptive, limiting their use in women of child-bearing age. We have shown that human peritoneal mesothelial cells (HPMCs) recovered from the pelvic peritoneum of women with endometriosis exhibit significantly higher glycolysis, lower mitochondrial respiration, decreased enzymatic activity of pyruvate dehydrogenase (PDH), and increased production of lactate compared to HPMCs from women without disease. Transforming growth factor-ß1 (TGF-ß1) is elevated in the peritoneal fluid from women with endometriosis, and exposure of HPMCs to TGF-ß1 exacerbates this abnormal phenotype. Treatment of endometriosis HPMCs with the pyruvate dehydrogenase kinase (PDK) inhibitor/PDH activator dichloroacetate (DCA) normalizes HPMC metabolism, reduces lactate secretion, and abrogates endometrial stromal cell proliferation in a coculture model. Oral DCA reduced peritoneal fluid lactate concentrations and endometriosis lesion size in a mouse model. These findings provide the rationale for targeting metabolic processes as a noncontraceptive treatment for women with endometriosis either as a primary nonhormonal treatment or to prevent recurrence after surgery.


Subject(s)
Dichloroacetic Acid/pharmacology , Drug Repositioning , Endometriosis , Animals , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Extracellular Space/drug effects , Female , Glycolysis/drug effects , Humans , Mice , Peritoneum/cytology
6.
J Endocrinol ; 242(3): 227-239, 2019 09.
Article in English | MEDLINE | ID: mdl-31319382

ABSTRACT

Selective androgen receptor modulators (SARMs) have been proposed as therapeutics for women suffering from breast cancer, muscle wasting or urinary incontinence. The androgen receptor (AR) is expressed in the uterus but the impact of SARMs on the function of this organ is unknown. We used a mouse model to compare the impact of SARMs (GTx-007/Andarine®, GTx-024/Enobosarm®), Danazol (a synthetic androstane steroid) and dihydrotestosterone (DHT) on tissue architecture, cell proliferation and gene expression. Ovariectomised mice were treated daily for 7 days with compound or vehicle control (VC). Uterine morphometric characteristics were quantified using high-throughput image analysis (StrataQuest; TissueGnostics), protein and gene expression were evaluated by immunohistochemistry and RT-qPCR, respectively. Treatment with GTx-024, Danazol or DHT induced significant increases in body weight, uterine weight and the surface area of the endometrial stromal and epithelial compartments compared to VC. Treatment with GTx-007 had no impact on these parameters. GTx-024, Danazol and DHT all significantly increased the percentage of Ki67-positive cells in the stroma, but only GTx-024 had an impact on epithelial cell proliferation. GTx-007 significantly increased uterine expression of Wnt4 and Wnt7a, whereas GTx-024 and Danazol decreased their expression. In summary, the impact of GTx-024 and Danazol on uterine cells mirrored that of DHT, whereas GTx-007 had minimal impact on the tested parameters. This study has identified endpoints that have revealed differences in the effects of SARMs on uterine tissue and provides a template for preclinical studies comparing the impact of compounds targeting the AR on endometrial function.


Subject(s)
Acetamides/pharmacology , Aminophenols/pharmacology , Anilides/pharmacology , Danazol/pharmacology , Dihydrotestosterone/pharmacology , Uterus/drug effects , Cell Proliferation/drug effects , Cell Proliferation/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Gene Expression/drug effects , Humans , Receptors, Androgen/metabolism , Uterus/cytology , Uterus/metabolism
7.
Int J Mol Sci ; 19(10)2018 Oct 22.
Article in English | MEDLINE | ID: mdl-30360364

ABSTRACT

Peripheral tissue metabolism of steroids (intracrinology) is now accepted as a key way in which tissues, such as the endometrium, can utilise inactive steroids present in the blood to respond to local physiological demands and 'fine-tune' the activation or inhibition of steroid hormone receptor-dependent processes. Expression of enzymes that play a critical role in the activation and inactivation of bioactive oestrogens (E1, E2) and androgens (A4, T, DHT), as well as expression of steroid hormone receptors, has been detected in endometrial tissues and cells recovered during the menstrual cycle. There is robust evidence that increased expression of aromatase is important for creating a local microenvironment that can support a pregnancy. Measurement of intra-tissue concentrations of steroids using liquid chromatography⁻tandem mass spectrometry has been important in advancing our understanding of a role for androgens in the endometrium, acting both as active ligands for the androgen receptor and as substrates for oestrogen biosynthesis. The emergence of intracrinology, associated with disordered expression of key enzymes such as aromatase, in the aetiology of common women's health disorders such as endometriosis and endometrial cancer has prompted renewed interest in the development of drugs targeting these pathways, opening up new opportunities for targeted therapies and precision medicine.


Subject(s)
Androgens/blood , Endometrium/pathology , Estrogens/blood , Uterine Diseases/blood , Uterine Diseases/pathology , Dehydroepiandrosterone/blood , Endometrium/metabolism , Female , Humans , Testosterone/blood
8.
Best Pract Res Clin Endocrinol Metab ; 32(3): 257-269, 2018 06.
Article in English | MEDLINE | ID: mdl-29779580

ABSTRACT

Endometriosis is a chronic incurable disorder that affects 1 in 10 women of reproductive age: associated symptoms include chronic pain and infertility. The aetiology of endometriosis remains poorly understood but patients, clinicians and researchers are all in agreement that new non-surgical therapies are urgently needed to reduce the severity of symptoms. Preclinical testing of drugs requires the development and validation of models that recapitulate the key features of the disorder. In this review we describe the best-validated animal models (primate, rodent, xenograft) and their contributions to our understanding of the factors underpinning the development of symptoms. We consider the evidence that these models have provided the platform for identification of new therapeutic interventions and reflect on future directions for research and drug validation.


Subject(s)
Chronic Pain , Disease Models, Animal , Endometriosis/etiology , Endometriosis/pathology , Infertility, Female , Animals , Chronic Pain/etiology , Chronic Pain/pathology , Female , Heterografts , Humans , Infertility, Female/etiology , Pelvic Pain/etiology , Pelvic Pain/pathology , Primates , Prodromal Symptoms , Rodentia
9.
J Mol Endocrinol ; 61(2): M57-M65, 2018 08.
Article in English | MEDLINE | ID: mdl-29720512

ABSTRACT

In women, establishment of pregnancy is dependent upon 'fine-tuning' of the endometrial microenvironment, which is mediated by terminal differentiation (decidualisation) of endometrial stromal fibroblasts (ESFs). We have demonstrated that intracrine steroid metabolism plays a key role in regulating decidualisation and is essential for time-dependent expression of key factors required for endometrial receptivity. The primary aim of the current study was to determine whether sulphated steroids can act as precursors to bioactive sex steroids during decidualisation. We used primary human ESF and a robust in vitro model of decidualisation to assess the expression of genes associated with sulphation, desulphation and transport of sulphated steroids in human ESF as well as the impact of the steroid sulphatase (STS) inhibitor STX64 (Irosustat). We found evidence for an increase in both expression and activity of STS in response to a decidualisation stimulus with abrogation of oestrone biosynthesis and decreased secretion of the decidualisation marker IGFBP1 in the presence of STX64. These results provide novel insight into the contribution of STS to the intracrine regulation of decidualisation.


Subject(s)
Endometrium/metabolism , Signal Transduction/physiology , Steryl-Sulfatase/metabolism , Sulfates/metabolism , Animals , Embryo Implantation/physiology , Female , Humans , Pregnancy
10.
Fertil Steril ; 109(4): 728-734.e2, 2018 04.
Article in English | MEDLINE | ID: mdl-29397924

ABSTRACT

OBJECTIVE: To investigate the impact of the androgen precursor dehydroepiandrosterone (DHEA) on the decidualization of human endometrial stromal cells isolated from women of advanced reproductive age. DESIGN: In vitro study. SETTING: University research institute. PATIENT(S): Proliferative phase primary human endometrial stromal fibroblasts (hESFs) were isolated from women of advanced reproductive age (n = 16; mean age, 44.7 ± 2.3). None of the women were receiving hormone therapy or had endometriosis. INTERVENTION(S): Isolated hESFs were decidualized in vitro by incubation with P (1 µM) and cAMP (0.1 mg/mL) in the presence, or absence, of DHEA (10 nM, 100 nM). MAIN OUTCOME MEASURE(S): Secretion of androgens was assessed by ELISA. Expression of decidualization markers and endometrial receptivity markers was assessed by quantitative polymerase chain reaction and ELISA. RESULT(S): Decidualization responses were retained in hESF isolated from women of advanced reproductive age. Supplementation with DHEA increased androgen biosynthesis and concentrations of T and dihydrotestosterone were ∼3× greater after coincubation with DHEA compared with hESF stimulated with decidualization alone. Addition of DHEA to decidualized hESF increased expression of the decidualization markers IGFBP1 and PRL and the endometrial receptivity marker SPP1. DHEA enhanced secretion of IGFBP1, PRL, and SPP1 proteins maximally by day 8 of the decidualization time course concomitant with peak androgen concentrations. CONCLUSION(S): These novel results demonstrate DHEA can enhance in vitro decidualization responses of hESF from women of advanced reproductive age. Supplementation with DHEA during the receptive phase may augment endometrial function and improve pregnancy rates in natural or assisted reproductive cycles.


Subject(s)
Cell Proliferation/drug effects , Decidua/drug effects , Dehydroepiandrosterone/pharmacology , Fibroblasts/drug effects , Maternal Age , Reproductive Health , Stromal Cells/drug effects , Adult , Biomarkers/metabolism , Cells, Cultured , Decidua/cytology , Decidua/metabolism , Dihydrotestosterone/metabolism , Female , Fibroblasts/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 1/metabolism , Middle Aged , Osteopontin/metabolism , Prolactin/metabolism , Stromal Cells/metabolism , Time Factors
11.
Mol Cell Endocrinol ; 465: 48-60, 2018 04 15.
Article in English | MEDLINE | ID: mdl-28919297

ABSTRACT

Androgens are synthesised in both the ovary and adrenals in women and play an important role in the regulation of female fertility, as well as in the aetiology of disorders such as polycystic ovarian syndrome, endometriosis and endometrial cancer. The endometrium is an androgen target tissue and the impact of AR-mediated effects has been studied using human endometrial tissue samples and rodent models. In this review we highlight recent evidence that endometrial androgen biosynthesis and intracrine action is important in preparation of a tissue microenvironment that can support implantation and establishment of pregnancy. The impact of androgens on endometrial cell proliferation, in repair of the endometrial wound at the time of menstruation and in endometrial disorders is discussed. Future directions for research focused on AR function as a therapeutic target are considered.


Subject(s)
Androgens/metabolism , Endometrium/metabolism , Androgens/biosynthesis , Animals , Cell Proliferation , Endometrium/pathology , Female , Humans , Menstrual Cycle , Receptors, Androgen/metabolism , Wound Healing
12.
Sci Rep ; 7(1): 2001, 2017 05 17.
Article in English | MEDLINE | ID: mdl-28515460

ABSTRACT

Miscarriage affects ~20% of pregnancies and maternal infections account for ~15% of early miscarriages. Chlamydia trachomatis (Ct) has been associated with miscarriage but the underlying mechanisms are unknown. Successful implantation requires endometrial stromal cell (ESC) decidualisation. Maintenance of pregnancy requires angiogenesis, establishment of the correct cellular milieu and trophoblast invasion, all of which involve the action of chemokines. Our objective was to determine whether Ct infection impacts upon ESC decidualisation and chemokine secretion. Human primary ESC were decidualised in-vitro, infected with Ct serovar E, and changes in expression of genes of interest were measured using RT-PCR, proteomic array and ELISA. We demonstrate for the first time that Ct can infect and proliferate in ESC. Expression of the decidualisation marker prolactin was decreased in Ct-infected ESC at both mRNA and protein levels. Ct infection altered the chemokine profile of decidualised ESC as shown by proteomic array. Chemokines CXCL12 and CXCL16, important for trophoblast invasion, were analysed further and expression was reduced in infected decidualised cells at mRNA and protein levels. Our data indicate that Ct infection of ESC impairs decidualisation and alters chemokine release. These findings at least partially explain how Ct infection could result in adverse pregnancy outcomes.


Subject(s)
Chemokines/biosynthesis , Chlamydia Infections/metabolism , Chlamydia Infections/microbiology , Chlamydia trachomatis/physiology , Decidua/metabolism , Decidua/microbiology , Stromal Cells/metabolism , Stromal Cells/microbiology , Cells, Cultured , Chlamydia Infections/pathology , Decidua/pathology , Female , Humans , Immunity, Innate , Proteome , Proteomics/methods
13.
J Mol Endocrinol ; 57(1): R35-47, 2016 07.
Article in English | MEDLINE | ID: mdl-27067639

ABSTRACT

During the establishment of pregnancy, the ovarian-derived hormones progesterone and oestradiol regulate remodelling of the endometrium to promote an environment that is able to support and maintain a successful pregnancy. Decidualisation is characterised by differentiation of endometrial stromal cells that secrete growth factors and cytokines that regulate vascular remodelling and immune cell influx. This differentiation process is critical for reproduction, and inadequate decidualisation is implicated in the aetiology of pregnancy disorders such as foetal growth restriction and preeclampsia. In contrast to progesterone and oestradiol, the role of androgens in regulating endometrial function is poorly understood. Androgen receptors are expressed in the endometrium, and androgens are reported to regulate both the transcriptome and the secretome of endometrial stromal cells. In androgen-target tissues, circulating precursors are activated to mediate local effects, and recent studies report that steroid concentrations detected in endometrial tissue are distinct to those detected in the peripheral circulation. New evidence suggests that decidualisation results in dynamic changes in the expression of androgen biosynthetic enzymes, highlighting a role for pre-receptor regulation of androgen action during the establishment of pregnancy. These results suggest that such enzymes could be future therapeutic targets for the treatment of infertility associated with endometrial dysfunction. In conclusion, these data support the hypothesis that androgens play a beneficial role in regulating the establishment and maintenance of pregnancy. Future studies should be focussed on investigating the safety and efficacy of androgen supplementation with the potential for utilisation of novel therapeutics, such as selective androgen receptor modulators, to improve reproductive outcomes in women.


Subject(s)
Androgens/metabolism , Reproduction/physiology , Animals , Decidua/metabolism , Endometrium/metabolism , Estradiol/metabolism , Female , Gene Expression Regulation , Gene Expression Regulation, Enzymologic , Humans , Pregnancy , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Signal Transduction , Steroids/metabolism
14.
Endocrinology ; 157(5): 2116-28, 2016 05.
Article in English | MEDLINE | ID: mdl-26963473

ABSTRACT

The endometrium consists of stromal and epithelial compartments (luminal and glandular) with distinct functions in the regulation of uterine homeostasis. Ovarian sex steroids, namely 17ß-estradiol and progesterone, play essential roles in modulating uterine cell proliferation, stromal-epithelial cross-talk and differentiation in preparation for pregnancy. The effect of androgens on uterine function remains poorly understood. The current study investigated the effect of the non-aromatizable androgen dihydrotestosterone (DHT) on mouse endometrial function. Ovx female mice were given a single sc injection (short treatment) or 7 daily injections (long treatment) of vehicle alone (5% ethanol, 0.4% methylcellulose) or vehicle with the addition of 0.2 mg DHT (n=8/group) and a single injection of bromodeoxyuridine 2 hours prior to tissue recovery. Treatment with DHT increased uterine weight, the area of the endometrial compartment and immunoexpression of the androgen receptor in the luminal and glandular epithelium. Treatment-dependent proliferation of epithelial cells was identified by immunostaining for MKi67 and bromodeoxyuridine. Real-time PCR identified significant DHT-dependent changes in the concentrations of mRNAs encoded by genes implicated in the regulation of the cell cycle (Wee1, Ccnd1, Rb1) and stromal-epithelial interactions (Wnt4, Wnt5a, Wnt7a, Cdh1, Vcl, Igf1, Prl8, Prlr) as well as a striking effect on the number of endometrial glands. This study has revealed a novel role for androgens in regulating uterine function with an effect on the glandular compartment of the endometrium. This previously unrecognized role for androgens has implications for our understanding of the role of androgens in regulation of endometrial function and fertility in women.


Subject(s)
Androgens/pharmacology , Cell Proliferation/drug effects , Dihydrotestosterone/pharmacology , Endometrium/drug effects , Epithelial Cells/drug effects , Animals , Cell Cycle/genetics , Endometrium/cytology , Endometrium/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Gene Expression/drug effects , Mice , Receptors, Androgen/metabolism
15.
Sci Rep ; 6: 19970, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26817618

ABSTRACT

The endometrium is a complex, steroid-dependent tissue that undergoes dynamic cyclical remodelling. Transformation of stromal fibroblasts (ESC) into specialised secretory cells (decidualization) is fundamental to the establishment of a receptive endometrial microenvironment which can support and maintain pregnancy. Androgen receptors (AR) are present in ESC; in other tissues local metabolism of ovarian and adrenal-derived androgens regulate AR-dependent gene expression. We hypothesised that altered expression/activity of androgen biosynthetic enzymes would regulate tissue availability of bioactive androgens and the process of decidualization. Primary human ESC were treated in vitro for 1-8 days with progesterone and cAMP (decidualized) in the presence or absence of the AR antagonist flutamide. Time and treatment-dependent changes in genes essential for a) intra-tissue biosynthesis of androgens (5α-reductase/SRD5A1, aldo-keto reductase family 1 member C3/AKR1C3), b) establishment of endometrial decidualization (IGFBP1, prolactin) and c) endometrial receptivity (SPP1, MAOA, EDNRB) were measured. Decidualization of ESC resulted in significant time-dependent changes in expression of AKR1C3 and SRD5A1 and secretion of T/DHT. Addition of flutamide significantly reduced secretion of IGFBP1 and prolactin and altered the expression of endometrial receptivity markers. Intracrine biosynthesis of endometrial androgens during decidualization may play a key role in endometrial receptivity and offer a novel target for fertility treatment.


Subject(s)
Androgens/metabolism , Decidua/metabolism , Endometrium/metabolism , Gene Expression Regulation , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , Female , Flutamide/pharmacology , Gene Expression Regulation/drug effects , Humans , Membrane Proteins/genetics , Pregnancy , Testosterone/biosynthesis , Testosterone/metabolism
16.
Endocrinology ; 156(12): 4522-33, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26393303

ABSTRACT

Androgens increase skeletal muscle mass, but their clinical use is hampered by a lack of tissue selectivity and subsequent side effects. Selective androgen receptor modulators elicit muscle-anabolic effects while only sparingly affecting reproductive tissues. The selective androgen receptor modulator, GTx-024 (enobosarm), is being investigated for cancer cachexia, sarcopenia, and muscle wasting diseases. Here we investigate the role of muscle androgen receptor (AR) in the anabolic effect of GTx-024. In mice lacking AR in the satellite cell lineage (satARKO), the weight of the androgen-sensitive levator ani muscle was lower but was decreased further upon orchidectomy. GTx-024 was as effective as DHT in restoring levator ani weights to sham levels. Expression of the muscle-specific, androgen-responsive genes S-adenosylmethionine decarboxylase and myostatin was decreased by orchidectomy and restored by GTx-024 and DHT in control mice, whereas the expression was low and unaffected by androgen status in satARKO. In contrast, insulin-like growth factor 1Ea expression was not different between satARKO and control muscle, decreased upon castration, and was restored by DHT and GTx-024 in both genotypes. These data indicate that GTx-024 does not selectively modulate AR in the satellite cell lineage and that cells outside this lineage remain androgen responsive in satARKO muscle. Indeed, residual AR-positive cells were present in satARKO muscle, coexpressing the fibroblast-lineage marker vimentin. AR positive, muscle-resident fibroblasts could therefore be involved in the indirect effects of androgens on muscle. In conclusion, both DHT and GTx-024 target AR pathways in the satellite cell lineage, but cells outside this lineage also contribute to the anabolic effects of androgens.


Subject(s)
Amides/pharmacology , Anabolic Agents/pharmacology , Body Composition/drug effects , Muscle, Skeletal/drug effects , Receptors, Androgen/metabolism , Satellite Cells, Skeletal Muscle/drug effects , Androgens/pharmacology , Anilides , Animals , Cachexia/drug therapy , Cell Lineage , Dihydrotestosterone/pharmacology , Male , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Orchiectomy , Organ Size/drug effects , Receptors, Androgen/genetics , Sarcopenia/drug therapy , Satellite Cells, Skeletal Muscle/metabolism , Wasting Syndrome/drug therapy
17.
FASEB J ; 29(3): 894-910, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25404712

ABSTRACT

Leydig cell number and function decline as men age, and low testosterone is associated with all "Western" cardio-metabolic disorders. However, whether perturbed androgen action within the adult Leydig cell lineage predisposes individuals to this late-onset degeneration remains unknown. To address this, we generated a novel mouse model in which androgen receptor (AR) is ablated from ∼75% of adult Leydig stem cell/cell progenitors, from fetal life onward (Leydig cell AR knockout mice), permitting interrogation of the specific roles of autocrine Leydig cell AR signaling through comparison to adjacent AR-retaining Leydig cells, testes from littermate controls, and to human testes, including from patients with complete androgen insensitivity syndrome (CAIS). This revealed that autocrine AR signaling is dispensable for the attainment of final Leydig cell number but is essential for Leydig cell maturation and regulation of steroidogenic enzymes in adulthood. Furthermore, these studies reveal that autocrine AR signaling in Leydig cells protects against late-onset degeneration of the seminiferous epithelium in mice and inhibits Leydig cell apoptosis in both adult mice and patients with CAIS, possibly via opposing aberrant estrogen signaling. We conclude that autocrine androgen action within Leydig cells is essential for the lifelong support of spermatogenesis and the development and lifelong health of Leydig cells.


Subject(s)
Androgen-Insensitivity Syndrome/pathology , Androgens/pharmacology , Apoptosis/drug effects , Leydig Cells/pathology , Receptors, Androgen/physiology , Testis/pathology , Adolescent , Adult , Androgen-Insensitivity Syndrome/drug therapy , Androgen-Insensitivity Syndrome/metabolism , Animals , Autocrine Communication , Blotting, Western , Cells, Cultured , Child , Humans , Immunoenzyme Techniques , Leydig Cells/drug effects , Leydig Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protective Agents/pharmacology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Spermatogenesis/drug effects , Testis/drug effects , Testis/metabolism , Young Adult
18.
Endocr Relat Cancer ; 21(4): T203-18, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24623742

ABSTRACT

Endometrial cancer (EC) and ovarian cancer are common gynaecological malignancies. The impact of androgen action in these cancers is poorly understood; however, there is emerging evidence to suggest that targeting androgen signalling may be of therapeutic benefit. Epidemiological evidence suggests that there is an increased risk of EC associated with exposure to elevated levels of androgens, and genetic variants in genes related to both androgen biosynthesis and action are associated with an increased risk of both EC and ovarian cancer. Androgen receptors (ARs) may be a potential therapeutic target in EC due to reported anti-proliferative activities of androgens. By contrast, androgens may promote growth of some ovarian cancers and anti-androgen therapy has been proposed. Introduction of new therapies targeting ARs expressed in EC or ovarian cancer will require a much greater understanding of the impacts of cell context-specific AR-dependent signalling and how ARs can crosstalk with other steroid receptors during progression of disease. This review considers the evidence that androgens may be important in the aetiology of EC and ovarian cancer with discussion of evidence for androgen action in normal and malignant endometrial and ovarian tissue.


Subject(s)
Androgens/pharmacology , Endometrial Neoplasms/etiology , Ovarian Neoplasms/etiology , Androgen Antagonists/therapeutic use , Androgens/metabolism , Endometrial Neoplasms/epidemiology , Endometrial Neoplasms/therapy , Endometrium/drug effects , Endometrium/metabolism , Female , Humans , Molecular Targeted Therapy , Ovarian Neoplasms/epidemiology , Ovarian Neoplasms/therapy , Ovary/drug effects , Ovary/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...