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










Publication year range
1.
Methods Mol Biol ; 2846: 35-45, 2024.
Article in English | MEDLINE | ID: mdl-39141228

ABSTRACT

Chromatin immunoprecipitation (ChIP) coupled to qPCR or sequencing is a crucial experiment to determine direct transcriptional regulation under the control of specific transcriptional factors or co-regulators at loci-specific or pan-genomic levels.Here we provide a reliable method for processing ChIP from adipocytes or frozen adipose tissue collection, isolation of nuclei, cross-linking of protein-DNA complexes, chromatin shearing, immunoprecipitation, and DNA purification. We also discuss critical steps for optimizing the experiment to perform a successful ChIP in lipid-rich cells/tissues.


Subject(s)
Adipocytes , Adipose Tissue , Chromatin Immunoprecipitation , DNA , Transcription Factors , Adipocytes/metabolism , Adipocytes/cytology , Adipose Tissue/metabolism , Adipose Tissue/cytology , Chromatin Immunoprecipitation/methods , DNA/metabolism , DNA/genetics , Transcription Factors/metabolism , Humans , Animals , Protein Binding , Chromatin/metabolism , Chromatin/genetics
2.
Methods Mol Biol ; 2846: 91-107, 2024.
Article in English | MEDLINE | ID: mdl-39141231

ABSTRACT

ChIP-exo is a powerful tool for achieving enhanced sensitivity and single-base-pair resolution of transcription factor (TF) binding, which utilizes a combination of chromatin immunoprecipitation (ChIP) and lambda exonuclease digestion (exo) followed by high-throughput sequencing. ChIP-nexus (chromatin immunoprecipitation experiments with nucleotide resolution through exonuclease, unique barcode, and single ligation) is an updated and simplified version of the original ChIP-exo method, which has reported an efficient adapter ligation through the DNA circularization step. Building upon an established method, we present a protocol for generating NGS (next-generation sequencing) ready and high-quality ChIP-nexus library for glucocorticoid receptor (GR). This method is specifically optimized for bone marrow-derived macrophage (BMDM) cells. The protocol is initiated by the formation of DNA-protein cross-links in intact cells. This is followed by chromatin shearing, chromatin immunoprecipitation, ligation of sequencing adapters, digestion of adapter-ligated DNA using lambda exonuclease, and purification of single-stranded DNA for circularization and library amplification.


Subject(s)
Chromatin Immunoprecipitation , DNA , High-Throughput Nucleotide Sequencing , Macrophages , Receptors, Glucocorticoid , Animals , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/genetics , Mice , Macrophages/metabolism , DNA/metabolism , DNA/genetics , High-Throughput Nucleotide Sequencing/methods , Chromatin Immunoprecipitation/methods , Protein Binding , Binding Sites
3.
Res Sq ; 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39070610

ABSTRACT

Circadian disruption enhances cancer risk, and many tumors exhibit disordered circadian gene expression. We show rhythmic gene expression is unexpectedly robust in clear cell renal cell carcinoma (ccRCC). Furthermore, the clock gene BMAL1 is higher in ccRCC than in healthy kidneys, unlike in other tumor types. BMAL1 is closely related to ARNT, and we show that BMAL1-HIF2α regulates a subset of HIF2α target genes in ccRCC cells. Depletion of BMAL1 reprograms HIF2α chromatin association and target gene expression and reduces ccRCC growth in culture and in xenografts. Analysis of pre-existing data reveals higher BMAL1 in patient-derived xenografts that are sensitive to growth suppression by a HIF2α antagonist (PT2399). We show that BMAL1-HIF2α is more sensitive than ARNT-HIF2α to suppression by PT2399, and increasing BMAL1 sensitizes 786O cells to growth inhibition by PT2399. Together, these findings indicate that an alternate HIF2α heterodimer containing the circadian partner BMAL1 contributes to HIF2α activity, growth, and sensitivity to HIF2α antagonist drugs in ccRCC cells.

4.
bioRxiv ; 2024 Jun 09.
Article in English | MEDLINE | ID: mdl-38895384

ABSTRACT

Circadian disruption enhances cancer risk, and many tumors exhibit disordered circadian gene expression. We show rhythmic gene expression is unexpectedly robust in clear cell renal cell carcinoma (ccRCC). Furthermore, the clock gene BMAL1 is higher in ccRCC than in healthy kidneys, unlike in other tumor types. BMAL1 is closely related to ARNT, and we show that BMAL1-HIF2α regulates a subset of HIF2α target genes in ccRCC cells. Depletion of BMAL1 reprograms HIF2α chromatin association and target gene expression and reduces ccRCC growth in culture and in xenografts. Analysis of pre-existing data reveals higher BMAL1 in patient-derived xenografts that are sensitive to growth suppression by a HIF2α antagonist (PT2399). We show that BMAL1-HIF2α is more sensitive than ARNT-HIF2α to suppression by PT2399, and increasing BMAL1 sensitizes 786O cells to growth inhibition by PT2399. Together, these findings indicate that an alternate HIF2α heterodimer containing the circadian partner BMAL1 contributes to HIF2α activity, growth, and sensitivity to HIF2α antagonist drugs in ccRCC cells.

5.
FEBS Lett ; 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38348593

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) begins with lipid accumulation and progresses toward inflammation and fibrosis. Nuclear receptors (NRs), like the Peroxisome Proliferator-Activated Receptors alpha and gamma (PPARα and PPARy), the Farnesoid X Receptor (FXR), and the Liver X receptor (LXR), regulate genes by heterodimerizing with Retinoid X receptor (RXR). These receptors are emerging targets for pharmaceutical intervention for metabolic diseases.

6.
Eur J Endocrinol ; 188(5): R111-R130, 2023 May 10.
Article in English | MEDLINE | ID: mdl-37119521

ABSTRACT

Glucocorticoids are essential hormones produced by the adrenal cortex with prominent circadian rhythmicity and in times of stress. Glucocorticoids maintain liver homeostasis through coordinated activities that control the major pathways of energy metabolism. Glucocorticoids activate the glucocorticoid receptor (GR), a nuclear hormone receptor that regulates the transcription of hundreds of genes in response to ligand. This review aims to provide a comprehensive overview of glucocorticoid receptor signaling impact on glucose, amino acid, and lipid metabolism in the liver. We integrate fundamental and current findings elucidating key GR-regulated pathways from a physiologic, biochemical, and molecular point-of-view. Here, we focus on the transcriptional regulation of well-characterized hepatic GR target genes, and on those GR co-factors that coordinate nutritional and hormonal signals.


Subject(s)
Glucocorticoids , Receptors, Glucocorticoid , Humans , Gene Expression Regulation , Genomics , Glucocorticoids/metabolism , Receptors, Glucocorticoid/metabolism , Steroids
7.
Comput Struct Biotechnol J ; 20: 5622-5638, 2022.
Article in English | MEDLINE | ID: mdl-36284713

ABSTRACT

Glucocorticoids such as dexamethasone (Dex) are widely used to treat both acute and chronic inflammatory conditions. They regulate immune responses by dampening cell-mediated immunity in a glucocorticoid receptor (GR)-dependent manner, by suppressing the expression of pro-inflammatory cytokines and chemokines and by stimulating the expression of anti-inflammatory mediators. Despite its evident clinical benefit, the mechanistic underpinnings of the gene regulatory networks transcriptionally controlled by GR in a context-specific manner remain mysterious. Next generation sequencing methods such mRNA sequencing (RNA-seq) and Ribosome profiling (ribo-seq) provide tools to investigate the transcriptional and post-transcriptional mechanisms that govern gene expression. Here, we integrate matched RNA-seq data with ribo-seq data from human acute monocytic leukemia (THP-1) cells treated with the TLR4 ligand lipopolysaccharide (LPS) and with Dex, to investigate the global transcriptional and translational regulation (translational efficiency, ΔTE) of Dex-responsive genes. We find that the expression of most of the Dex-responsive genes are regulated at both the transcriptional and the post-transcriptional level, with the transcriptional changes intensified on the translational level. Overrepresentation pathway analysis combined with STRING protein network analysis and manual functional exploration, identified these genes to encode immune effectors and immunomodulators that contribute to macrophage-mediated immunity and to the maintenance of macrophage-mediated immune homeostasis. Further research into the translational regulatory network underlying the GR anti-inflammatory response could pave the way for the development of novel immunomodulatory therapeutic regimens with fewer undesirable side effects.

8.
Int J Mol Sci ; 22(23)2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34884498

ABSTRACT

Palmitoleic acid (C16:1n7) has been identified as a regulator of physiological cardiac hypertrophy. In the present study, we aimed to investigate the molecular pathways involved in C16:1n7 responses in primary murine cardiomyocytes (PCM) and a mouse model of isoproterenol (ISO)-induced cardiac damage. PCMs were stimulated with C16:1n7 or a vehicle. Afterwards, RNA sequencing was performed using an Illumina HiSeq sequencer. Confirmatory analysis was performed in PCMs and HL-1 cardiomyocytes. For an in vivo study, 129 sv mice were orally treated with a vehicle or C16:1n7 for 22 days. After 5 days of pre-treatment, the mice were injected with ISO (25 mg/kg/d s. c.) for 4 consecutive days. Cardiac phenotyping was performed using echocardiography. In total, 129 genes were differentially expressed in PCMs stimulated with C16:1n7, including Angiopoietin-like factor 4 (Angptl4) and Pyruvate Dehydrogenase Kinase 4 (Pdk4). Both Angptl4 and Pdk4 are proxisome proliferator-activated receptor α/δ (PPARα/δ) target genes. Our in vivo results indicated cardioprotective and anti-fibrotic effects of C16:1n7 application in mice. This was associated with the C16:1n7-dependent regulation of the cardiac PPAR-specific signaling pathways. In conclusion, our experiments demonstrated that C16:1n7 might have protective effects on cardiac fibrosis and inflammation. Our study may help to develop future lipid-based therapies for catecholamine-induced cardiac damage.


Subject(s)
Cardiomegaly/drug therapy , Cardiotonic Agents/pharmacology , Catecholamines/toxicity , Fatty Acids, Monounsaturated/pharmacology , Gene Expression Regulation/drug effects , PPAR alpha/metabolism , PPAR delta/metabolism , Animals , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Cardiomegaly/pathology , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , PPAR alpha/genetics , PPAR delta/genetics
9.
Nat Commun ; 12(1): 6883, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824281

ABSTRACT

In female mammals, the cessation of ovarian functions is associated with significant metabolic alterations, weight gain, and increased susceptibility to a number of pathologies associated with ageing. The molecular mechanisms triggering these systemic events are unknown because most tissues are responsive to lowered circulating sex steroids. As it has been demonstrated that isoform alpha of the estrogen receptor (ERα) may be activated by both estrogens and amino acids, we test the metabolic effects of a diet enriched in specific amino acids in ovariectomized (OVX) mice. This diet is able to block the OVX-induced weight gain and fat deposition in the liver. The use of liver-specific ERα KO mice demonstrates that the hepatic ERα, through the control of liver lipid metabolism, has a key role in the systemic response to OVX. The study suggests that the liver ERα might be a valuable target for dietary treatments for the post-menopause.


Subject(s)
Amino Acids, Essential/pharmacology , Estrogen Receptor alpha/metabolism , Liver/drug effects , Ovariectomy/adverse effects , Amino Acids, Branched-Chain/pharmacology , Amino Acids, Branched-Chain/therapeutic use , Amino Acids, Essential/therapeutic use , Animals , Diet Therapy , Estrogen Receptor alpha/deficiency , Female , Lipid Metabolism/drug effects , Liver/metabolism , Male , Mice , Mice, Knockout , Sex Characteristics , Transcriptome/drug effects , Weight Gain/drug effects
10.
STAR Protoc ; 2(3): 100609, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34189474

ABSTRACT

Quantifying differential genome occupancy by chromatin immunoprecipitation (ChIP) remains challenging due to variation in chromatin fragmentation, immunoprecipitation efficiencies, and intertube variability. In this protocol, we add heterologous spike-ins from Drosophila chromatin as an internal control to the mice chromatin before immunoprecipitation to normalize for technical variation in ChIP-qPCR or ChIP-seq. The choice of spike-in depends on the evolutionary conservation of the protein of interest and the antibody used. For complete details on the use and execution of this protocol, please refer to Greulich et al. (2021).


Subject(s)
Chromatin Immunoprecipitation/methods , Proteins/metabolism , Animals , Binding Sites , Drosophila melanogaster , High-Throughput Nucleotide Sequencing/methods , Mice , Real-Time Polymerase Chain Reaction
11.
Cells ; 11(1)2021 12 23.
Article in English | MEDLINE | ID: mdl-35011590

ABSTRACT

Glucocorticoids are potent anti-inflammatory drugs; however, their molecular mode of action remains complex and elusive. They bind to the glucocorticoid receptor (GR), a nuclear receptor that controls gene expression in almost all tissues in a cell type-specific manner. While GR's transcriptional targets mediate beneficial reactions in immune cells, they also harbor the potential of adverse metabolic effects in other cell types such as hepatocytes. Here, we have profiled nascent transcription upon glucocorticoid stimulation in LPS-activated primary murine macrophages using 4sU-seq. We compared our results to publicly available nascent transcriptomics data from murine liver and bioinformatically identified non-coding RNAs transcribed from intergenic GR binding sites in a tissue-specific fashion. These tissue-specific enhancer RNAs (eRNAs) correlate with target gene expression, reflecting cell type-specific glucocorticoid responses. We further associate GR-mediated eRNA expression with changes in H3K27 acetylation and BRD4 recruitment in inflammatory macrophages upon glucocorticoid treatment. In summary, we propose a common mechanism by which GR-bound enhancers regulate target gene expression by changes in histone acetylation, BRD4 recruitment and eRNA expression. We argue that local eRNAs are potential therapeutic targets downstream of GR signaling which may modulate glucocorticoid response in a cell type-specific way.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation , Glucocorticoids/pharmacology , Macrophages/metabolism , RNA/genetics , Acetylation/drug effects , Animals , Binding Sites , Gene Expression Profiling , Gene Expression Regulation/drug effects , Histones/metabolism , Lysine/metabolism , Macrophages/drug effects , Male , Mice, Inbred C57BL , Nuclear Proteins/metabolism , Organ Specificity/drug effects , RNA/metabolism , Receptors, Glucocorticoid/metabolism , Transcription Factors/metabolism , Transcription, Genetic/drug effects
12.
Nucleic Acids Res ; 48(15): 8393-8407, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32619221

ABSTRACT

The glucocorticoid receptor is an important immunosuppressive drug target and metabolic regulator that acts as a ligand-gated transcription factor. Generally, GR's anti-inflammatory effects are attributed to the silencing of inflammatory genes, while its adverse effects are ascribed to the upregulation of metabolic targets. GR binding directly to DNA is proposed to activate, whereas GR tethering to pro-inflammatory transcription factors is thought to repress transcription. Using mice with a point mutation in GR's zinc finger, that still tether via protein-protein interactions while being unable to recognize DNA, we demonstrate that DNA binding is essential for both transcriptional activation and repression. Performing ChIP-Seq, RNA-Seq and proteomics under inflammatory conditions, we show that DNA recognition is required for the assembly of a functional co-regulator complex to mediate glucocorticoid responses. Our findings may contribute to the development of safer immunomodulators with fewer side effects.


Subject(s)
DNA-Binding Proteins/genetics , DNA/genetics , Inflammation/genetics , Receptors, Glucocorticoid/genetics , Animals , DNA/metabolism , Gene Expression Regulation/genetics , Glucocorticoids/genetics , Glucocorticoids/metabolism , Humans , Inflammation/pathology , Mice , Protein Interaction Domains and Motifs/genetics , RNA-Seq , Transcriptional Activation/genetics
13.
Curr Opin Pharmacol ; 53: 35-44, 2020 08.
Article in English | MEDLINE | ID: mdl-32416533

ABSTRACT

Glucocorticoids (GCs) are widely used immunomodulators. They regulate gene expression by binding and activating the Glucocorticoid Receptor (GR), but underlying transcriptional mechanisms remain enigmatic. This review summarizes recent findings identifyingspecific GR-bound DNA sequences whose configuration may affect transcriptional output. Additional factors affecting GR's anti-inflammatory actions, including different chromatin states such as DNAse hypersensitive regions and histone marks will be discussed, together with the relevant transcriptional co-regulators and promoter/enhancer features. Furthermore, the involvement of non-coding RNAs such as lncRNAs, miRNAs and eRNAs adds another level of regulation to the GR's transcriptional activity. Characterizing and understanding these multiple mechanisms will be crucial for developing more targeted immunomodulatory therapies with reduced adverse effects such as obesity, diabetes and osteoporosis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Glucocorticoids/pharmacology , Animals , Genomics , Humans , RNA, Untranslated , Receptors, Glucocorticoid/genetics , Transcription, Genetic
14.
Mol Cell ; 76(4): 531-545.e5, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31706703

ABSTRACT

The glucocorticoid receptor (GR) is a potent metabolic regulator and a major drug target. While GR is known to play integral roles in circadian biology, its rhythmic genomic actions have never been characterized. Here we mapped GR's chromatin occupancy in mouse livers throughout the day and night cycle. We show how GR partitions metabolic processes by time-dependent target gene regulation and controls circulating glucose and triglycerides differentially during feeding and fasting. Highlighting the dominant role GR plays in synchronizing circadian amplitudes, we find that the majority of oscillating genes are bound by and depend on GR. This rhythmic pattern is altered by high-fat diet in a ligand-independent manner. We find that the remodeling of oscillatory gene expression and postprandial GR binding results from a concomitant increase of STAT5 co-occupancy in obese mice. Altogether, our findings highlight GR's fundamental role in the rhythmic orchestration of hepatic metabolism.


Subject(s)
Chromatin/metabolism , Circadian Clocks , Circadian Rhythm , Diet, High-Fat , Dietary Fats/metabolism , Energy Metabolism , Liver/metabolism , Obesity/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Blood Glucose/metabolism , Circadian Clocks/genetics , Circadian Rhythm/genetics , Dietary Fats/administration & dosage , Dietary Fats/blood , Disease Models, Animal , Energy Metabolism/genetics , Fasting/metabolism , Gene Expression Regulation , Glucocorticoids/metabolism , Gluconeogenesis , Ligands , Male , Mice, Inbred C57BL , Mice, Knockout , Obesity/blood , Obesity/genetics , PPAR alpha/genetics , PPAR alpha/metabolism , Postprandial Period , Receptors, Glucocorticoid/deficiency , Receptors, Glucocorticoid/genetics , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Secretory Pathway , Signal Transduction , Time Factors , Transcription, Genetic , Triglycerides/blood
15.
Front Immunol ; 10: 1859, 2019.
Article in English | MEDLINE | ID: mdl-31440248

ABSTRACT

For many decades, glucocorticoids have been widely used as the gold standard treatment for inflammatory conditions. Unfortunately, their clinical use is limited by severe adverse effects such as insulin resistance, cardiometabolic diseases, muscle and skin atrophies, osteoporosis, and depression. Glucocorticoids exert their effects by binding to the Glucocorticoid Receptor (GR), a ligand-activated transcription factor which both positively, and negatively regulates gene expression. Extensive research during the past several years has uncovered novel mechanisms by which the GR activates and represses its target genes. Genome-wide studies and mouse models have provided valuable insight into the molecular mechanisms of inflammatory gene regulation by GR. This review focusses on newly identified target genes and GR co-regulators that are important for its anti-inflammatory effects in innate immune cells, as well as mutations within the GR itself that shed light on its transcriptional activity. This research progress will hopefully serve as the basis for the development of safer immune suppressants with reduced side effect profiles.


Subject(s)
Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Glucocorticoids/immunology , Immunosuppressive Agents/immunology , Receptors, Glucocorticoid/immunology , Animals , Glucocorticoids/pharmacology , Humans , Immunosuppressive Agents/pharmacology
16.
Methods Mol Biol ; 1966: 39-70, 2019.
Article in English | MEDLINE | ID: mdl-31041738

ABSTRACT

Chromatin immunoprecipitation coupled to next generation sequencing (ChIP-seq) is a powerful tool to map context-dependent genome-wide binding of nuclear hormone receptors and their coregulators. This information can provide important mechanistic insight into where, when and how DNA-protein interactions are linked to target gene regulation. Here we describe a simple, yet reliable ChIP-seq method, including nuclear isolation from frozen tissue samples, cross-linking DNA-protein complexes, chromatin shearing, immunoprecipitation, and purification of ChIP DNA. We also include a standard ChIP-seq data analysis pipeline to elaborate and analyze raw single-end or paired-end sequencing data, including quality control steps, peak calling, annotation, and motif enrichment.


Subject(s)
Chromatin Immunoprecipitation/methods , High-Throughput Nucleotide Sequencing/methods , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , DNA/metabolism , Humans , Sequence Analysis, DNA/methods
17.
Dev Cell ; 47(2): 205-221.e7, 2018 10 22.
Article in English | MEDLINE | ID: mdl-30352176

ABSTRACT

Lipid metabolism is highly compartmentalized between cellular organelles that dynamically adapt their compositions and interactions in response to metabolic challenges. Here, we investigate how diet-induced hepatic lipid accumulation, observed in non-alcoholic fatty liver disease (NAFLD), affects protein localization, organelle organization, and protein phosphorylation in vivo. We develop a mass spectrometric workflow for protein and phosphopeptide correlation profiling to monitor levels and cellular distributions of ∼6,000 liver proteins and ∼16,000 phosphopeptides during development of steatosis. Several organelle contact site proteins are targeted to lipid droplets (LDs) in steatotic liver, tethering organelles orchestrating lipid metabolism. Proteins of the secretory pathway dramatically redistribute, including the mis-localization of the COPI complex and sequestration of the Golgi apparatus at LDs. This correlates with reduced hepatic protein secretion. Our systematic in vivo analysis of subcellular rearrangements and organelle-specific phosphorylation reveals how nutrient overload leads to organellar reorganization and cellular dysfunction.


Subject(s)
Fatty Liver/physiopathology , Lipid Droplets/physiology , Organelles/physiology , Animals , Diet , Diet, High-Fat , Golgi Apparatus/physiology , Lipid Droplets/metabolism , Lipid Metabolism , Lipids/physiology , Liver , Mass Spectrometry/methods , Mice , Mice, Inbred C57BL , Mitochondrial Membranes , Nutrients/metabolism , Organelles/drug effects , Phosphorylation , Protein Transport , Proteomics/methods , Secretory Pathway
18.
Cell Rep ; 15(2): 360-71, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27050513

ABSTRACT

Lipoprotein synthesis is controlled by estrogens, but the exact mechanisms underpinning this regulation and the role of the hepatic estrogen receptor α (ERα) in cholesterol physiology are unclear. Utilizing a mouse model involving selective ablation of ERα in the liver, we demonstrate that hepatic ERα couples lipid metabolism to the reproductive cycle. We show that this receptor regulates the synthesis of cholesterol transport proteins, enzymes for lipoprotein remodeling, and receptors for cholesterol uptake. Additionally, ERα is indispensable during proestrus for the generation of high-density lipoproteins efficient in eliciting cholesterol efflux from macrophages. We propose that a specific interaction with liver X receptor α (LXRα) mediates the broad effects of ERα on the hepatic lipid metabolism.


Subject(s)
Estrogen Receptor alpha/metabolism , Liver/metabolism , Reproduction , Adiposity , Animals , Cholesterol/metabolism , Collagen/metabolism , Estrous Cycle , Female , Gene Deletion , Lipoproteins/metabolism , Lipoproteins, HDL/metabolism , Liver X Receptors/metabolism , Mice, Knockout , PPAR alpha/metabolism , Protein Binding , Transcription, Genetic
19.
Reproduction ; 142(4): 489-95, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21810859

ABSTRACT

Since the discovery of the conserved forkhead (Fkh) DNA binding domain more than 20 years ago, members of the Fkh or forkhead box (FOX) family of transcription factors have been shown to act as important regulators of numerous developmental and homeostatic processes. The human genome contains 44 Fkh genes, several of which have recently been reported to be essential for female fertility. In this review, we highlight the roles of specific FOX proteins in ovarian folliculogenesis and present our current understanding of their molecular function. In particular, we describe what we have learned from loss-of-function studies using mouse models as well as human genetics and illustrate how different stages of folliculogenesis, both in oocytes and in somatic granulosa and theca cells, are regulated by FOXC1, FOXL2, and FOXO subfamily members.


Subject(s)
Forkhead Transcription Factors/physiology , Ovarian Follicle/physiology , Ovary/physiology , Animals , Female , Homeostasis/physiology , Humans , Mice , Models, Animal , Reproduction/physiology
20.
Mol Genet Metab ; 88(3): 225-34, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16647286

ABSTRACT

Foxl2 is a forkhead transcription factor essential for proper reproductive function in females. Human patients carrying mutations in the FOXL2 gene display blepharophimosis/ptosis/epicanthus inversus syndrome (BPES), an autosomal dominant disease associated with eyelid defects and premature ovarian failure in females. Recently, animal models for BPES have been developed that in combination with a catalogue of human FOXL2 mutations provide further insight into its molecular function. Mice homozygous mutant for Foxl2 display craniofacial malformations and female infertility. The analysis of the murine phenotype has revealed that Foxl2 is required for granulosa cell function. These ovarian somatic cells surround and nourish the oocyte and play an important role in follicle formation and activation. Mutations upstream of FOXL2 in humans, not affecting the coding sequence itself, have also been shown to cause BPES, which points to the existence of a distant regulatory element necessary for proper gene expression. The same regulatory sequences may be deleted in the goat polled intersex syndrome (PIS), in which FoxL2 expression is severely reduced. Sequence comparison of FoxL2 from several vertebrate species has shown that it is a highly conserved gene involved in ovary development. Thus, the detailed understanding of Foxl2 function and regulation and the identification of its transcriptional targets may open new avenues for the treatment of female infertility in the future.


Subject(s)
Forkhead Transcription Factors/physiology , Ovary/metabolism , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Amino Acid Sequence , Animals , Blepharophimosis/genetics , Blepharophimosis/metabolism , Blepharoptosis/genetics , Blepharoptosis/metabolism , Evolution, Molecular , Eyelids/abnormalities , Female , Forkhead Box Protein L2 , Forkhead Transcription Factors/genetics , Humans , Infertility, Female/genetics , Infertility, Female/metabolism , Mice , Molecular Sequence Data , Mutation , Ovary/abnormalities , Ovary/embryology , Primary Ovarian Insufficiency/genetics , Primary Ovarian Insufficiency/metabolism , Regulatory Elements, Transcriptional , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL