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










Publication year range
1.
Oncogene ; 42(38): 2854-2867, 2023 09.
Article in English | MEDLINE | ID: mdl-37587334

ABSTRACT

Increasing evidence points towards a causal link between exposure to persistent organic pollutants (POPs) with increased incidence and aggressivity of various cancers. Among these POPs, dioxin and PCB-153 are widely found in our environment and represent a significant source of contamination. Dioxin exposure has already been linked to cancer such as non-Hodgkin's lymphoma, but remains to be more extensively investigated in other cancers. Potential implications of dioxin and PCB-153 in prostate cancer progression spurred us to challenge both ex vivo and in vivo models with low doses of these POPs. We found that dioxin or PCB-153 exposure increased hallmarks of growth and metastasis of prostate cancer cells ex vivo and in grafted NOD-SCID mice. Exposure induced histopathological carcinoma-like patterns in the Ptenpc-/- mice. We identified up-regulation of Acetyl-CoA Acetyltransferase-1 (ACAT1) involved in ketone bodies pathway as a potential target. Mechanistically, genetic inhibition confirmed that ACAT1 mediated dioxin effect on cell migration. Using public prostate cancer datasets, we confirmed the deregulation of ACAT1 and associated gene encoded ketone bodies pathway enzymes such as OXCT1, BDH1 and HMGCL in advanced prostate cancer. To further explore this link between dioxin and ACAT1 deregulation, we analyzed a unique prostate-tumour tissue collection from the USA veterans exposed to agent orange, known to be highly contaminated by dioxin because of industrial production. We found that ACAT1 histoscore is significantly increased in exposed patients. Our studies reveal the implication of dioxin and PCB-153 to induce a prometastatic programme in prostate tumours and identify ACAT1 deregulation as a key event in this process.


Subject(s)
Dioxins , Polychlorinated Dibenzodioxins , Prostatic Neoplasms , Male , Humans , Animals , Mice , Mice, Inbred NOD , Mice, SCID , Persistent Organic Pollutants , Dioxins/toxicity , Prostatic Neoplasms/chemically induced , Prostatic Neoplasms/genetics , Acetyltransferases
2.
Int J Mol Sci ; 23(23)2022 Dec 06.
Article in English | MEDLINE | ID: mdl-36499726

ABSTRACT

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.


Subject(s)
Bile Acids and Salts , Leydig Cells , Male , Humans , Leydig Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Homeostasis , Testosterone
3.
Adv Sci (Weinh) ; 9(17): e2200626, 2022 06.
Article in English | MEDLINE | ID: mdl-35435331

ABSTRACT

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.


Subject(s)
Busulfan , Drug Resistance, Neoplasm , Kruppel-Like Transcription Factors , Nerve Tissue Proteins , Receptors, G-Protein-Coupled , Tumor Suppressor Protein p53 , Animals , Busulfan/metabolism , Busulfan/pharmacology , Homeostasis , Kruppel-Like Transcription Factors/genetics , Male , Mice , Nerve Tissue Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Spermatogonia/metabolism , Tumor Suppressor Protein p53/genetics
4.
Cells ; 10(9)2021 09 13.
Article in English | MEDLINE | ID: mdl-34572051

ABSTRACT

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.


Subject(s)
Busulfan/pharmacology , Fertility/drug effects , Gene Expression Regulation/drug effects , Immunosuppressive Agents/pharmacology , Testis/drug effects , Animals , Dose-Response Relationship, Drug , Female , Male , Mice , Mice, Inbred C57BL , RNA-Seq , Testis/metabolism
5.
PLoS Biol ; 18(12): e3000948, 2020 12.
Article in English | MEDLINE | ID: mdl-33284790

ABSTRACT

Chronic inflammation is now a well-known precursor for cancer development. Infectious prostatitis are the most common causes of prostate inflammation, but emerging evidence points the role of metabolic disorders as a potential source of cancer-related inflammation. Although the widely used treatment for prostate cancer based on androgen deprivation therapy (ADT) effectively decreases tumor size, it also causes profound alterations in immune tumor microenvironment within the prostate. Here, we demonstrate that prostates of a mouse model invalidated for nuclear receptors liver X receptors (LXRs), crucial lipid metabolism and inflammation integrators, respond in an unexpected way to androgen deprivation. Indeed, we observed profound alterations in immune cells composition, which was associated with chronic inflammation of the prostate. This was explained by the recruitment of phagocytosis-deficient macrophages leading to aberrant hyporesponse to castration. This phenotypic alteration was sufficient to allow prostatic neoplasia. Altogether, these data suggest that ADT and inflammation resulting from metabolic alterations interact to promote aberrant proliferation of epithelial prostate cells and development of neoplasia. This raises the question of the benefit of ADT for patients with metabolic disorders.


Subject(s)
Immunity/physiology , Liver X Receptors/metabolism , Prostate/metabolism , Androgen Antagonists/immunology , Androgens/metabolism , Animals , Disease Models, Animal , Immunity/immunology , Liver X Receptors/genetics , Liver X Receptors/immunology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Neoplasms/etiology , Neoplasms/immunology , Neoplasms/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Receptors, Cytoplasmic and Nuclear/metabolism , Tumor Microenvironment
6.
Mol Cell Endocrinol ; 518: 110995, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32827571

ABSTRACT

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.


Subject(s)
Gonadal Hormones/metabolism , Leydig Cells/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Endocrine Cells/metabolism , Gene Expression Regulation , Male , Mice , Pituitary Gland/metabolism , Protein Binding , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/physiology
7.
J Steroid Biochem Mol Biol ; 194: 105460, 2019 11.
Article in English | MEDLINE | ID: mdl-31470110

ABSTRACT

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.


Subject(s)
Chorionic Gonadotropin/pharmacology , Receptors, Cytoplasmic and Nuclear/genetics , Testis/drug effects , Animals , Cell Line , Male , Mice, Inbred C57BL , Phosphoproteins/genetics , Progesterone/metabolism , Receptors, LH/genetics , Signal Transduction/drug effects , Testis/metabolism , Testosterone/blood , Testosterone/metabolism
8.
Cell Mol Life Sci ; 76(24): 4849-4859, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31407019

ABSTRACT

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.


Subject(s)
Genitalia, Male/growth & development , Prostate/growth & development , Receptors, Cytoplasmic and Nuclear/genetics , Testis/growth & development , Animals , Bile Acids and Salts/metabolism , Genitalia, Male/metabolism , Homeostasis , Humans , Male , Mice , Prostate/metabolism , Signal Transduction/genetics , Testis/metabolism , Transcription Factors/genetics
9.
Eur Urol Oncol ; 2(2): 198-206, 2019 03.
Article in English | MEDLINE | ID: mdl-31017097

ABSTRACT

BACKGROUND: Deregulation of cholesterol metabolism represents a hallmark of prostate cancer (PCa) and promotes its development. OBJECTIVE: To compare cholesterol metabolism on individual paired normal and tumour prostate tissues obtained from patients with PCa. DESIGN, SETTING, AND PARTICIPANTS: Between 2008 and 2012, normal and tumour paired tissue samples were collected from radical prostatectomy specimens from a cohort of 69 patients treated for localised PCa. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Tumour and normal tissues were subjected to gene analysis, sterol measurement, and immunohistochemistry. The Wilcoxon paired test and Spearman test were applied for comparison and correlation analyses, respectively. Principal component analysis was also carried out to investigate relationships between quantitative variables. RESULTS AND LIMITATIONS: Overall, cholesterol concentrations were not significantly different between tissue pairs. However, tumour samples were significantly associated with downregulated de novo cholesterol synthesis, but exhibited 54.7% overexpression of SCARB1 that could increase high-density lipoprotein uptake in PCa. Tumour tissues showed different trafficking of available cholesterol, with significantly lower ACAT1, and an altered efflux via APOE. Furthermore, cholesterol metabolism in tumour tissues was characterised by higher accumulation of 7α-hydroxycholesterol (OHC), 7ßOHC, and 7-ketosterol, and a lower level of 27OHC. CONCLUSIONS: Focusing on individually paired prostate tissues, our results highlighted several differences between normal and tumour samples linked to a metabolic shift in cholesterol flux. PCa samples exhibited a specific tissue signature characterised by higher SCARB1 expression, higher accumulation of OHC species, and clear downregulation of de novo cholesterol synthesis. PATIENT SUMMARY: Comparing normal and tumour tissues from the same prostates, our study identified a set of alterations in prostate cancer samples in terms of their use of cholesterol. These included higher cholesterol uptake, accumulation of oxidised cholesterol derivatives, and autonomous cellular production of cholesterol. Together, these data provide promising clinical targets to fight prostate cancer.


Subject(s)
Cholesterol/metabolism , Gene Regulatory Networks , Prostatic Neoplasms/surgery , Acetyl-CoA C-Acetyltransferase/genetics , Acetyl-CoA C-Acetyltransferase/metabolism , Aged , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Principal Component Analysis , Prostatectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism
10.
Sci Rep ; 8(1): 16875, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30443025

ABSTRACT

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.


Subject(s)
Bile/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Cell Lineage/drug effects , Cholic Acid/pharmacology , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/drug effects , DNA Methylation/genetics , Diet , Gene Expression Regulation/drug effects , Male , Mice, Inbred C57BL , Phenotype , Signal Transduction/drug effects , Spermatozoa/cytology , Spermatozoa/drug effects , Spermatozoa/metabolism , DNA Methyltransferase 3B
11.
Stem Cell Reports ; 9(1): 315-328, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28669602

ABSTRACT

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.


Subject(s)
Receptors, Cytoplasmic and Nuclear/metabolism , Spermatogenesis , Spermatozoa/cytology , Aging , Animals , Cells, Cultured , Female , Fertility , Gene Deletion , Gene Expression Regulation , Leydig Cells/cytology , Leydig Cells/metabolism , Male , Mice, Inbred C57BL , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Reproduction , Sertoli Cells/cytology , Sertoli Cells/metabolism , Spermatozoa/metabolism , Testis/cytology , Testis/metabolism , Testis/ultrastructure
12.
Sci Rep ; 7: 42182, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28181583

ABSTRACT

Bile acids (BAs) are molecules with endocrine activities controlling several physiological functions such as immunity, glucose homeostasis, testicular physiology and male fertility. The role of the nuclear BA receptor FXRα in the control of BA homeostasis has been well characterized. The present study shows that testis synthetize BAs. We demonstrate that mice invalidated for the gene encoding FXRα have altered BA homeostasis in both liver and testis. In the absence of FXRα, BA exposure differently alters hepatic and testicular expression of genes involved in BA synthesis. Interestingly, Fxrα-/- males fed a diet supplemented with BAs show alterations of testicular physiology and sperm production. This phenotype was correlated with the altered testicular BA homeostasis and the production of intermediate metabolites of BAs which led to the modulation of CAR signaling pathways within the testis. The role of the CAR signaling pathways within testis was validated using specific CAR agonist (TCPOBOP) and inverse agonist (androstanol) that respectively inhibited or reproduced the phenotype observed in Fxrα-/- males fed BA-diet. These data open interesting perspectives to better define how BA homeostasis contributes to physiological or pathophysiological conditions via the modulation of CAR activity.


Subject(s)
Bile Acids and Salts/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Reproduction/genetics , Testis/metabolism , Androstanols/pharmacology , Animals , Bile Acids and Salts/biosynthesis , Constitutive Androstane Receptor , Gene Expression Regulation/drug effects , Homeostasis/genetics , Liver/metabolism , Male , Mice , Mice, Knockout , Pyridines/pharmacology , Signal Transduction/drug effects , Spermatozoa/metabolism
13.
Oncotarget ; 7(15): 19468-82, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26848619

ABSTRACT

The bile acid receptor Farnesol-X-Receptor alpha (FRXα) is a member of the nuclear receptor superfamily. FRXα is expressed in the interstitial compartment of the adult testes, which contain the Leydig cells. In adult, short term treatment (12 hours) with FRXα agonist inhibits the expression of steroidogenic genes via the induction of the Small heterodimer partner (SHP). However the consequences of FRXα activation on testicular pathophysiology have never been evaluated. We demonstrate here that mice fed a diet supplemented with bile acid during pubertal age show increased incidence of infertility. This is associated with altered differentiation and increase apoptosis of germ cells due to lower testosterone levels. At the molecular level, next to the repression of basal steroidogenesis via the induction expression of Shp and Dax-1, two repressors of steroidogenesis, the main action of the BA-FRXα signaling is through lowering the Leydig cell sensitivity to the hypothalamo-pituitary axis, the main regulator of testicular endocrine function. In conclusion, BA-FRXα signaling is a critical actor during sexual maturation.


Subject(s)
Bile Acids and Salts/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Sexual Maturation/drug effects , Signal Transduction/drug effects , Animals , Bile Acids and Salts/administration & dosage , Cell Line , Cholic Acid/administration & dosage , Cholic Acid/pharmacology , Fertility/drug effects , Isoxazoles/pharmacology , Leydig Cells/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , RNA Interference , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction/genetics , Testis/cytology , Testis/metabolism , Testosterone/metabolism
14.
PLoS One ; 10(10): e0139946, 2015.
Article in English | MEDLINE | ID: mdl-26439743

ABSTRACT

Bile acids have recently been demonstrated as molecules with endocrine activities controlling several physiological functions such as immunity and glucose homeostases. They act mainly through two receptors, the nuclear receptor Farnesol-X-Receptor alpha (FXRα) and the G-protein coupled receptor (TGR5). These recent studies have led to the idea that molecules derived from bile acids (BAs) and targeting their receptors must be good targets for treatment of metabolic diseases such as obesity or diabetes. Thus it might be important to decipher the potential long term impact of such treatment on different physiological functions. Indeed, BAs have recently been demonstrated to alter male fertility. Here we demonstrate that in mice with overweight induced by high fat diet, BA exposure leads to increased rate of male infertility. This is associated with the altered germ cell proliferation, default of testicular endocrine function and abnormalities in cell-cell interaction within the seminiferous epithelium. Even if the identification of the exact molecular mechanisms will need more studies, the present results suggest that both FXRα and TGR5 might be involved. We believed that this work is of particular interest regarding the potential consequences on future approaches for the treatment of metabolic diseases.


Subject(s)
Bile Acids and Salts/pharmacology , Fertility/drug effects , Infertility, Male/chemically induced , Metabolic Syndrome/metabolism , Overweight/metabolism , Animals , Blood-Testis Barrier/drug effects , Blood-Testis Barrier/metabolism , Cell Proliferation/drug effects , Diet, High-Fat , Infertility, Male/metabolism , Liver/metabolism , Male , Metabolic Syndrome/complications , Mice , Overweight/complications , Signal Transduction
15.
Endocrinology ; 156(2): 660-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25426871

ABSTRACT

The small heterodimer partner (SHP, nuclear receptor subfamily 0, group B, member 2; NR0B2) is an atypical nuclear receptor known mainly for its role in bile acid homeostasis in the enterohepatic tract. We previously showed that NR0B2 controls testicular functions such as testosterone synthesis. Moreover, NR0B2 mediates the deleterious testicular effects of estrogenic endocrine disruptors leading to infertility. The endocrine homeostasis is essential for health, because it controls many physiological functions. This is supported by a large number of studies demonstrating that alterations of steroid activity lead to several kinds of diseases such as obesity and infertility. Within the testis, the functions of the Leydig cells are mainly controlled by the hypothalamo-pituitary axis via LH/chorionic gonadotropin (CG). Here, we show that LH/CG represses Nr0b2 expression through the protein kinase A-AMP protein kinase pathway. Moreover, using a transgenic mouse model invalidated for Nr0b2, we point out that NR0B2 mediates the repression of testosterone synthesis and subsequent germ cell apoptosis induced by exposure to anti-GnRH compound. Together, our data demonstrate a new link between hypothalamo-pituitary axis and NR0B2 in testicular androgen metabolism, making NR0B2 a major actor of testicular physiology in case of alteration of LH/CG levels.


Subject(s)
Hypothalamo-Hypophyseal System/physiology , Leydig Cells/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Testosterone/biosynthesis , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line , Chorionic Gonadotropin , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation , Humans , Male , Mice, Inbred C57BL , Receptors, LHRH/antagonists & inhibitors , Signal Transduction
16.
Hepatology ; 60(3): 1054-65, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24798773

ABSTRACT

UNLABELLED: Bile acids (BAs) are signaling molecules that are involved in many physiological functions, such as glucose and energy metabolism. These effects are mediated through activation of the nuclear and membrane receptors, farnesoid X receptor (FXR-α) and TGR5 (G-protein-coupled bile acid receptor 1; GPBAR1). Although both receptors are expressed within the testes, the potential effect of BAs on testis physiology and male fertility has not been explored thus far. Here, we demonstrate that mice fed a diet supplemented with cholic acid have reduced fertility subsequent to testicular defects. Initially, germ cell sloughing and rupture of the blood-testis barrier occur and are correlated with decreased protein accumulation of connexin-43 (Cx43) and N-cadherin, whereas at later stages, apoptosis of spermatids is observed. These abnormalities are associated with increased intratesticular BA levels in general and deoxycholic acid, a TGR5 agonist, in particular. We demonstrate here that Tgr5 is expressed within the germ cell lineage, where it represses Cx43 expression through regulation of the transcriptional repressor, T-box transcription factor 2 gene. Consistent with this finding, mice deficient for Tgr5 are protected against the deleterious testicular effects of BA exposure. CONCLUSIONS: These data identify the testis as a new target of BAs and emphasize TGR5 as a critical element in testicular pathophysiology. This work may open new perspectives on the potential effect of BAs on testis physiology during liver dysfunction.


Subject(s)
Cholic Acid/metabolism , Fertility , Infertility, Male/metabolism , Receptors, G-Protein-Coupled/metabolism , Testis/drug effects , Animals , Cholic Acid/administration & dosage , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Inbred C57BL , Signal Transduction , Spermatozoa/drug effects , Testosterone/blood
18.
Mol Cell Endocrinol ; 300(1-2): 197-204, 2009 Mar 05.
Article in English | MEDLINE | ID: mdl-19041690

ABSTRACT

The Cre-loxP system combined with gene targeting strategies has proven to be very useful for gene inactivation in specific tissues and/or cell types. To achieve adrenal cortex specific recombination in vivo, we used a 0.5-kb fragment of the 5'-flanking region of the akr1b7 gene to drive Cre expression in adrenocortical cells. The resulting 0.5 akr1b7-Cre mice express Cre in all steroidogenic zones of the adrenal cortex but not in the gonads. Although recombination of the ROSA26R reporter locus was not observed in all cortical cells, we provide evidence that Cre is expressed in all the cells of the cortex in adult mice. In addition, Cre activity was found in collecting ducts and maturing glomeruli of the kidney. This line is the first to show specific Cre expression in the adrenal cortex in the absence of Cre expression in the gonads. This transgene thus provides a valuable tool for specific gene recombination in the adrenal cortex and kidney.


Subject(s)
Adrenal Cortex/enzymology , Integrases/metabolism , Mice, Transgenic , Adrenal Cortex/cytology , Adrenal Cortex/embryology , Animals , Female , Gonads/cytology , Gonads/enzymology , Integrases/genetics , Kidney/cytology , Kidney/enzymology , Male , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...