Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
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
2.
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
3.
Sci Total Environ ; 829: 154383, 2022 Jul 10.
Article in English | MEDLINE | ID: mdl-35276143

ABSTRACT

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.


Subject(s)
Metals , Volcanic Eruptions , Animals , Humans , Isotopes , Male , Mice , Mice, Inbred C57BL , Volcanic Eruptions/adverse effects
5.
Cells ; 10(9)2021 09 10.
Article in English | MEDLINE | ID: mdl-34572036

ABSTRACT

Prostate cancer is the most common cancer in aging men. Despite recent progress, there are still few effective treatments to cure its aggressive and metastatic stages. A better understanding of the molecular mechanisms driving disease initiation and progression appears essential to support the development of more efficient therapies and improve patient care. To do so, multiple research models, such as cell culture and mouse models, have been developed over the years and have improved our comprehension of the biology of the disease. Recently, a new model has been added with the use of the Drosophila accessory gland. With a high level of conservation of major signaling pathways implicated in human disease, this functional equivalent of the prostate represents a powerful, inexpensive, and rapid in vivo model to study epithelial carcinogenesis. The purpose of this review is to quickly overview the existing prostate cancer models, including their strengths and limitations. In particular, we discuss how the Drosophila accessory gland can be integrated as a convenient complementary model by bringing new understanding in the mechanisms driving prostate epithelial tumorigenesis, from initiation to metastatic formation.


Subject(s)
Disease Models, Animal , Drosophila/physiology , Genitalia, Male/pathology , Prostatic Neoplasms/pathology , Animals , Humans , Male
6.
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
7.
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
8.
Nat Commun ; 11(1): 2300, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32385236

ABSTRACT

One of the most important but less understood step of epithelial tumourigenesis occurs when cells acquire the ability to leave their epithelial compartment. This phenomenon, described as basal epithelial cell extrusion (basal extrusion), represents the first step of tumour invasion. However, due to lack of adequate in vivo model, implication of emblematic signalling pathways such as Ras/Mitogen-Activated Protein Kinase (MAPK) and phosphoinositide 3 kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signalling pathways, is scarcely described in this phenomenon. We have developed a unique model of basal extrusion in the Drosophila accessory gland. There, we demonstrate that both Ras/MAPK and PI3K/AKT/mTOR pathways are necessary for basal extrusion. Furthermore, as in prostate cancer, we show that these pathways are co-activated. This occurs through set up of Epidermal Growth Factor Receptor (EGFR) and Insulin Receptor (InR) dependent autocrine loops, a phenomenon that, considering human data, could be relevant for prostate cancer.


Subject(s)
Drosophila Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Drosophila , Exocrine Glands/metabolism , Male , Prostatic Neoplasms/metabolism , Signal Transduction/physiology
9.
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
10.
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
11.
Int J Mol Sci ; 19(11)2018 Nov 17.
Article in English | MEDLINE | ID: mdl-30453651

ABSTRACT

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.


Subject(s)
Bile Acids and Salts/metabolism , Disease , Health , Receptors, Cytoplasmic and Nuclear/metabolism , Xenobiotics/metabolism , Animals , Bile Acids and Salts/chemistry , Humans , Inactivation, Metabolic
12.
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
13.
Stem Cell Reports ; 11(4): 944-958, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30245210

ABSTRACT

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.


Subject(s)
Benzhydryl Compounds/toxicity , Cell Differentiation , Germ Cells/pathology , Homeostasis , Infertility, Male/metabolism , Infertility, Male/pathology , Phenols/toxicity , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Adult , Animals , Animals, Newborn , Cell Differentiation/drug effects , Fetus/drug effects , Fetus/pathology , Germ Cells/drug effects , Germ Cells/metabolism , Homeostasis/drug effects , Humans , Male , Mice , Middle Aged , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Signal Transduction/drug effects , Stigmasterol/toxicity
14.
Article in English | MEDLINE | ID: mdl-30072948

ABSTRACT

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.

15.
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
16.
Mol Aspects Med ; 56: 101-109, 2017 08.
Article in English | MEDLINE | ID: mdl-28511935

ABSTRACT

Next to their involvement in digestion, bile acids have been defined as signaling molecules. They have been demonstrated to control many physiological functions among which lipid homeostasis, glucose and energy metabolisms. Bile acids are ligands of several receptors and multiple studies using transgenic mouse models defined the major roles of their respective nuclear and membrane receptors namely the Farnesoid-X-Receptor (FXRα) and the G-protein-coupled bile acid receptor 1(GPBAR1; TGR5). Here we review the reports highlighting the impacts of bile acids on testicular physiology and on male reproductive functions. The studies on mouse models open perspectives to better understand the deleterious effects of bile acids on testicular pathophysiologies and fertility disorders. Additional studies are needed to corroborate these correlations in humans.


Subject(s)
Bile Acids and Salts/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, G-Protein-Coupled/genetics , Spermatogenesis/drug effects , Testis/metabolism , Animals , Bile Acids and Salts/chemistry , Bile Acids and Salts/pharmacology , Energy Metabolism/genetics , Fertility/drug effects , Fertility/genetics , Gene Expression Regulation, Developmental , Glucose/metabolism , Homeostasis , Humans , Male , Mice , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Spermatogenesis/genetics , Testis/cytology , Testis/drug effects , Testis/growth & development , Testosterone/biosynthesis
17.
Proc Natl Acad Sci U S A ; 114(15): 3909-3914, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28348226

ABSTRACT

Lysine Specific Demethylase 1 (LSD1) removes mono- and dimethyl groups from lysine 4 of histone H3 (H3K4) or H3K9, resulting in repressive or activating (respectively) transcriptional histone marks. The mechanisms that control the balance between these two antagonist activities are not understood. We here show that LSD1 and the orphan nuclear receptor estrogen-related receptor α (ERRα) display commonly activated genes. Transcriptional activation by LSD1 and ERRα involves H3K9 demethylation at the transcriptional start site (TSS). Strikingly, ERRα is sufficient to induce LSD1 to demethylate H3K9 in vitro. The relevance of this mechanism is highlighted by functional data. LSD1 and ERRα coregulate several target genes involved in cell migration, including the MMP1 matrix metallo-protease, also activated through H3K9 demethylation at the TSS. Depletion of LSD1 or ERRα reduces the cellular capacity to invade the extracellular matrix, a phenomenon that is rescued by MMP1 reexpression. Altogether our results identify a regulatory network involving a direct switch in the biochemical activities of a histone demethylase, leading to increased cell invasion.


Subject(s)
Histone Demethylases/metabolism , Histones/metabolism , Receptors, Estrogen/metabolism , Cell Movement , Gene Expression Regulation , HEK293 Cells , Histone Demethylases/genetics , Humans , Lysine/metabolism , Matrix Metalloproteinase 1/metabolism , Methylation , Promoter Regions, Genetic , Receptors, Estrogen/genetics , Transcription Initiation Site , ERRalpha Estrogen-Related Receptor
18.
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
19.
Mol Aspects Med ; 56: 2-9, 2017 08.
Article in English | MEDLINE | ID: mdl-28153453

ABSTRACT

Primary bile acids are synthetized from cholesterol within the liver and then transformed by the bacteria in the intestine to secondary bile acids. In addition to their involvement in digestion and fat solubilization, bile acids also act as signaling molecules. Several receptors are sensors of bile acids. Among these receptors, this review focuses on the nuclear receptor FXRα and the G-protein-coupled receptor TGR5. This review briefly presents the potential links between bile acids and cancers that are discussed in more details in the other articles of this special issue of Molecular Aspects of Medicine focused on "Bile acids, roles in integrative physiology and pathophysiology".


Subject(s)
Bile Acids and Salts/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, G-Protein-Coupled/genetics , Animals , Biotransformation , Cholesterol/metabolism , Gastrointestinal Microbiome/physiology , Homeostasis/physiology , Humans , Intestinal Mucosa/metabolism , Intestines/microbiology , Liver/cytology , Liver/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Polymorphism, Genetic , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
20.
Methods Mol Biol ; 1443: 13-9, 2016.
Article in English | MEDLINE | ID: mdl-27246331

ABSTRACT

Screening of bona fide ligands for nuclear receptors is a real tour de force as the identified molecules are supposed to be able to activate the targeted proteins in cell culture as well as in vivo. Indeed orphan nuclear receptors are putative pharmacologically targets for various diseases. It is thus necessary to have quick and reproductive systems that help in identifying new ligands, agonist or antagonist, before using them in vivo in animal models to check for secondary effects. Here, we describe the transient transfections (homologous and heterologous) used for the screening of ligands for liver X receptor α (LXRα, NR1H3) in HeLa cells.


Subject(s)
High-Throughput Screening Assays/methods , Ligands , Liver X Receptors/chemistry , Liver X Receptors/metabolism , Transfection/methods , Animals , HeLa Cells , Homeostasis , Humans , Lipid Metabolism , Lipids/chemistry , Liver X Receptors/genetics , Luciferases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...