Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 16(11)2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38893271

ABSTRACT

Epidemiological studies point to cholesterol as a possible key factor for both prostate cancer incidence and progression. It could represent a targetable metabolite as the most aggressive tumors also appear to be sensitive to therapies designed to decrease hypercholesterolemia, such as statins. However, it remains unknown whether and how cholesterol, through its dietary uptake and its metabolism, could be important for early tumorigenesis. Oncogene clonal induction in the Drosophila melanogaster accessory gland allows us to reproduce tumorigenesis from initiation to early progression, where tumor cells undergo basal extrusion to form extra-epithelial tumors. Here we show that these tumors accumulate lipids, and especially esterified cholesterol, as in human late carcinogenesis. Interestingly, a high-cholesterol diet has a limited effect on accessory gland tumorigenesis. On the contrary, cell-specific downregulation of cholesterol uptake, intracellular transport, or metabolic response impairs the formation of such tumors. Furthermore, in this context, a high-cholesterol diet suppresses this impairment. Interestingly, expression data from primary prostate cancer tissues indicate an early signature of redirection from cholesterol de novo synthesis to uptake. Taken together, these results reveal that during early tumorigenesis, tumor cells strongly increase their uptake and use of dietary cholesterol to specifically promote the step of basal extrusion. Hence, these results suggest the mechanism by which a reduction in dietary cholesterol could lower the risk and slow down the progression of prostate cancer.

2.
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
3.
Br J Pharmacol ; 178(16): 3277-3293, 2021 08.
Article in English | MEDLINE | ID: mdl-33080050

ABSTRACT

Liver X receptors (LXRs) are members of the nuclear receptor superfamily that are canonically activated by oxidized derivatives of cholesterol. Since the mid-90s, numerous groups have identified LXRs as endocrine receptors that are involved in the regulation of various physiological functions. As a result, when their expression is genetically modified in mice, phenotypic analyses reveal endocrine disorders ranging from infertility to diabetes and obesity, nervous system pathologies such Alzheimer's or Parkinson's disease, immunological disturbances, inflammatory response, and enhancement of tumour development. Based on such findings, it appears that LXRs could constitute good pharmacological targets to prevent and/or to treat these diseases. This review discusses the various aspects of LXR drug discovery, from the tools available for the screening of potential LXR modulators to the current situational analysis of the drugs in development. LINKED ARTICLES: This article is part of a themed issue on Oxysterols, Lifelong Health and Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.16/issuetoc.


Subject(s)
Diabetes Mellitus , Parkinson Disease , Animals , Cholesterol , Liver X Receptors , Mice , Receptors, Cytoplasmic and Nuclear
4.
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
5.
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
6.
J Steroid Biochem Mol Biol ; 190: 173-182, 2019 06.
Article in English | MEDLINE | ID: mdl-30959154

ABSTRACT

Liver X receptors (LXRs) α (NR1H3) and ß (NR1H2) are nuclear receptors that have been involved in the regulation of many physiological processes, principally in the control of cholesterol homeostasis, as well as in the control of the cell death and proliferation balance. These receptors are thus promising therapeutic targets in various pathologies such as dyslipidemia, atherosclerosis, diabetes and/or cancers. These receptors are known to be activated by specific oxysterol compounds. The screening for LXR-specific ligands is a challenging process: indeed, these molecules should present a specificity towards each LXR-isoform. Because some natural products have significant effects in the regulation of the LXR-regulated homeostasis and are enriched in flavonoids, we have decided to test in cell culture the effects of 4 selected flavonoids (galangin, quercetin, apigenin and naringenin) on the modulation of LXR activity using double-hybrid experiments. In silico, molecular docking suggests specific binding pattern between agonistic and antagonistic molecules. Altogether, these results allow a better understanding of the ligand binding pocket of LXRα/ß. They also improve our knowledge about flavonoid mechanism of action, allowing the selection and development of better LXR selective ligands.


Subject(s)
Flavonoids/pharmacology , Liver X Receptors/agonists , Liver X Receptors/antagonists & inhibitors , Apigenin/pharmacology , Drug Evaluation, Preclinical , Flavanones/pharmacology , HeLa Cells , Humans , Liver X Receptors/metabolism , Molecular Docking Simulation , Quercetin/pharmacology , Structure-Activity Relationship
7.
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
8.
J Steroid Biochem Mol Biol ; 189: 108-115, 2019 05.
Article in English | MEDLINE | ID: mdl-30831197

ABSTRACT

Antiandrogens have a peculiar place in the treatment of metastatic prostate cancer by blocking the androgen receptor (AR). Unfortunately, aggressive tumors could rapidly develop into a castration resistant state. It is therefore essential to look for new molecules that are more effective, affecting not only the androgen signaling and with minimum undesirable effects. Natural products are an interesting source of new therapeutics, especially for cancer therapy as 70% of them have botanical origin. Based on an ethnobotany screening, we evaluated the effects of ethanolic extract of propolis (EEP) from Algeria on LNCaP cells. Results pointed out that EEP reduces the survival of LNCaP cells with an IC50 of 0.04 mg/ml, induces the apoptosis and blocks the cell cycle at G0/G1 phase. Interestingly, EEP decreased the accumulation of AR suggesting some anti-androgen activity. Indeed, secreted amount of the androgen target protein PSA was decreased when LNCaP cells were incubated with EEP, starting after 4 h of treatment. This anti-androgen activity was also shown on the androgen target genes Fkbp5 and Sgk1. Finally, the capacity of EEP to block AR functioning was demonstrated in transient transfections with human AR and the reporter gene ARE-tk-Luc. Propolis antagonizes the induction of the luciferase activity induced by the natural androgen DHT (10-8M) or the synthetic AR agonist R1881 (10-7M). Altogether, these results highlight the potential pharmacological effects of EEP in future treatments of prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation/drug effects , Propolis/pharmacology , Prostatic Neoplasms/drug therapy , Receptors, Androgen/genetics , Androgen Antagonists/pharmacology , Animals , Apoptosis/drug effects , Bees , Cell Cycle/drug effects , Cell Line, Tumor , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Transcriptional Activation/drug effects
9.
Inflammopharmacology ; 27(1): 15-25, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30467620

ABSTRACT

Stress is a reflex response, both psychological and physiological, of the body to a difficult situation that requires adaptation. Stress is at the intersection of the objective event and the subjective event. The physiological mechanisms involved in chronic stress are numerous and can contribute to a wide variety of disorders, in all systems including the immune system. Stress modifies the Th1/Th2 balance via the HPA axis and a set of immune mediators. This will make the body more vulnerable to external infections in a scientific way while others claim the opposite, stress could be considered immune stimulatory. The development of synthetic LXR ligands such as T0901317 and GW3965 as well as an understanding of the direct involvement of these receptors in the regulation of proopiomelanocortin (POMC) gene expression and indirectly by producing a variety of cytokines in a stressor response, will open in the near future new therapeutic methods against the undesirable effects of stress on the behavior of the immune system.


Subject(s)
Immunologic Factors/immunology , Liver X Receptors/immunology , Stress, Physiological/immunology , Stress, Psychological/immunology , Animals , Cytokines/immunology , Humans
10.
Int J Mol Sci ; 19(9)2018 Aug 28.
Article in English | MEDLINE | ID: mdl-30154328

ABSTRACT

Prostate cancer (PCa) incidence has been dramatically increasing these last years in westernized countries. Though localized PCa is usually treated by radical prostatectomy, androgen deprivation therapy is preferred in locally advanced disease in combination with chemotherapy. Unfortunately, PCa goes into a castration-resistant state in the vast majority of the cases, leading to questions about the molecular mechanisms involving the steroids and their respective nuclear receptors in this relapse. Interestingly, liver X receptors (LXRα/NR1H3 and LXRß/NR1H2) have emerged as new actors in prostate physiology, beyond their historical roles of cholesterol sensors. More importantly LXRs have been proposed to be good pharmacological targets in PCa. This rational has been based on numerous experiments performed in PCa cell lines and genetic animal models pointing out that using selective liver X receptor modulators (SLiMs) could actually be a good complementary therapy in patients with a castration resistant PCa. Hence, this review is focused on the interaction among the androgen receptors (AR/NR3C4), estrogen receptors (ERα/NR3A1 and ERß/NR3A2), and LXRs in prostate homeostasis and their putative pharmacological modulations in parallel to the patients' support.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Prostatic Neoplasms/etiology , Prostatic Neoplasms/metabolism , Androgens/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Disease Management , Endocrine Disruptors/adverse effects , Environmental Exposure/adverse effects , Estrogens/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lipid Metabolism , Liver X Receptors/genetics , Liver X Receptors/metabolism , Male , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Oxysterols/metabolism , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/therapy , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction
11.
Chem Phys Lipids ; 207(Pt B): 214-222, 2017 10.
Article in English | MEDLINE | ID: mdl-28411017

ABSTRACT

Natural products are an interesting source of new therapeutics, especially for cancer therapy as 70% of them have botany origin. Propolis, a resinous mixture that honey bees collect and transform from tree buds, sap flows, or other botanical sources, has been used by ethnobotany and traditional practitioners as early in Egypt as 3000 BCE. Enriched in flavonoids, phenol acids and terpene derivatives, propolis has been widely used for its antibacterial, antifungal and anti-inflammatory properties. Even though it is a challenge to standardize propolis composition, chemical analyses have pointed out interesting molecules that also present anti-oxidant and anti-proliferative properties that are of interest in the field of anti-cancer therapy. This review describes the various geographical origins and compositions of propolis, and analyzes how the main compounds of propolis could modulate cell signaling. A focus is made on the putative use of propolis in prostate cancer.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Antifungal Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Neoplasms/drug therapy , Plant Extracts/pharmacology , Propolis/pharmacology , Animals , Anti-Bacterial Agents/chemistry , Anti-Inflammatory Agents/chemistry , Antifungal Agents/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Cell Proliferation/drug effects , Humans , Neoplasms/pathology , Plant Extracts/chemistry , Propolis/chemistry
12.
Chem Phys Lipids ; 207(Pt B): 59-68, 2017 10.
Article in English | MEDLINE | ID: mdl-28259649

ABSTRACT

Liver X receptors (LXRs) are members of the nuclear receptor superfamily that have been shown to regulate various physiological functions such as lipid metabolism and cholesterol homeostasis. Concordant reports have elicited the possibility to target them to cure many human diseases including arteriosclerosis, cancer, arthritis, and diabetes. The high relevance of modulating LXR activities to treat numerous skin diseases, mainly those with exacerbated inflammation processes, contrasts with the lack of approved therapeutic use. This review makes an assessment to sum up the findings regarding the physiological roles of LXRs in skin and help progress towards the therapeutic and safe management of their activities. It focuses on the possible pharmacological targeting of LXRs to cure or prevent selected skin diseases.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Atherosclerosis/drug therapy , Diabetes Mellitus/drug therapy , Liver X Receptors/metabolism , Neoplasms/drug therapy , Skin/drug effects , Arthritis, Rheumatoid/metabolism , Atherosclerosis/metabolism , Diabetes Mellitus/metabolism , Humans , Ligands , Liver X Receptors/chemistry , Neoplasms/metabolism , Skin/metabolism , Skin Physiological Phenomena/drug effects
13.
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
14.
Mol Aspects Med ; 49: 31-46, 2016 06.
Article in English | MEDLINE | ID: mdl-27091047

ABSTRACT

Liver X receptors (LXRs) α and ß are nuclear receptors whose transcriptional activity is regulated by oxysterols, the oxidized forms of cholesterol. Described in the late 1990s as lipid sensors, both LXRs regulate cholesterol and fatty acid homeostasis. Over the years, deep phenotypic analyses of mouse models deficient for LXRα and/or LXRß have pointed out various other physiological functions including glucose homeostasis, immunology, and neuroprotection. This review enlightens the "endocrine" functions of LXRs; they deeply impact plasma glucose directly and by modulating insulin signaling, renin-angiotensin-aldosterone axis, thyroid and pituitary hormone levels, and bone homeostasis. Besides, LXR signaling is also involved in adrenal physiology, steroid synthesis, and male and female reproduction. Hence, LXRs are definitely involved in the endocrine system and could thus be considered as endocrine receptors, even though oxysterols do not fully correspond to the definition of hormones. Finally, because they are ligand-regulated transcription factors, LXRs are potential pharmacological targets with promising beneficial metabolic effects.


Subject(s)
Endocrine System/physiology , Liver X Receptors/metabolism , Animals , Cholesterol/metabolism , Gene Expression Regulation , Glucose/metabolism , Hormones/metabolism , Humans , Insulin/metabolism , Lipid Metabolism , Liver X Receptors/chemistry , Liver X Receptors/genetics , Organ Specificity , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Reproduction , Transcription Factors/chemistry , Transcription Factors/metabolism
15.
Biol Reprod ; 94(3): 55, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26792941

ABSTRACT

After its production in the testis, a spermatozoon has to undergo posttesticular maturation steps to become fully motile and fertile. The first step is epididymal maturation, during which immature spermatozoa are transformed into biochemically mature cells ready to proceed to the next step, capacitation, a physiological process occurring in the female genital tract. The biochemical transformations include modification of sperm lipid composition during epididymal transit, with significant changes in fatty acids, phospholipids, and sterols between the caput and the cauda epididymal spermatozoa. Although quantitative aspects of these changes are well documented for several mammalian species, molecular mechanisms governing these steps are poorly understood. Transgenic male mice invalidated for the two liver X receptors (LXRalpha and LXRbeta, nuclear oxysterol receptors regulating cholesterol and lipid metabolism) become sterile when aging, showing an epididymal phenotype. We used single-knockout-model mice to characterize the role of each LXR isoform during sperm maturation in the epididymis. We show here that although a certain redundancy exists in the functions of the two LXR isoforms, some physiological processes are more under the influence of only one of them. In both cases, aging males showed slight subfertility, associated with dyslipidemia, emphasizing the importance of lipid metabolism in relation with male fertility.


Subject(s)
Epididymis/metabolism , Gene Expression Regulation/physiology , Liver X Receptors/metabolism , Aging , Animals , Cholesterol/metabolism , Epididymis/pathology , Female , Homeostasis , Infertility, Male/genetics , Infertility, Male/metabolism , Lipid Metabolism , Liver X Receptors/genetics , Male , Mice , Mice, Knockout , Pregnancy , Pregnancy Rate , Protein Isoforms
16.
Endocrinology ; 156(12): 4545-57, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26402841

ABSTRACT

Liver X receptors (LXRs) are key regulators of lipid homeostasis and are involved in multiple testicular functions. The Lxrα(-/-);Lxrß(-/-) mice have illuminated the roles of both isoforms in maintenance of the epithelium in the seminiferous tubules, spermatogenesis, and T production. The requirement for LXRß in Sertoli cells have been emphasized by early abnormal cholesteryl ester accumulation in the Lxrß(-/-) and Lxrα(-/-);Lxrß(-/-) mice. Other phenotypes, such as germ cell loss and hypogonadism, occur later in life in the Lxrα(-/-);Lxrß(-/-) mice. Thus, LXRß expression in Sertoli cells seems to be essential for normal testicular physiology. To decipher the roles of LXRß within the Sertoli cells, we generated Lxrα(-/-);Lxrß(-/-):AMH-Lxrß transgenic mice, which reexpress Lxrß in Sertoli cells in the context of Lxrα(-/-);Lxrß(-/-) mice. In addition to lipid homeostasis, LXRß is necessary for maintaining the blood-testis barrier and the integrity of the germ cell epithelium. LXRß is also implicated in the paracrine action of Sertoli cells on Leydig cells to modulate T synthesis. The Lxrα(-/-);Lxrß(-/-) and Lxrα(-/-);Lxrß(-/-):AMH-Lxrß mice exhibit lipid accumulation in germ cells after the Abcg8 down-regulation, suggesting an intricate LXRß-dependent cooperation between the Sertoli cells and germ cells to ensure spermiogenesis. Further analysis revealed also peritubular smooth muscle defects (abnormal lipid accumulation and disorganized smooth muscle actin) and spermatozoa stagnation in the seminiferous tubules. Together the present work elucidates specific roles of LXRß in Sertoli cell physiology in vivo beyond lipid homeostasis.


Subject(s)
Blood-Testis Barrier/metabolism , Lipid Metabolism/genetics , Orphan Nuclear Receptors/genetics , Seminiferous Epithelium/metabolism , Sertoli Cells/metabolism , Spermatozoa/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 8 , ATP-Binding Cassette Transporters/genetics , Animals , Anti-Mullerian Hormone/genetics , Down-Regulation , Gene Expression , Homeostasis , Leydig Cells/metabolism , Lipoproteins/genetics , Liver X Receptors , Male , Mice , Mice, Knockout , Mice, Transgenic , Muscle, Smooth/metabolism , Paracrine Communication/genetics , Seminiferous Tubules/metabolism , Testis/metabolism , Testosterone/biosynthesis
17.
Proc Natl Acad Sci U S A ; 112(24): 7587-92, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26023184

ABSTRACT

The identification of new pathways governing myelination provides innovative avenues for remyelination. Liver X receptors (LXRs) α and ß are nuclear receptors activated by oxysterols that originated from the oxidation of cholesterol. They are crucial for cholesterol homeostasis, a major lipid constituent of myelin sheaths that are formed by oligodendrocytes. However, the role of LXRs in myelin generation and maintenance is poorly understood. Here, we show that LXRs are involved in myelination and remyelination processes. LXRs and their ligands are present in oligodendrocytes. We found that mice invalidated for LXRs exhibit altered motor coordination and spatial learning, thinner myelin sheaths, and reduced myelin gene expression. Conversely, activation of LXRs by either 25-hydroxycholesterol or synthetic TO901317 stimulates myelin gene expression at the promoter, mRNA, and protein levels, directly implicating LXRα/ß in the transcriptional control of myelin gene expression. Interestingly, activation of LXRs also promotes oligodendroglial cell maturation and remyelination after lysolecithin-induced demyelination of organotypic cerebellar slice cultures. Together, our findings represent a conceptual advance in the transcriptional control of myelin gene expression and strongly support a new role of LXRs as positive modulators in central (re)myelination processes.


Subject(s)
Cerebellum/physiology , Myelin Sheath/physiology , Orphan Nuclear Receptors/physiology , Animals , Cell Differentiation/drug effects , Cerebellum/cytology , Cerebellum/drug effects , Cholesterol/metabolism , Gene Expression Regulation/drug effects , Homeostasis , Hydrocarbons, Fluorinated/pharmacology , Hydroxycholesterols/pharmacology , Liver X Receptors , Male , Mice , Mice, Knockout , Myelin Sheath/drug effects , Myelin Sheath/genetics , Oligodendroglia/cytology , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Organ Culture Techniques , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/deficiency , Promoter Regions, Genetic , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spatial Learning/drug effects , Spatial Learning/physiology , Sulfonamides/pharmacology
18.
Arterioscler Thromb Vasc Biol ; 35(6): 1357-65, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25838428

ABSTRACT

OBJECTIVE: Liver X receptors (LXRs) modulate cholesterol and fatty acid homeostasis as well as inflammation. This study aims to decipher the role of LXRs in the regulation of polyunsaturated fatty acid (PUFA) synthesis in macrophages in the context of atherosclerosis. APPROACH AND RESULTS: Transcriptomic analysis in human monocytes and macrophages was used to identify putative LXR target genes among enzymes involved in PUFA biosynthesis. In parallel, the consequences of LXR activation or LXR invalidation on PUFA synthesis and distribution were determined. Finally, we investigated the impact of LXR activation on PUFA metabolism in vivo in apolipoprotein E-deficient mice. mRNA levels of acyl-CoA synthase long-chain family member 3, fatty acid desaturases 1 and 2, and fatty acid elongase 5 were significantly increased in human macrophages after LXR agonist treatment, involving both direct and sterol responsive element binding protein-1-dependent mechanisms. Subsequently, pharmacological LXR agonist increased long chain PUFA synthesis and enhanced arachidonic acid content in the phospholipids of human macrophages. Increased fatty acid desaturases 1 and 2 and acyl-CoA synthase long-chain family member 3 mRNA levels as well as increased arachidonic acid to linoleic acid and docosahexaenoic acid to eicosapentaenoic acid ratios were also found in atheroma plaque and peritoneal foam cells from LXR agonist-treated mice. By contrast, murine LXR-deficient macrophages displayed reduced expression of fatty acid elongase 5, acyl-CoA synthase long-chain family member 3 and fatty acid desaturases 1, as well as decreased cellular levels of docosahexaenoic acid and arachidonic acid. CONCLUSIONS: Our results indicate that LXR activation triggers PUFA synthesis in macrophages, which results in significant alterations in the macrophage lipid composition. Moreover, we demonstrate here that LXR agonist treatment modulates PUFA metabolism in atherosclerotic arteries.


Subject(s)
Atherosclerosis/metabolism , Fatty Acids, Unsaturated/biosynthesis , Macrophages/metabolism , Orphan Nuclear Receptors/metabolism , Animals , Arachidonic Acid/metabolism , Arteries/metabolism , Foam Cells/metabolism , Humans , Liver X Receptors , Mice , Orphan Nuclear Receptors/agonists , Sterol Regulatory Element Binding Protein 1/metabolism
19.
Steroids ; 99(Pt B): 266-71, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25708389

ABSTRACT

Enolase is a glycolytic enzyme known to inhibit cholesteryl ester hydrolases (CEHs). Cholesteryl ester loading of macrophages, as occurs during atherosclerosis, is accompanied by increased Enolase protein and activity. Here, we describe that J774 macrophages treated with LXR agonists exhibit reduced Enolase transcript and protein abundance. Moreover, we show that this reduction is further potentiated by activation of the LXR/RXR heterodimer with the RXR ligand 9-cis retinoic acid. Enolase levels are also reduced in vivo following activation of LXRs in the intestine, but not in the liver. This effect is lost in Lxrαß-/- mice. In aggregate, our study identified Enolase as a new target of LXRs in vivo, which may promote cholesterol mobilization for subsequent efflux.


Subject(s)
Orphan Nuclear Receptors/metabolism , Phosphopyruvate Hydratase/metabolism , Animals , Intestines/enzymology , Liver X Receptors , Macrophages/enzymology , Mice , Microvilli/enzymology , Models, Biological , Orphan Nuclear Receptors/agonists , Phosphopyruvate Hydratase/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL