Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
2.
PLoS One ; 18(9): e0290846, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37656709

RESUMEN

Sertoli cells support the development of sperm and the function of various somatic cells in the interstitium between the tubules. Sertoli cells regulate the function of the testicular vasculature and the development and function of the Leydig cells that produce testosterone for fertility and virility. However, the Sertoli cell-derived factors that regulate these cells are largely unknown. To define potential mechanisms by which Sertoli cells could support testicular somatic cell function, we aimed to identify Sertoli cell-enriched proteins in the testicular interstitial fluid (TIF) between the tubules. We previously resolved the proteome of TIF in mice and humans and have shown it to be a rich source of seminiferous tubule-derived proteins. In the current study, we designed bioinformatic strategies to interrogate relevant proteomic and genomic datasets to identify Sertoli cell-enriched proteins in mouse and human TIF. We analysed proteins in mouse TIF that were significantly reduced after one week of acute Sertoli cell ablation in vivo and validated which of these are likely to arise primarily from Sertoli cells based on relevant mouse testis RNASeq datasets. We used a different, but complementary, approach to identify Sertoli cell-enriched proteins in human TIF, taking advantage of high-quality human testis genomic, proteomic and immunohistochemical datasets. We identified a total of 47 and 40 Sertoli cell-enriched proteins in mouse and human TIF, respectively, including 15 proteins that are conserved in both species. Proteins with potential roles in angiogenesis, the regulation of Leydig cells or steroidogenesis, and immune cell regulation were identified. The data suggests that some of these proteins are secreted, but that Sertoli cells also deposit specific proteins into TIF via the release of extracellular vesicles. In conclusion, we have identified novel Sertoli cell-enriched proteins in TIF that are candidates for regulating somatic cell-cell communication and testis function.


Asunto(s)
Células de Sertoli , Testículo , Humanos , Masculino , Animales , Ratones , Líquido Extracelular , Proteómica , Semen
3.
Front Public Health ; 11: 1195751, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37457264

RESUMEN

Introduction: Vaccine hesitancy is a global health threat undermining control of many vaccine-preventable diseases. Patient-level education has largely been ineffective in reducing vaccine concerns and increasing vaccine uptake. We built and evaluated a personalized vaccine risk communication website called LetsTalkShots in English, Spanish and French (Canadian) for vaccines across the lifespan. LetsTalkShots tailors lived experiences, credible sources and informational animations to disseminate the right message from the right messenger to the right person, applying a broad range of behavioral theories. Methods: We used mixed-methods research to test our animation and some aspects of credible sources and personal narratives. We conducted 67 discussion groups (n = 325 persons), stratified by race/ethnicity (African American, Hispanic, and White people) and population (e.g., parents, pregnant women, adolescents, younger adults, and older adults). Using a large Ipsos survey among English-speaking respondents (n = 2,272), we tested animations aligned with vaccine concerns and specific to population (e.g., parents of children, parents of adolescents, younger adults, older adults). Results: Discussion groups provided robust feedback specific to each animation as well as areas for improvements across animations. Most respondents indicated that the information presented was interesting (85.5%), clear (96.0%), helpful (87.0%), and trustworthy (82.2%). Discussion: Tailored vaccine risk communication can assist decision makers as they consider vaccination for themselves, their families, and their communities. LetsTalkShots presents a model for personalized communication in other areas of medicine and public health.


Asunto(s)
Comunicación , Vacunación , Vacunas , Adolescente , Anciano , Niño , Femenino , Humanos , Embarazo , Negro o Afroamericano , Canadá , Medicina de Precisión , Vacilación a la Vacunación , Riesgo , Salud Pública , Promoción de la Salud , Educación en Salud/métodos , Hispánicos o Latinos , Blanco , Adulto Joven , Padres
4.
Int J Mol Sci ; 23(23)2022 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-36499341

RESUMEN

Glucocorticoids are steroids involved in key physiological processes such as development, metabolism, inflammatory and stress responses and are mostly used exogenously as medications to treat various inflammation-based conditions. They act via the glucocorticoid receptor (GR) expressed in most cells. Exogenous glucocorticoids can negatively impact the function of the Leydig cells in the testis, leading to decreased androgen production. However, endogenous glucocorticoids are produced by the adrenal and within the testis, but whether their action on GR in Leydig cells regulates steroidogenesis is unknown. This study aimed to define the role of endogenous GR signalling in adult Leydig cells. We developed and compared two models; an inducible Cre transgene driven by expression of the Cyp17a1 steroidogenic gene (Cyp17-iCre) that depletes GR during development and a viral vector-driven Cre (AAV9-Cre) to deplete GR in adulthood. The delivery of AAV9-Cre ablated GR in adult mouse Leydig cells depleted Leydig cell GR more efficiently than the Cyp17-iCre model. Importantly, adult depletion of GR in Leydig cells caused reduced expression of luteinising hormone receptor (Lhcgr) and of steroidogenic enzymes required for normal androgen production. These findings reveal that Leydig cell GR signalling plays a physiological role in the testis and highlight that a normal balance of glucocorticoid activity in the testis is important for steroidogenesis.


Asunto(s)
Células Intersticiales del Testículo , Receptores de Glucocorticoides , Ratones , Masculino , Animales , Células Intersticiales del Testículo/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Glucocorticoides/genética , Glucocorticoides/metabolismo , Andrógenos/metabolismo , Ratones Noqueados , Testículo/metabolismo , Expresión Génica
5.
Front Endocrinol (Lausanne) ; 13: 1027164, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36465608

RESUMEN

Decidualization is the hormone-dependent process of endometrial remodeling that is essential for fertility and reproductive health. It is characterized by dynamic changes in the endometrial stromal compartment including differentiation of fibroblasts, immune cell trafficking and vascular remodeling. Deficits in decidualization are implicated in disorders of pregnancy such as implantation failure, intra-uterine growth restriction, and pre-eclampsia. Androgens are key regulators of decidualization that promote optimal differentiation of stromal fibroblasts and activation of downstream signaling pathways required for endometrial remodeling. We have shown that androgen biosynthesis, via 5α-reductase-dependent production of dihydrotestosterone, is required for optimal decidualization of human stromal fibroblasts in vitro, but whether this is required for decidualization in vivo has not been tested. In the current study we used steroid 5α-reductase type 1 (SRD5A1) deficient mice (Srd5a1-/- mice) and a validated model of induced decidualization to investigate the role of SRD5A1 and intracrine androgen signaling in endometrial decidualization. We measured decidualization response (weight/proportion), transcriptomic changes, and morphological and functional parameters of vascular development. These investigations revealed a striking effect of 5α-reductase deficiency on the decidualization response. Furthermore, vessel permeability and transcriptional regulation of angiogenesis signaling pathways, particularly those that involved vascular endothelial growth factor (VEGF), were disrupted in the absence of 5α-reductase. In Srd5a1-/- mice, injection of dihydrotestosterone co-incident with decidualization restored decidualization responses, vessel permeability, and expression of angiogenesis genes to wild type levels. Androgen availability declines with age which may contribute to age-related risk of pregnancy disorders. These findings show that intracrine androgen signaling is required for optimal decidualization in vivo and confirm a major role for androgens in the development of the vasculature during decidualization through regulation of the VEGF pathway. These findings highlight new opportunities for improving age-related deficits in fertility and pregnancy health by targeting androgen-dependent signaling in the endometrium.


Asunto(s)
3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa , Decidua , Remodelación Vascular , Animales , Femenino , Ratones , Embarazo , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/genética , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/metabolismo , Andrógenos/farmacología , Colestenona 5 alfa-Reductasa/genética , Colestenona 5 alfa-Reductasa/metabolismo , Decidua/efectos de los fármacos , Decidua/metabolismo , Dihidrotestosterona/farmacología , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Remodelación Vascular/efectos de los fármacos , Remodelación Vascular/genética , Remodelación Vascular/fisiología
6.
Int J Mol Sci ; 23(24)2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36555196

RESUMEN

Androgens such as testosterone and dihydrotestosterone (DHT) are essential for male sexual development, masculinisation, and fertility. Testosterone is produced via the canonical androgen production pathway and is essential for normal masculinisation and testis function. Disruption to androgen production can result in disorders of sexual development (DSD). In the canonical pathway, 17ß-hydroxysteroid dehydrogenase type 3 (HSD17B3) is viewed as a critical enzyme in the production of testosterone, performing the final conversion required. HSD17B3 deficiency in humans is associated with DSD due to low testosterone concentration during development. Individuals with HSD17B3 mutations have poorly masculinised external genitalia that can appear as ambiguous or female, whilst having internal Wolffian structures and testes. Recent studies in mice deficient in HSD17B3 have made the surprising finding that testosterone production is maintained, male mice are masculinised and remain fertile, suggesting differences between mice and human testosterone production exist. We discuss the phenotypic differences observed and the possible other pathways and enzymes that could be contributing to testosterone production and male development. The identification of alternative testosterone synthesising enzymes could inform the development of novel therapies to endogenously regulate testosterone production in individuals with testosterone deficiency.


Asunto(s)
Andrógenos , Testosterona , Humanos , Masculino , Femenino , Ratones , Animales , Virilismo/genética , Mutación , Dihidrotestosterona , 17-Hidroxiesteroide Deshidrogenasas/metabolismo
7.
Curr Opin Cell Biol ; 77: 102104, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35671587

RESUMEN

Spermatozoa are comprised of many unique proteins not expressed elsewhere. Sperm-specific proteins are first expressed at puberty, after the development of immune tolerance to self-antigens, and have been assumed to remain confined inside the seminiferous tubules, protected from immune cell recognition by various mechanisms of testicular immune privilege. However, new data has shown that sperm-specific proteins are released by the tubules into the surrounding interstitial fluid; from here they can contact immune cells, potentially promote immune tolerance, and enter the circulation. These new findings have clinical implications for diagnostics and therapeutics targeted at a specific class of proteins known as cancer-testis antigens (CTA), the opportunity to identify new communication pathways in the testis, and to discover new ways to monitor testis function.


Asunto(s)
Neoplasias , Semen , Humanos , Inmunoterapia , Masculino , Neoplasias/diagnóstico , Neoplasias/terapia , Túbulos Seminíferos , Espermatozoides
8.
Mol Cell Endocrinol ; 544: 111556, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35031431

RESUMEN

Testicular Leydig cells (LCs) are the principal source of circulating testosterone in males. LC steroidogenesis maintains sexual function, fertility and general health, and is influenced by various paracrine factors. The leukemia inhibitory factor receptor (LIFR) is expressed in the testis and activated by different ligands, including leukemia inhibitory factor (LIF), produced by peritubular myoid cells. LIF can modulate LC testosterone production in vitro under certain circumstances, but the role of consolidated signalling through LIFR in adult LC function in vivo has not been established. We used a conditional Lifr allele in combination with adenoviral vectors expressing Cre-recombinase to generate an acute model of LC Lifr-KO in the adult mouse testis, and showed that LC Lifr is not required for short term LC survival or basal steroidogenesis. However, LIFR-signalling negatively regulates steroidogenic enzyme expression and maximal gonadotrophin-stimulated testosterone biosynthesis, expanding our understanding of the intricate regulation of LC steroidogenic function.


Asunto(s)
Células Intersticiales del Testículo , Testosterona , Animales , Factor Inhibidor de Leucemia/metabolismo , Células Intersticiales del Testículo/metabolismo , Masculino , Ratones , Receptores OSM-LIF/metabolismo , Testículo/metabolismo , Testosterona/metabolismo
9.
Semin Cell Dev Biol ; 121: 2-9, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34229950

RESUMEN

Sertoli cells are the orchestrators of spermatogenesis; they support fetal germ cell commitment to the male pathway and are essential for germ cell development, from maintenance of the spermatogonial stem cell niche and spermatogonial populations, through meiosis and spermiogeneis and to the final release of mature spermatids during spermiation. However, Sertoli cells are also emerging as key regulators of other testis somatic cells, including supporting peritubular myoid cell development in the pre-pubertal testis and supporting the function of the testicular vasculature and in contributing to testicular immune privilege. Sertoli cells also have a major role in regulating androgen production within the testis, by specifying interstitial cells to a steroidogenic fate, contributing to androgen production in the fetal testis, and supporting fetal and adult Leydig cell development and function. Here, we provide an overview of the specific roles for Sertoli cells in the testis and highlight how these cells are key drivers of testicular sperm output, and of adult testis size and optimal function of other testicular somatic cells, including the steroidogenic Leydig cells.


Asunto(s)
Células Intersticiales del Testículo/metabolismo , Células de Sertoli/metabolismo , Animales , Humanos , Masculino , Ratas , Testículo
10.
Front Cell Dev Biol ; 9: 695546, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34262907

RESUMEN

Leydig cells (LC) are the main testicular androgen-producing cells. In eutherian mammals, two types of LCs emerge successively during testicular development, fetal Leydig cells (FLCs) and adult Leydig cells (ALCs). Both display significant differences in androgen production and regulation. Using bulk RNA sequencing, we compared the transcriptomes of both LC populations to characterize their specific transcriptional and functional features. Despite similar transcriptomic profiles, a quarter of the genes show significant variations in expression between FLCs and ALCs. Non-transcriptional events, such as alternative splicing was also observed, including a high rate of intron retention in FLCs compared to ALCs. The use of single-cell RNA sequencing data also allowed the identification of nine FLC-specific genes and 50 ALC-specific genes. Expression of the corticotropin-releasing hormone 1 (Crhr1) receptor and the ACTH receptor melanocortin type 2 receptor (Mc2r) specifically in FLCs suggests a dual regulation of steroidogenesis. The androstenedione synthesis by FLCs is stimulated by luteinizing hormone (LH), corticotrophin-releasing hormone (CRH), and adrenocorticotropic hormone (ACTH) whereas the testosterone synthesis by ALCs is dependent exclusively on LH. Overall, our study provides a useful database to explore LC development and functions.

11.
Andrology ; 9(5): 1652-1661, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33998165

RESUMEN

BACKGROUND: Circulating prolactin concentration in rodents and humans is sexually dimorphic. Oestrogens are a well-characterised stimulator of prolactin release. Circulating prolactin fluctuates throughout the menstrual/oestrous cycle of females in response to oestrogen levels, but remains continually low in males. We have previously identified androgens as an inhibitor of prolactin release through characterisation of males of a mouse line with a conditional pituitary androgen receptor knockout (PARKO) which have an increase in circulating prolactin, but unchanged lactotroph number. OBJECTIVES: In the present study, we aimed to specify the cell type that androgens act on to repress prolactin release. MATERIALS AND METHODS: PARKO, lactotroph-specific, Pit1 lineage-specific and neural-specific conditional androgen receptor knockout male mice were investigated using prolactin ELISA, pituitary electron microscopy, immunohistochemistry and qRT-PCR. RESULTS: Lactotroph-specific, Pit1 lineage-specific and neural-specific conditional AR knockouts did not duplicate the high circulating prolactin seen in the PARKO line. Using electron microscopy to examine ultrastructure, we showed that pituitary androgen receptor knockout male mice develop lactotrophs that resemble those seen in female mice. Castrated PARKO males have significantly reduced circulating prolactin compared to intact males. When expression of selected oestrogen-regulated anterior pituitary genes was examined, there were no differences in expression level between controls and knockouts. DISCUSSION: The cell type that androgens act on to repress prolactin release is not the lactotroph, cells in the Pit1-lineage, or the dopaminergic neurons in the hypothalamus. PARKO males develop a female-specific lactotroph ultrastructure that this is likely to contribute to the increase in circulating prolactin. Castrated PARKO males have significantly reduced circulating prolactin compared to intact males, which suggests that removal of both circulating oestrogens and androgens reduces the stimulation of pituitary prolactin release. CONCLUSION: Further investigation is needed into prolactin regulation by changes in androgen-oestrogen balance, which is involved sexual dimorphism of development and diseases including hyperprolactinemia.


Asunto(s)
Hiperprolactinemia/genética , Lactotrofos , Receptores Androgénicos/deficiencia , Animales , Estrógenos/metabolismo , Masculino , Ratones , Ratones Noqueados , Hipófisis/metabolismo , Prolactina/metabolismo
12.
FASEB J ; 35(3): e21397, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33565176

RESUMEN

Sperm develop from puberty in the seminiferous tubules, inside the blood-testis barrier to prevent their recognition as "non-self" by the immune system, and it is widely assumed that human sperm-specific proteins cannot access the circulatory or immune systems. Sperm-specific proteins aberrantly expressed in cancer, known as cancer-testis antigens (CTAs), are often pursued as cancer biomarkers and therapeutic targets based on the assumption they are neoantigens absent from the circulation in healthy men. Here, we identify a wide range of germ cell-derived and sperm-specific proteins, including multiple CTAs, that are selectively deposited by the Sertoli cells of the adult mouse and human seminiferous tubules into testicular interstitial fluid (TIF) that is "outside" the blood-testis barrier. From TIF, the proteins can access the circulatory- and immune systems. Disruption of spermatogenesis decreases the abundance of these proteins in mouse TIF, and a sperm-specific CTA is significantly decreased in TIF from infertile men, suggesting that exposure of certain CTAs to the immune system could depend on fertility status. The results provide a rationale for the development of blood-based tests useful in the management of male infertility and indicate CTA candidates for cancer immunotherapy and biomarker development that could show sex-specific and male-fertility-related responses.


Asunto(s)
Antígenos de Neoplasias/análisis , Proteínas/análisis , Túbulos Seminíferos/metabolismo , Espermatozoides/química , Animales , Barrera Hematotesticular , Líquido Extracelular/química , Humanos , Inmunoterapia , Infertilidad Masculina/metabolismo , Masculino , Ratones , Neoplasias/terapia , Proteoma , Células de Sertoli/fisiología , Espermatogénesis , Testículo/metabolismo
13.
Andrology ; 9(1): 460-473, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32996275

RESUMEN

BACKGROUND: Despite the increasing popularity of deliverable transgenics, a robust and fully validated method for targeting Leydig cells, capable of delivering long-term transgene expression, is yet to be defined. OBJECTIVES: We compared three viral vector systems in terms of their cell targeting specificity, longevity of gene expression and impact on targeted cell types when delivered to the interstitial compartment of the mouse testis. MATERIALS & METHODS: We delivered lentiviral, adenoviral and adeno-associated (AAV) viral particles to the interstitial compartment of adult mouse testis. Immunolocalization and stereology were performed to characterize ability of vectors to target and deliver transgenes to Leydig cells. RESULTS: Viral vectors utilized in this study were found to specifically target Leydig cells when delivered interstitially. Transgene expression in lentiviral-targeted Leydig cells was detected for 7 days post-injection before Leydig cells underwent apoptosis. Adenoviral-delivered transgene expression was detected for 10 days post-injection with no evidence of targeted cell apoptosis. We found serotype differences in AAV injected testis with AAV serotype 9 targeting a significant proportion of Leydig cells. Targeting efficiency increased to an average of 59.63% (and a maximum of 80%) of Leydig cells with the addition of neuraminidase during injection. In AAV injected testis sections, transgene expression was detectable for up to 50 days post-injection. DISCUSSION & CONCLUSION: Lentivirus, Adenovirus and Adeno-Associated virus delivery to the testis resulted in key variances in targeting efficiency of Leydig cells and in longevity of transgene expression, but identified AAV9 + Neuraminidase as an efficient vector system for transgene delivery and long-term expression. Simple viral delivery procedures and the commercial availability of viral vectors suggests AAV9 + Neuraminidase will be of significant utility to researchers investigating the genetics underpinning Leydig cell function and holds promise to inform the development of novel therapeutics for the treatment of male reproductive disorders.


Asunto(s)
Dependovirus , Técnicas de Transferencia de Gen , Vectores Genéticos , Células Intersticiales del Testículo , Adenoviridae , Animales , Lentivirus , Masculino , Ratones
14.
Sci Rep ; 10(1): 19022, 2020 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-33149175

RESUMEN

Focal dysgenesis is a consistent feature of testicular dysgenesis syndrome (TDS) in humans. Rodent studies show that perturbation of androgens (e.g. following phthalate exposure) during a fetal masculinisation programming window (MPW) predisposes to a TDS phenotype. This study aimed to determine whether dissociation and reconstitution of rat fetal testis tissue during the MPW can be used to model and manipulate seminiferous cord development, including induction of focal dysgenesis, as described in TDS. Dissociated fetal rat testes were xenotransplanted subcutaneously into recipient mice for 4 weeks. Transplanted mice were treated with vehicle or di-n-butyl-phthalate (DBP, a plasticising chemical known to induce testicular dysgenesis in vivo in rats). Testosterone production by the transplants was measured in recipient mice and immunofluorescence was performed on the retrieved transplants to identify features consistent with focal testicular dysgenesis. Re-aggregation of rat fetal testis tissue xenotransplants during the MPW results in reconstitution of seminiferous cords. Features of focal testicular dysgenesis were present in re-aggregated testis, including ectopic Sertoli cells and intratubular Leydig cells (ITLCs). DBP exposure of recipient mice reduced androgen-dependent seminal vesicle weight (8.3 vs 26.7 mg; p < 0.05), but did not enhance features of focal dysgenesis including number of ITLCs (0.07 vs 0.10 cells/mm2; p > 0.05). We conclude that seminiferous cord reformation during the MPW results in development of focal dysgenesis. The system may be used to separate specific effects (e.g. androgen suppression) of individual chemical exposures from other mechanisms that may be conserved in TDS.


Asunto(s)
Disgenesia Gonadal/patología , Testículo/embriología , Animales , Femenino , Masculino , Embarazo , Ratas , Ratas Wistar , Testículo/crecimiento & desarrollo
15.
Cancers (Basel) ; 12(10)2020 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-33008013

RESUMEN

Survivors of childhood cancer are at risk for long-term treatment-induced health sequelae, including gonadotoxicity and iatrogenic infertility. At present, for prepubertal boys there are no viable clinical options to preserve future reproductive potential. We investigated the effect of a pubertal induction regimen with gonadotrophins on prepubertal human testis xenograft development. Human testis tissue was obtained from patients with cancer and non-malignant haematological disorders (n = 6; aged 1-14 years) who underwent testis tissue cryopreservation for fertility preservation. Fresh and frozen-thawed testis fragments were transplanted subcutaneously or intratesticularly into immunocompromised mice. Graft-bearing mice received injections of vehicle or exogenous gonadotrophins, human chorionic gonadotrophin (hCG, 20 IU), and follicle-stimulating hormone (FSH, 12.5 IU) three times a week for 12 weeks. The gross morphology of vehicle and gonadotrophin-exposed grafts was similar for both transplantation sites. Exposure of prepubertal human testis tissue xenografts to exogenous gonadotrophins resulted in limited endocrine function of grafts, as demonstrated by the occasional expression of the steroidogenic cholesterol side-chain cleavage enzyme (CYP11A1). Plasma testosterone concentrations (0.13 vs. 0.25 ng/mL; p = 0.594) and seminal vesicle weights (10.02 vs. 13.93 mg; p = 0.431) in gonadotrophin-exposed recipient mice were comparable to vehicle-exposed controls. Regardless of the transplantation site and treatment, initiation and maintenance of androgen receptor (AR) expression were observed in Sertoli cells, indicating commitment towards a more differentiated status. However, neither exogenous gonadotrophins (in castrated host mice) nor endogenous testosterone (in intact host mice) were sufficient to repress the expression of markers associated with immature Sertoli cells, such as anti-Müllerian hormone (AMH) and Ki67, or to induce the redistribution of junctional proteins (connexin 43, CX43; claudin 11, CLDN11) to areas adjacent to the basement membrane. Spermatogonia did not progress developmentally but remained the most advanced germ cell type in testis xenografts. Overall, these findings demonstrate that exogenous gonadotrophins promote partial activation and maturation of the somatic environment in prepubertal testis xenografts. However, alternative hormone regimens or additional factors for pubertal induction are required to complete the functional maturation of the spermatogonial stem cell (SSC) niche.

16.
J Neuroendocrinol ; 32(10): e12903, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32959418

RESUMEN

The anterior and intermediate lobes of the pituitary are composed of endocrine cells, as well as vasculature and supporting cells, such as folliculostellate cells. Folliculostellate cells form a network with several postulated roles in the pituitary, including production of paracrine signalling molecules and cytokines, coordination of endocrine cell hormone release, phagocytosis, and structural support. Folliculostellate cells in rats are characterised by expression of S100B protein, and in humans by glial fibrillary acid protein. However, there is evidence for another network of supporting cells in the anterior pituitary that has properties of mural cells, such as vascular smooth muscle cells and pericytes. The present study aims to characterise the distribution of cells that express the mural cell marker platelet derived growth factor receptor beta (PDGFRß) in the mouse pituitary and establish whether these cells are folliculostellate. By immunohistochemical localisation, we determine that approximately 80% of PDGFRß+ cells in the mouse pituitary have a non-perivascular location and 20% are pericytes. Investigation of gene expression in a magnetic cell sorted population of PDGFRß+ cells shows that, despite a mostly non-perivascular location, this population is enriched for mural cell markers but not enriched for rat or human folliculostellate cell markers. This is confirmed by immunohistochemistry. The present study concludes that a mural cell network is present throughout the anterior pituitary of the mouse and that this population does not express well-characterised human or rat folliculostellate cell markers.


Asunto(s)
Comunicación Celular/fisiología , Hipófisis/citología , Animales , Biomarcadores/metabolismo , Células Endocrinas/citología , Células Endocrinas/fisiología , Células Endoteliales/citología , Células Endoteliales/fisiología , Ratones , Ratones Endogámicos C57BL , Pericitos/citología , Pericitos/fisiología , Hipófisis/metabolismo , Adenohipófisis/citología , Adenohipófisis/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factores de Transcripción SOXB1/metabolismo
17.
FASEB J ; 34(8): 10373-10386, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32557858

RESUMEN

Male development, fertility, and lifelong health are all androgen-dependent. Approximately 95% of circulating testosterone is synthesized by the testis and the final step in this canonical pathway is controlled by the activity of the hydroxysteroid-dehydrogenase-17-beta-3 (HSD17B3). To determine the role of HSD17B3 in testosterone production and androgenization during male development and function we have characterized a mouse model lacking HSD17B3. The data reveal that developmental masculinization and fertility are normal in mutant males. Ablation of HSD17B3 inhibits hyperstimulation of testosterone production by hCG, although basal testosterone levels are maintained despite the absence of HSD17B3. Reintroduction of HSD17B3 via gene-delivery to Sertoli cells in adulthood partially rescues the adult phenotype, showing that, as in development, different cell-types in the testis are able to work together to produce testosterone. Together, these data show that HS17B3 acts as a rate-limiting-step for the maximum level of testosterone production by the testis but does not control basal testosterone production. Measurement of other enzymes able to convert androstenedione to testosterone identifies HSD17B12 as a candidate enzyme capable of driving basal testosterone production in the testis. Together, these findings expand our understanding of testosterone production in males.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas/metabolismo , Células de Sertoli/metabolismo , Testículo/metabolismo , Testosterona/metabolismo , Andrógenos/metabolismo , Animales , Femenino , Fertilidad/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
18.
J Dent Hyg ; 94(3): 48-55, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32554415

RESUMEN

Purpose: Many school-aged children have not received dental care in West Virginia, despite mandated statewide requirements of a dental evaluation and dental treatment before entering school, and the provision of Medicaid/CHIP insurance coverage for children from families below the federal poverty level. An innovative mobile oral health program to educate children, provide preventive care, and bring technology to public schools was developed for West Virginia children in a need shortage area. It was unknown if the unmet dental needs challenge was greater for male or female children residing in that area. The purpose of this study was to determine whether there was a difference by sex in the number of attendees and the incidence of dental caries for children who visited a school-based mobile dental facility.Methods: School-aged children who had not had a dental examination within the previous year were offered school-based examinations/assessments, preventive care, and oral health education via a mobile oral health program following parental/guardian consent. Data were collected concerning the number of current carious teeth in need of restoration. Descriptive statistics and chi square analyses were conducted to analyze the data.Results: There were 429 students evaluated at the school-based mobile dental facility. Half (50.3%) were male. Referrals for additional necessary oral/medical care were made for 214 (50.1%) children; 45.9% of males and 53.3% of females (p= 0.287) had dental caries.Conclusion: Results from this study indicate that sex was not a statistically significant factor in school-based mobile dental facility attendance nor in current dental caries incidence among school-aged children in an underserved area of West Virginia.


Asunto(s)
Caries Dental , Niño , Atención Odontológica , Instituciones Odontológicas , Femenino , Humanos , Masculino , Salud Bucal , West Virginia
19.
J Clin Med ; 9(1)2020 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-31963729

RESUMEN

The future fertility of prepubertal boys with cancer may be irreversibly compromised by chemotherapy and/or radiotherapy. Successful spermatogenesis has not been achieved following the xenotransplantation of prepubertal human testis tissue, which is likely due to the failure of somatic cell maturation and function. We used a validated xenograft model to identify the factors required for Leydig and Sertoli cell development and function in immature human testis. Importantly, we compared the maturation status of Sertoli cells in xenografts with that of human testis tissues (n = 9, 1 year-adult). Human fetal testis (n = 6; 14-21 gestational weeks) tissue, which models many aspects of prepubertal testicular development, was transplanted subcutaneously into castrated immunocompromised mice for ~12 months. The mice received exogenous human chorionic gonadotropin (hCG; 20IU, 3×/week). In xenografts exposed continuously to hCG, we demonstrate the maintenance of Leydig cell steroidogenesis, the acquisition of features of Sertoli cell maturation (androgen receptor, lumen development), and the formation of the blood-testis barrier (connexin 43), none of which were present prior to the transplantation or in xenografts in which hCG was withdrawn after 7 months. These studies provide evidence that hCG plays a role in Sertoli cell maturation, which is relevant for future investigations, helping them generate functional gametes from immature testis tissue for clinical application.

20.
Front Endocrinol (Lausanne) ; 11: 599869, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33584538

RESUMEN

Adrenal androgens are fundamental mediators of ovarian folliculogenesis, embryonic implantation, and breast development. Although adrenal androgen function in target tissues are well characterized, there is little research covering the role of androgen-signaling within the adrenal itself. Adrenal glands express AR which is essential for the regression of the X-zone in male mice. Female mice also undergo X-zone regression during their first pregnancy, however whether this is also controlled by AR signaling is unknown. To understand the role of the androgen receptor (AR) in the female adrenal, we utilized a Cyp11a1-Cre to specifically ablate AR from the mouse adrenal cortex. Results show that AR-signaling is dispensable for adrenal gland development in females, and for X-zone regression during pregnancy, but is required to suppress elevation of corticosterone levels post-partum. Additionally, following disruption to adrenal AR, aberrant spindle cell development is observed in young adult females. These results demonstrate sexually dimorphic regulation of the adrenal X-zone by AR and point to dysfunctional adrenal androgen signaling as a possible mechanism in the early development of adrenal spindle cell hyperplasia.


Asunto(s)
Corteza Suprarrenal/citología , Andrógenos/farmacología , Corticosterona/metabolismo , Periodo Posparto/metabolismo , Sustancias Protectoras/farmacología , Receptores Androgénicos/química , Corteza Suprarrenal/efectos de los fármacos , Corteza Suprarrenal/metabolismo , Animales , Femenino , Masculino , Ratones , Periodo Posparto/efectos de los fármacos , Embarazo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA