Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Int J Mol Sci ; 23(23)2022 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-36499726

RESUMEN

Understanding the regulation of the testicular endocrine function leading to testosterone production is a major objective as the alteration of endocrine function is associated with the development of many diseases such as infertility. In the last decades, it has been demonstrated that several endogenous molecules regulate the steroidogenic pathway. Among them, bile acids have recently emerged as local regulators of testicular physiology and particularly endocrine function. Bile acids act through the nuclear receptor FXRα (Farnesoid-X-receptor alpha; NR1H4) and the G-protein-coupled bile acid receptor (GPBAR-1; TGR5). While FXRα has been demonstrated to regulate testosterone synthesis within Leydig cells, no data are available regarding TGR5. Here, we investigated the potential role of TGR5 within Leydig cells using cell culture approaches combined with pharmacological exposure to the TGR5 agonist INT-777. The data show that activation of TGR5 results in a decrease in testosterone levels. TGR5 acts through the PKA pathway to regulate steroidogenesis. In addition, our data show that TGR5 activation leads to an increase in cholesterol ester levels. This suggests that altered lipid homeostasis may be a mechanism explaining the TGR5-induced decrease in testosterone levels. In conclusion, the present work highlights the impact of the TGR5 signaling pathway on testosterone production and reinforces the links between bile acid signaling pathways and the testicular endocrine function. The testicular bile acid pathways need to be further explored to increase our knowledge of pathologies associated with impaired testicular endocrine function, such as fertility disorders.


Asunto(s)
Ácidos y Sales Biliares , Células Intersticiales del Testículo , Masculino , Humanos , Células Intersticiales del Testículo/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Homeostasis , Testosterona
2.
Adv Sci (Weinh) ; 9(17): e2200626, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35435331

RESUMEN

Spermatogonial stem cells regenerate and maintain spermatogenesis throughout life, making testis a good model for studying stem cell biology. The effects of chemotherapy on fertility have been well-documented previously. This study investigates how busulfan, an alkylating agent that is often used for chemotherapeutic purposes, affects male fertility. Specifically, the role of the TGR5 pathway is investigated on spermatogonia homeostasis using in vivo, in vitro, and pharmacological methods. In vivo studies are performed using wild-type and Tgr5-deficient mouse models. The results clearly show that Tgr5 deficiency can facilitate restoration of the spermatogonia homeostasis and allow faster resurgence of germ cell lineage after exposure to busulfan. TGR5 modulates the expression of key genes of undifferentiated spermatogonia such as Gfra1 and Fgfr2. At the molecular level, the present data highlight molecular mechanisms underlying the interactions among the TGR5, GLIS2, and TP53 pathways in spermatogonia associated with germ cell apoptosis following busulfan exposure. This study makes a significant contribution to the literature because it shows that TGR5 plays key role on undifferentiated germ cell homeostasis and that modulating the TGR5 signaling pathway could be used as a potential therapeutic tool for fertility disorders.


Asunto(s)
Busulfano , Resistencia a Antineoplásicos , Factores de Transcripción de Tipo Kruppel , Proteínas del Tejido Nervioso , Receptores Acoplados a Proteínas G , Proteína p53 Supresora de Tumor , Animales , Busulfano/metabolismo , Busulfano/farmacología , Homeostasis , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Proteínas del Tejido Nervioso/genética , Receptores Acoplados a Proteínas G/genética , Transducción de Señal , Espermatogonias/metabolismo , Proteína p53 Supresora de Tumor/genética
3.
Sci Total Environ ; 829: 154383, 2022 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-35276143

RESUMEN

Volcanic ash exposure can lead to significant health risks. Damage to the respiratory and pulmonary systems are the most evident toxic side effects although the causes of these symptoms remain unclear. Conversely, the effects on other organs remain largely under-explored, limiting our understanding of the long-term volcanic ash-related risk at the whole-body scale. The metallome i.e. metal concentrations and isotopic compositions within the body, is suspected to be affected by volcanic ash exposure, having thus the potential for capturing some specificities of ash toxicity. However, the means by and extent to which the metallome is affected at the entire body scale and how the consequent chemical and isotopic deregulations correlate with pathophysiological dysfunctions are currently poorly understood. Here, we adopt a transdisciplinary approach combining high precision chemical analyses (major and trace element concentrations) and CuZn isotope measurements in seven organs and two biological fluids of isogenic mice (C57BL/6) exposed to eruption products from La Soufrière de Guadeloupe (Eastern Carribean), in tandem with biological parameters including physiological and morphological data. Based on principal component analysis, we show that after one month of exposure to volcanic ash deposits, the mice metallome; originally organ-specific and isotopically-typified, is highly disrupted as shown for example by heavy metal accumulation in testis (e.g., Fe, Zn) and Cu, Zn isotopic divergence in liver, intestine and blood. These metallomic variations are correlated with early testicular defects and might reflect the warning signs of premature (entero)hepatic impairments that may seriously affect fertility and favor the emergence of liver diseases after prolonged exposure. Monitoring the temporal evolution of the Cu and Zn isotope compositions seems to be a promising technique to identify the main biological processes and vital functions that are vulnerable to environmental volcanogenic pollutants although this will require further validation on human subjects.


Asunto(s)
Metales , Erupciones Volcánicas , Animales , Humanos , Isótopos , Masculino , Ratones , Ratones Endogámicos C57BL , Erupciones Volcánicas/efectos adversos
5.
Biol Reprod ; 106(3): 463-476, 2022 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-34875016

RESUMEN

Infertility represents a growing burden worldwide, with one in seven couples presenting difficulties conceiving. Among these, 10-15% of the men have idiopathic infertility that does not correlate with any defect in the classical sperm parameters measured. In the present study, we used a mouse model to investigate the effects of maternal undernutrition on fertility in male progeny. Our results indicate that mothers fed on a low-protein diet during gestation and lactation produce male offspring with normal sperm morphology, concentration, and motility but exhibiting an overall decrease of fertility when they reach adulthood. Particularly, in contrast to control, sperm from these offspring show a remarkable lower capacity to fertilize oocytes when copulation occurs early in the estrus cycle relative to ovulation, due to an altered sperm capacitation. Our data demonstrate for the first time that maternal nutritional stress can have long-term consequences on the reproductive health of male progeny by affecting sperm physiology, especially capacitation, with no observable impact on spermatogenesis and classical quantitative and qualitative sperm parameters. Moreover, our experimental model could be of major interest to study, explain, and ultimately treat certain categories of infertilities.


Asunto(s)
Infertilidad Masculina , Desnutrición , Adulto , Animales , Femenino , Fertilidad , Humanos , Infertilidad Masculina/etiología , Lactancia , Masculino , Desnutrición/complicaciones , Ratones , Embarazo , Capacitación Espermática , Motilidad Espermática , Espermatozoides/fisiología
6.
Cells ; 10(9)2021 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-34572051

RESUMEN

Spermatogenesis is a process within the testis that leads to the production of spermatozoa. It is based on a population of spermatogonial stem cells, which have the capacity to self-renew and to differentiate throughout life to ensure the functions of reproduction are maintained. Male fertility disorders are responsible for half of the cases of infertility in couples worldwide. It is well known that cancer treatments are associated with reversible or irreversible fertility disorders. Busulfan (Bu) is an alkylating agent that significantly inhibits spermatogenesis. The present study relied on a combination of in vivo and in vitro approaches as well as RNAseq analysis to characterize the effects of Bu, in which mouse testes were used as a model. An in silico analysis revealed that many of the Bu-modulated genes are potentially regulated by the SIN3 Transcription Regulator Family Member A (SIN3A) and E2F Transcription Factor (E2F) families of transcription factors. The results demonstrate that the deregulated genes function in processes related to the cell cycle, DNA repair, and cell death mechanisms, including the Tumor Protein 53 (TP53) pathway. This reinforces the role of the TP53 signaling pathway as a major player in Bu effects. In addition, Bu altered the patterns of mRNA accumulation for various genes in undifferentiated spermatogonia. This work provides significant insight into the kinetics and impacts of busulfan, which could pave the way for developing strategies to minimize the impact of chemodrugs and, thus, could lead to germ cell lineage regeneration following anticancer treatments.


Asunto(s)
Busulfano/farmacología , Fertilidad/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Inmunosupresores/farmacología , Testículo/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , RNA-Seq , Testículo/metabolismo
7.
Mol Aspects Med ; 78: 100956, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33676761
10.
Mol Cell Endocrinol ; 518: 110995, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32827571

RESUMEN

The hypothalamic-pituitary axis exert a major control over endocrine and exocrine testicular functions. The hypothalamic-pituitary axis corresponds to a cascade with the Gonadotropin Releasing Hormone secreted by the hypothalamus, which stimulates the synthesis and the release of Luteinizing Hormone (LH) and Follicle Stimulating Hormone by the gonadotropic cells of the anterior pituitary. The LH signaling pathway controls the steroidogenic activity of the Leydig cells via the activation of the luteinizing hormone/choriogonadotropin receptor. In order to avoid a runaway system, sex steroids exert a negative feedback within hypothalamus and pituitary. Testicular steroidogenesis is locally controlled within Leydig cells. The present work reviews some local regulations of steroidogenesis within the Leydig cells focusing mainly on the roles of the Farnesoid-X-Receptor-alpha and its interactions with several orphan members of the nuclear receptor superfamily. Further studies are required to reinforce our knowledge of the regulation of testicular endocrine function, which is necessary to ensure a better understanding of fertility disorders and then proposed an adequate treatment of the diseases.


Asunto(s)
Hormonas Gonadales/metabolismo , Células Intersticiales del Testículo/metabolismo , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Células Endocrinas/metabolismo , Regulación de la Expresión Génica , Masculino , Ratones , Hipófisis/metabolismo , Unión Proteica , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal/fisiología
11.
J Steroid Biochem Mol Biol ; 194: 105460, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31470110

RESUMEN

The bile acid receptor Farnesoid-X-Receptor alpha (FXRα), a member of the nuclear receptor superfamily, is well known for its roles in the enterohepatic tract. In addition, FXRα regulates testicular physiology through the control of both endocrine and exocrine functions. The endocrine function of the Leydig cells is mainly controlled by the hypothalamo-pituitary axis viaLH/chorionic gonadotropin (CG). If FXRα was demonstrated to control the expression of the Lhcgr gene, encoding the LH receptor; the impact of the LH/CG signaling on the Fxrα expression has not been defined so far. Here, we demonstrate that hCG increases the Fxrα gene expression through the protein kinase-A signaling pathway. Fxrα is then involved in a negative feedback of steroid synthesis. These data improve our knowledge of the local control of the testicular steroidogenesis with the identification of the link between the hypothalamo-pituitary axis and the FXRα signaling pathway.


Asunto(s)
Gonadotropina Coriónica/farmacología , Receptores Citoplasmáticos y Nucleares/genética , Testículo/efectos de los fármacos , Animales , Línea Celular , Masculino , Ratones Endogámicos C57BL , Fosfoproteínas/genética , Progesterona/metabolismo , Receptores de HL/genética , Transducción de Señal/efectos de los fármacos , Testículo/metabolismo , Testosterona/sangre , Testosterona/metabolismo
12.
Cell Mol Life Sci ; 76(24): 4849-4859, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31407019

RESUMEN

The farnesoid-X-receptorα (FXRα; NR1H4) is one of the main bile acid (BA) receptors. During the last decades, through the use of pharmalogical approaches and transgenic mouse models, it has been demonstrated that the nuclear receptor FXRα controls numerous physiological functions such as glucose or energy metabolisms. It is also involved in the etiology or the development of several pathologies. Here, we will review the unexpected roles of FXRα on the male reproductive tract. FXRα has been demonstrated to play functions in the regulation of testicular and prostate homeostasis. Even though additional studies are needed to confirm these findings in humans, the reviewed reports open new field of research to better define the effects of bile acid-FXRα signaling pathways on fertility disorders and cancers.


Asunto(s)
Genitales Masculinos/crecimiento & desarrollo , Próstata/crecimiento & desarrollo , Receptores Citoplasmáticos y Nucleares/genética , Testículo/crecimiento & desarrollo , Animales , Ácidos y Sales Biliares/metabolismo , Genitales Masculinos/metabolismo , Homeostasis , Humanos , Masculino , Ratones , Próstata/metabolismo , Transducción de Señal/genética , Testículo/metabolismo , Factores de Transcripción/genética
13.
Sci Rep ; 8(1): 16875, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30443025

RESUMEN

Besides their well-known roles in digestion and fat solubilization, bile acids (BAs) have been described as signaling molecules activating the nuclear receptor Farnesoid-X-receptor (FXRα) or the G-protein-coupled bile acid receptor-1 (GPBAR-1 or TGR5). In previous reports, we showed that BAs decrease male fertility due to abnormalities of the germ cell lineage dependent on Tgr5 signaling pathways. In the presentstudy, we tested whether BA exposure could impact germ cell DNA integrity leading to potential implications for progeny. For that purpose, adult F0 male mice were fed a diet supplemented with cholic acid (CA) or the corresponding control diet during 3.5 months prior mating. F1 progeny from CA exposed founders showed higher perinatal lethality, impaired BA homeostasis and reduced postnatal growth, as well as altered glucose metabolism in later life. The majority of these phenotypic traits were maintained up to the F2 generation. In F0 sperm cells, differential DNA methylation associated with CA exposure may contribute to the initial programming of developmental and metabolic defects observed in F1 and F2 offspring. Tgr5 knock-out mice combined with in vitro strategies defined the critical role of paternal Tgr5 dependent pathways in the multigenerational impacts of ancestral CA exposure.


Asunto(s)
Bilis/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Animales , Linaje de la Célula/efectos de los fármacos , Ácido Cólico/farmacología , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Dieta , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Fenotipo , Transducción de Señal/efectos de los fármacos , Espermatozoides/citología , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo , ADN Metiltransferasa 3B
14.
Int J Mol Sci ; 19(11)2018 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-30453651

RESUMEN

Structural and functional studies have provided numerous insights over the past years on how members of the nuclear hormone receptor superfamily tightly regulate the expression of drug-metabolizing enzymes and transporters. Besides the role of the farnesoid X receptor (FXR) in the transcriptional control of bile acid transport and metabolism, this review provides an overview on how this metabolic sensor prevents the accumulation of toxic byproducts derived from endogenous metabolites, as well as of exogenous chemicals, in coordination with the pregnane X receptor (PXR) and the constitutive androstane receptor (CAR). Decrypting this network should provide cues to better understand how these metabolic nuclear receptors participate in physiologic and pathologic processes with potential validation as therapeutic targets in human disabilities and cancers.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Enfermedad , Salud , Receptores Citoplasmáticos y Nucleares/metabolismo , Xenobióticos/metabolismo , Animales , Ácidos y Sales Biliares/química , Humanos , Inactivación Metabólica
15.
Stem Cell Reports ; 11(4): 944-958, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30245210

RESUMEN

Several studies have reported an association between the farnesoid X receptor alpha (FXRα) and estrogenic signaling pathways. Fxrα could thus be involved in the reprotoxic effects of endocrine disruptors such as bisphenol-A (BPA). To test this hypothesis, mice were exposed to BPA and/or stigmasterol (S), an FXRα antagonist. Following the exposure to both molecules, wild-type animals showed impaired fertility and lower sperm cell production associated with the alteration of the establishment and maintenance of the undifferentiated germ cell pool. The crosstalk between BPA and FXRα is further supported by the lower impact of BPA in mice genetically ablated for Fxrα and the fact that BPA counteracted the effects of FXRα agonists. These effects might result from the downregulation of Fxrα expression following BPA exposure. BPA and S act additively in human testis. Our data demonstrate that FXRα activity modulates the impact of BPA on male gonads and on undifferentiated germ cell population.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Diferenciación Celular , Células Germinativas/patología , Homeostasis , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Fenoles/toxicidad , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal , Adulto , Animales , Animales Recién Nacidos , Diferenciación Celular/efectos de los fármacos , Feto/efectos de los fármacos , Feto/patología , Células Germinativas/efectos de los fármacos , Células Germinativas/metabolismo , Homeostasis/efectos de los fármacos , Humanos , Masculino , Ratones , Persona de Mediana Edad , Células de Sertoli/efectos de los fármacos , Células de Sertoli/metabolismo , Transducción de Señal/efectos de los fármacos , Estigmasterol/toxicidad
16.
Artículo en Inglés | MEDLINE | ID: mdl-30072948

RESUMEN

Cholesterol is essential for mammalian cell functions and integrity. It is an important structural component maintaining the permeability and fluidity of the cell membrane. The balance between synthesis and catabolism of cholesterol should be tightly regulated to ensure normal cellular processes. Male reproductive function has been demonstrated to be dependent on cholesterol homeostasis. Here we review data highlighting the impacts of cholesterol homeostasis on male fertility and the molecular mechanisms implicated through the signaling pathways of some nuclear receptors.

18.
Nat Commun ; 8(1): 445, 2017 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-28874658

RESUMEN

Advanced prostate cancer (PCa) is a clinical challenge as no curative therapeutic is available. In this context, a better understanding of metastasis and resistance mechanisms in PCa is an important issue. As phosphatase and tensin homolog (PTEN) loss is the most common genetic lesion in such cancer, we investigate human data sets for mechanisms that can constrain cancer evolution in this setting. Here we report a liver X receptor (LXR) signature, which tightly correlates with PTEN loss, in PCa. Accordingly, the LXR pathway is deregulated in prostate carcinomas in Pten-null mice. Genetic ablation of LXRs in Pten-null mice, exacerbates PCa invasiveness and metastatic dissemination, which involves mesenchymal transition and accumulation of matrix metalloproteinases. Mechanistically, PTEN deletion governed LXR transcriptional activity through deregulation of cholesterol de novo synthesis, resulting in accumulation of endogenous LXR ligands. Our study therefore reveals a functional circuit linking PTEN and LXR, and highlights LXRs as metabolic gatekeepers that are able to constrain PCa progression.Treatment of prostate cancer, especially in its advanced stage, is still challenging; therefore, strategies to prevent metastatic dissemination are of great interest. Here the authors reveal a crucial role for liver X receptors in suppressing prostate carcinogenesis and metastatic progression in PTEN-null tumors.


Asunto(s)
Receptores X del Hígado/genética , Fosfohidrolasa PTEN/genética , Neoplasias de la Próstata/genética , Transducción de Señal/genética , Animales , Línea Celular Tumoral , Células Cultivadas , Colesterol/metabolismo , Progresión de la Enfermedad , Perfilación de la Expresión Génica/métodos , Humanos , Estimación de Kaplan-Meier , Receptores X del Hígado/deficiencia , Masculino , Ratones Noqueados , Metástasis de la Neoplasia , Fosfohidrolasa PTEN/deficiencia , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología
19.
Stem Cell Reports ; 9(1): 315-328, 2017 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-28669602

RESUMEN

Spermatogenesis is the process by which spermatozoa are generated from spermatogonia. This cell population is heterogeneous, with self-renewing spermatogonial stem cells (SSCs) and progenitor spermatogonia that will continue on a path of differentiation. Only SSCs have the ability to regenerate and sustain spermatogenesis. This makes the testis a good model to investigate stem cell biology. The Farnesoid X Receptor alpha (FXRα) was recently shown to be expressed in the testis. However, its global impact on germ cell homeostasis has not yet been studied. Here, using a phenotyping approach in Fxrα-/- mice, we describe unexpected roles of FXRα on germ cell physiology independent of its effects on somatic cells. FXRα helps establish and maintain an undifferentiated germ cell pool and in turn influences male fertility. FXRα regulates the expression of several pluripotency factors. Among these, in vitro approaches show that FXRα controls the expression of the pluripotency marker Lin28 in the germ cells.


Asunto(s)
Receptores Citoplasmáticos y Nucleares/metabolismo , Espermatogénesis , Espermatozoides/citología , Envejecimiento , Animales , Células Cultivadas , Femenino , Fertilidad , Eliminación de Gen , Regulación de la Expresión Génica , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/metabolismo , Masculino , Ratones Endogámicos C57BL , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Reproducción , Células de Sertoli/citología , Células de Sertoli/metabolismo , Espermatozoides/metabolismo , Testículo/citología , Testículo/metabolismo , Testículo/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...