Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
iScience ; 27(1): 108692, 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38226167

ABSTRACT

Adipocyte hypertrophy is associated with metabolic complications independent of obesity. We aimed to determine: 1) the association between adipocyte size and postprandial fatty acid metabolism; 2) the potential mechanisms driving the obesity-independent, hypertrophy-associated dysmetabolism in vivo and at a single-cell resolution. Tracers with positron emission tomography were used to measure fatty acid metabolism in 40 men and women with normal or impaired glucose tolerance (NCT02808182), and single nuclei RNA-sequencing (snRNA-seq) to determine transcriptional dynamics of subcutaneous adipose tissue (AT) between individuals with AT hypertrophy vs. hyperplasia matched for sex, ethnicity, glucose-tolerance status, BMI, total and percent body fat, and waist circumference. Adipocyte size was associated with high postprandial total cardiac fatty acid uptake and higher visceral AT dietary fatty acid uptake, but lower lean tissue dietary fatty acid uptake. We found major shifts in cell transcriptomal dynamics with AT hypertrophy that were consistent with in vivo metabolic changes.

2.
Animals (Basel) ; 13(18)2023 Sep 08.
Article in English | MEDLINE | ID: mdl-37760251

ABSTRACT

Cystic ovarian disease (COD) in dairy cattle is characterized by preovulatory follicles that become cysts, fail to ovulate and persist in the ovary; consequently, interfering with normal ovarian cyclicity. The intraovarian key players that orchestrate the alterations occurring in the preovulatory follicle and that culminate with cyst formation and persistence, however, remain uncertain. Interestingly, the Hippo pathway effector yes-associated protein (YAP) has been described in humans and mice as a key player of anovulatory cystic disorders. To start elucidating if YAP deregulation in ovarian follicle cells can be also involved in the pathogenesis of COD, we have generated a series of novel results using spontaneously occurring cystic follicles in cattle. We found that mRNA and protein levels of YAP are significantly higher in granulosa (GCs) and theca cells (TCs) isolated from cystic follicles (follicular structures of at least 20 mm in diameter) in comparison to respective cell types isolated from non-cystic large follicles (≥12 mm). In addition, immunohistochemistry and Western blot analyses used to determine YAP phosphorylation pattern suggest that YAP transcriptional activity is augmented is cystic GCs. These results were confirmed by a significant increase in the mRNA levels encoding for the classic YAP-TEAD transcriptional target genes CTGF, BIRC5 and ANKRD1 in GCs from follicle cysts in comparison to non-cystic large follicles. Taken together, these results provide considerable insight of a completely novel signaling pathway that seems to play an important role in ovarian cystic disease pathogenesis in dairy cattle.

3.
Proc Natl Acad Sci U S A ; 120(32): e2220849120, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37494420

ABSTRACT

The ovarian follicle reserve, formed pre- or perinatally, comprises all oocytes for lifetime reproduction. Depletion of this reserve results in infertility. Steroidogenic factor 1 (SF-1; Nr5a1) and liver receptor homolog 1 (LRH-1; Nr5a2) are two orphan nuclear receptors that regulate adult endocrine function, but their role in follicle formation is unknown. We developed models of conditional depletion of SF-1 or LRH-1 from prenatal ovaries. Depletion of SF-1, but not LRH-1, resulted in dramatically smaller ovaries and fewer primordial follicles. This was mediated by increased oocyte death, resulting from increased ovarian inflammation and increased Notch signaling. Major dysregulated genes were Iroquois homeobox 3 and 5 and their downstream targets involved in the establishment of the ovarian laminin matrix and oocyte-granulosa cell gap junctions. Disruptions of these pathways resulted in follicles with impaired basement membrane formation and compromised oocyte-granulosa communication networks, believed to render them more prone to atresia. This study identifies SF-1 as a key regulator of the formation of the ovarian reserve.


Subject(s)
Ovarian Reserve , Pregnancy , Female , Humans , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Ovarian Reserve/genetics , Ovarian Follicle/metabolism , Ovary/metabolism , Granulosa Cells/metabolism
4.
Cell Commun Signal ; 20(1): 72, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35619099

ABSTRACT

BACKGROUND: The LH surge is a pivotal event that triggers multiple key ovarian processes including oocyte maturation, cumulus expansion, follicular wall rupture and luteinization of mural granulosa and theca cells. Recently, LH-dependent activation of the Hippo signaling pathway has been shown to be required for the differentiation of granulosa cells into luteal cells. Still, the precise interactions between Hippo and LH signaling in murine granulosa cells remain to be elucidated. METHODS: To detect the expression of effectors of the Hippo pathway, western blot, immunohistochemical and RT-qPCR analyses were performed on granulosa cells treated with LH in vitro or isolated from immature mice treated with eCG and hCG. Cultured granulosa cells were pretreated with pharmacologic inhibitors to identify the signaling pathways involved in Hippo regulation by LH. To study the roles of Yap1 and Taz in the regulation of the LH signaling cascade, RT-qPCR and microarray analyses were done on granulosa cells from Yap1f/f;Tazf/f mice treated with an adenovirus to drive cre expression. RT-qPCR was performed to evaluate YAP1 binding to the Areg promoter following chromatin immunoprecipitation of granulosa cells collected from mice prior to or 60 min following hCG treatment. RESULTS: Granulosa cells showed a transient increase in LATS1, YAP1 and TAZ phosphorylation levels in response to the ovulatory signal. This Hippo activation by LH was mediated by protein kinase A. Furthermore, Yap1 and Taz are required for the induction of several LH target genes such as Areg, Pgr and Ptgs2, and for the activation of the ERK1/2 pathway. Consistent with these results, there was a substantial overlap between genes that are upregulated by LH and those that are downregulated following loss of Yap1/Taz, highlighting a major role for Hippo in mediating LH actions in the ovulation process. Finally, we showed that there is a marked recruitment of YAP1 to the Areg promoter of granulosa cells in response to hCG stimulation. CONCLUSIONS: Overall, these results indicate that Hippo collaborates with the cAMP/PKA and ERK1/2 pathways to participate in the precise regulation of the LH cascade, and that Areg, as a direct transcriptional target of YAP1, is involved in mediating its actions in the ovary. Video Abstract.


Subject(s)
Granulosa Cells , Luteinizing Hormone , Amphiregulin/metabolism , Animals , Female , Granulosa Cells/metabolism , Luteinizing Hormone/metabolism , Luteinizing Hormone/pharmacology , Mice , Phosphorylation , Signal Transduction
5.
Endocr Rev ; 43(1): 35-60, 2022 01 12.
Article in English | MEDLINE | ID: mdl-34100954

ABSTRACT

The obesity pandemic increasingly causes morbidity and mortality from type 2 diabetes, cardiovascular diseases and many other chronic diseases. Fat cell size (FCS) predicts numerous obesity-related complications such as lipid dysmetabolism, ectopic fat accumulation, insulin resistance, and cardiovascular disorders. Nevertheless, the scarcity of systematic literature reviews on this subject is compounded by the use of different methods by which FCS measurements are determined and reported. In this paper, we provide a systematic review of the current literature on the relationship between adipocyte hypertrophy and obesity-related glucose and lipid dysmetabolism, ectopic fat accumulation, and cardiovascular disorders. We also review the numerous mechanistic origins of adipocyte hypertrophy and its relationship with metabolic dysregulation, including changes in adipogenesis, cell senescence, collagen deposition, systemic inflammation, adipokine secretion, and energy balance. To quantify the effect of different FCS measurement methods, we performed statistical analyses across published data while controlling for body mass index, age, and sex.


Subject(s)
Cardiovascular Diseases , Diabetes Mellitus, Type 2 , Insulin Resistance , Adipocytes , Cardiovascular Diseases/etiology , Cell Size , Diabetes Mellitus, Type 2/complications , Humans , Hypertrophy/complications , Lipids , Obesity/complications
6.
Mol Reprod Dev ; 86(12): 1901-1908, 2019 12.
Article in English | MEDLINE | ID: mdl-31713287

ABSTRACT

Follicle growth and granulosa cell health are dependent on the secretion of estradiol from granulosa cells. Estradiol is synthesized from androgen precursor by cytochrome P450 aromatase (CYP19A1), and in cattle CYP19A1 messenger RNA has a short half-life but a long (3.5 kb) 3'-untranslated region (3'UTR), suggesting that posttranscriptional regulation may be important for control of enzyme activity. We tested this hypothesis by inserting the CYP19A1 3'UTR and fragments thereof into a reporter vector between the end of the luciferase coding region and the polyadenylation signal. The full-length aromatase 3'UTR suppressed luciferase activity to 10% of control levels, and smaller fragments showed that this inhibitory activity lies between +926 and +1134 of the 3'UTR. Protein-RNA cross-linking experiments revealed that these 3'UTR fragments formed an RNA-protein complex of approximately 70 kDa that was present in granulosa cells but not in corpus luteum, lung, liver, kidney, pancreas, or bladder extracts. The RNA-binding activity was specific to the 3'UTR, as shown by competition experiments with unlabeled RNA, and was present only in 3'UTR constructs that inhibited luciferase activity. These data suggest that posttranscriptional regulation is an important component of the control of CYP19A1 expression and involves protein binding to a specific sequence in the 3'UTR.


Subject(s)
3' Untranslated Regions , Aromatase/biosynthesis , Granulosa Cells/metabolism , Multiprotein Complexes/metabolism , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/metabolism , Animals , Cattle , Female , Granulosa Cells/cytology
7.
Development ; 146(20)2019 10 18.
Article in English | MEDLINE | ID: mdl-31575647

ABSTRACT

WNT signaling plays essential roles in the development and function of the female reproductive tract. Although crosstalk with the Hippo pathway is a key regulator of WNT signaling, whether Hippo itself plays a role in female reproductive biology remains largely unknown. Here, we show that conditional deletion of the key Hippo kinases Lats1 and Lats2 in mouse Müllerian duct mesenchyme cells caused them to adopt the myofibroblast cell fate, resulting in profound reproductive tract developmental defects and sterility. Myofibroblast differentiation was attributed to increased YAP and TAZ expression (but not to altered WNT signaling), leading to the direct transcriptional upregulation of Ctgf and the activation of the myofibroblast genetic program. Müllerian duct mesenchyme cells also became myofibroblasts in male mutant embryos, which impeded the development of the male reproductive tract and resulted in cryptorchidism. The inactivation of Lats1/2 in differentiated uterine stromal cells in vitro did not compromise their ability to decidualize, suggesting that Hippo is dispensable during implantation. We conclude that Hippo signaling is required to suppress the myofibroblast genetic program and maintain multipotency in Müllerian mesenchyme cells.


Subject(s)
Mullerian Ducts/cytology , Mullerian Ducts/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Chromatin Immunoprecipitation , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Endometrium/cytology , Endometrium/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myofibroblasts/cytology , Myofibroblasts/metabolism , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/genetics
8.
Cell Rep ; 28(9): 2443-2454.e4, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31461657

ABSTRACT

In the ovary, follicular growth and maturation are complicated processes that involve a series of morphological and physiological changes in oocytes and somatic cells leading to ovulation and luteinization, essential processes for fertility. Given the complexity of ovulation, characterization of genome-wide regulatory elements is essential to understand the mechanisms governing the expression of specific genes in the rapidly differentiating follicle. We therefore employed a systems biology approach to determine global transcriptional mechanisms during the early stages of the ovulatory process. We demonstrate that, following the hormonal signal that initiates ovulation, granulosa cells undergo major modification of distal regulatory elements, which coincides with cistrome reprogramming of the indispensable orphan nuclear receptor liver receptor homolog-1 (LRH-1). This cistromic reorganization correlates with the extensive changes in gene expression in granulosa cells leading to ovulation. Together, our study yields a highly detailed transcriptional map delineating ovarian cell differentiation during the initiation of ovulation.


Subject(s)
Chromatin Assembly and Disassembly , Ovarian Follicle/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cell Differentiation , Cells, Cultured , Female , Granulosa Cells/cytology , Granulosa Cells/metabolism , Mice , Mice, Inbred C57BL , Nucleotide Motifs , Ovarian Follicle/cytology , Ovulation
9.
FASEB J ; 33(10): 10819-10832, 2019 10.
Article in English | MEDLINE | ID: mdl-31268774

ABSTRACT

Recent reports suggest that the Hippo signaling pathway influences ovarian follicle development; however, its exact roles remain unknown. Here, we examined the ovarian functions of the Hippo kinases large tumor suppressors (LATS)1 and 2, which serve to inactivate the transcriptional coactivators Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ). Inactivation of Lats1/2 in murine granulosa cells either in vitro or in vivo resulted in a loss of granulosa cell morphology, function, and gene expression. Mutant cells further underwent changes in structure and gene expression suggestive of epithelial-to-mesenchymal transition and transdifferentiation into multiple lineages. In vivo, granulosa cell-specific loss of Lats1/2 caused the ovarian parenchyma to be mostly replaced by bone tissue and seminiferous tubule-like structures. Transdifferentiation into Sertoli-like cells and osteoblasts was attributed in part to the increased recruitment of YAP and TAZ to the promoters of sex-determining region Y box 9 and bone γ-carboxyglutamate protein, key mediators of male sex determination and osteogenesis, respectively. Together, these results demonstrate for the first time a critical role for Lats1/2 in the maintenance of the granulosa cell genetic program and further highlight the remarkable plasticity of granulosa cells.-Tsoi, M., Morin, M., Rico, C., Johnson, R. L., Paquet, M., Gévry, N., Boerboom, D. Lats1 and Lats2 are required for ovarian granulosa cell fate maintenance.


Subject(s)
Granulosa Cells/cytology , Granulosa Cells/metabolism , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Proteins/physiology , Acyltransferases , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Lineage , Cell Transdifferentiation , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation , Hippo Signaling Pathway , Infertility, Female/genetics , Infertility, Female/pathology , Infertility, Female/physiopathology , Male , Mice , Mice, Knockout , Osteoblasts/metabolism , Osteoblasts/pathology , Ovarian Follicle/physiology , Ovary/pathology , Ovary/physiopathology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Sertoli Cells/metabolism , Sertoli Cells/pathology , Signal Transduction , Transcription Factors/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , YAP-Signaling Proteins
10.
Front Immunol ; 10: 2874, 2019.
Article in English | MEDLINE | ID: mdl-31969876

ABSTRACT

Mycobacterium avium spp. paratuberculosis (MAP) is the causative agent of Johne's disease (JD), also known as paratuberculosis, in ruminants. The mechanisms of JD pathogenesis are not fully understood, but it is known that MAP subverts the host immune system by using macrophages as its primary reservoir. MAP infection in macrophages is often studied in healthy cows or experimentally infected calves, but reports on macrophages from naturally infected cows are lacking. In our study, primary monocyte-derived macrophages (MDMs) from cows diagnosed as positive (+) or negative (-) for JD were challenged in vitro with live MAP. Analysis using next-generation RNA sequencing revealed that macrophages from JD(+) cows did not present a definite pattern of response to MAP infection. Interestingly, a considerable number of genes, up to 1436, were differentially expressed in JD(-) macrophages. The signatures of the infection time course of 1, 4, 8, and 24 h revealed differential expression of ARG2, COL1A1, CCL2, CSF3, IL1A, IL6, IL10, PTGS2, PTX3, SOCS3, TNF, and TNFAIP6 among other genes, with major effects on host signaling pathways. While several immune pathways were affected by MAP, other pathways related to hepatic fibrosis/hepatic stellate cell activation, lipid homeostasis, such as LXR/RXR (liver X receptor/retinoid X receptor) activation pathways, and autoimmune diseases (rheumatoid arthritis or atherosclerosis) also responded to the presence of live MAP. Comparison of the profiles of the unchallenged MDMs from JD(+) vs. JD(-) cows showed that 868 genes were differentially expressed, suggesting that these genes were already affected before monocytes differentiated into macrophages. The downregulated genes predominantly modified the general cell metabolism by downregulating amino acid synthesis and affecting cholesterol biosynthesis and other energy production pathways while introducing a pro-fibrotic pattern associated with foam cells. The upregulated genes indicated that lipid homeostasis was already supporting fat storage in uninfected JD(+) MDMs. For JD(+) MDMs, differential gene expression expounds long-term mechanisms established during disease progression of paratuberculosis. Therefore, MAP could further promote disease persistence by influencing long-term macrophage behavior by using both tolerance and fat-storage states. This report contributes to a better understanding of MAP's controls over the immune cell response and mechanisms of MAP survival.


Subject(s)
Foam Cells/immunology , Immunity, Innate/immunology , Macrophages/immunology , Macrophages/microbiology , Mycobacterium avium subsp. paratuberculosis/immunology , Paratuberculosis/immunology , Transcriptome/immunology , Animals , Cattle , Cattle Diseases/immunology , Cattle Diseases/microbiology , Foam Cells/microbiology , Gene Expression Profiling/methods , Immune Tolerance/immunology , Monocytes/immunology , Monocytes/microbiology , Phenotype
11.
Int J Mol Sci ; 19(9)2018 Aug 24.
Article in English | MEDLINE | ID: mdl-30149509

ABSTRACT

MicroRNAs (miRNA) regulate mRNA networks to coordinate cellular functions. In this study, we constructed gene co-expression networks to detect miRNA modules (clusters of miRNAs with similar expression patterns) and miRNA⁻mRNA pairs associated with blood (triacylglyceride and nonesterified fatty acids) and milk (milk yield, fat, protein, and lactose) components and milk fatty acid traits following dietary supplementation of cows' diets with 5% linseed oil (LSO) (n = 6 cows) or 5% safflower oil (SFO) (n = 6 cows) for 28 days. Using miRNA transcriptome data from mammary tissues of cows for co-expression network analysis, we identified three consensus modules: blue, brown, and turquoise, composed of 70, 34, and 86 miRNA members, respectively. The hub miRNAs (miRNAs with the most connections with other miRNAs) were miR-30d, miR-484 and miR-16b for blue, brown, and turquoise modules, respectively. Cell cycle arrest, and p53 signaling and transforming growth factor⁻beta (TGF-ß) signaling pathways were the common gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways enriched for target genes of the three modules. Protein percent (p = 0.03) correlated with the turquoise module in LSO treatment while protein yield (p = 0.003) and milk yield (p = 7 × 10-04) correlated with the turquoise model, protein and milk yields and lactose percent (p < 0.05) correlated with the blue module and fat percent (p = 0.04) correlated with the brown module in SFO treatment. Several fatty acids correlated (p < 0.05) with the blue (CLA:9,11) and brown (C4:0, C12:0, C22:0, C18:1n9c and CLA:10,12) modules in LSO treatment and with the turquoise (C14:0, C18:3n3 and CLA:9,11), blue (C14:0 and C23:0) and brown (C6:0, C16:0, C22:0, C22:6n3 and CLA:10,12) modules in SFO treatment. Correlation of miRNA and mRNA data from the same animals identified the following miRNA⁻mRNA pairs: miR-183/RHBDD2 (p = 0.003), miR-484/EIF1AD (p = 0.011) and miR-130a/SBSPON (p = 0.004) with lowest p-values for the blue, brown, and turquoise modules, respectively. Milk yield, protein yield, and protein percentage correlated (p < 0.05) with 28, 31 and 5 miRNA⁻mRNA pairs, respectively. Our results suggest that, the blue, brown, and turquoise modules miRNAs, hub miRNAs, miRNA⁻mRNA networks, cell cycle arrest GO term, p53 signaling and TGF-ß signaling pathways have considerable influence on milk and blood phenotypes following dietary supplementation of dairy cows' diets with 5% LSO or 5% SFO.


Subject(s)
Gene Expression Regulation , Gene Regulatory Networks , Metabolome , MicroRNAs/genetics , Milk , Quantitative Trait, Heritable , RNA Interference , RNA, Messenger/genetics , Animals , Biomarkers , Computational Biology/methods , Gene Expression Profiling , Gene Ontology , Genetic Association Studies , Metabolomics/methods , Phenotype , Transcriptome
12.
Oncotarget ; 8(3): 4008-4042, 2017 Jan 17.
Article in English | MEDLINE | ID: mdl-28008141

ABSTRACT

Gynecological cancers are known for being very aggressive at their advanced stages. Indeed, the survival rate of both ovarian and endometrial cancers is very low when diagnosed lately and the success rate of current chemotherapy regimens is not very efficient. One of the main reasons for this low success rate is the acquired chemoresistance of these cancers during their progression. The mechanisms responsible for this acquired chemoresistance are numerous, including efflux pumps, repair mechanisms, survival pathways (PI3K/AKT, MAPK, EGFR, mTOR, estrogen signaling) and tumor suppressors (P53 and Par-4). To overcome these resistances, a new type of therapy has emerged named targeted therapy. The principle of targeted therapy is simple, taking advantage of changes acquired in malignant cancer cells (receptors, proteins, mechanisms) by using compounds specifically targeting these, thus limiting their action on healthy cells. Targeted therapies are emerging and many clinical trials targeting these pathways, frequently involved in chemoresistance, have been tested on gynecological cancers. Despite some targets being less efficient than expected as mono-therapies, the combination of compounds seems to be the promising avenue. For instance, we demonstrate using ChIP-seq analysis that estrogen downregulate tumor suppressor Par-4 in hormone-dependent cells by directly binding to its DNA regulatory elements and inhibiting estrogen signaling could reinstate Par-4 apoptosis-inducing abilities. This review will focus on the chemoresistance mechanisms and the clinical trials of targeted therapies associated with these, specifically for endometrial and ovarian cancers.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Endometrial Neoplasms/drug therapy , Molecular Targeted Therapy/methods , Ovarian Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis Regulatory Proteins/genetics , Clinical Trials as Topic , Endometrial Neoplasms/genetics , Estrogens/pharmacology , Estrogens/therapeutic use , Female , Gene Regulatory Networks/drug effects , Humans , Ovarian Neoplasms/genetics , Treatment Outcome
13.
Methods Mol Biol ; 1528: 229-243, 2017.
Article in English | MEDLINE | ID: mdl-27854025

ABSTRACT

Because of its profound influence on DNA accessibility for protein binding and thus on the regulation of diverse biological processes, nucleosome positioning has been studied for many years. In the past decade, high-throughput sequencing technologies have opened new perspectives in this research field by allowing the study of nucleosome positioning and occupancy on a genome-wide scale, therefore providing understanding on important aspects of chromatin packaging, as well as on various chromatin-template processes like transcription. In this chapter, we provide the protocol of MNase sequencing for the genome-wide mapping of nucleosomes using MNase to generate mononucleosomal DNA fragments and next-generation sequencing technology to identify their individual location.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Nucleosomes/metabolism , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/physiology , Chromosome Mapping , Histones/metabolism , Humans , Sequence Analysis, DNA/methods
14.
Oncotarget ; 7(47): 77276-77290, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27764788

ABSTRACT

Chromosomal and genome abnormalities at the 3p21.3 locus are frequent events linked to epithelial cancers, including ovarian and breast cancers. Genes encoded in the 3p21.3 cluster include HYAL1, HYAL2 and HYAL3 members of hyaluronidases involved in the breakdown of hyaluronan, an abundant component of the vertebrate extracellular matrix. However, the transcriptional regulation of HYAL genes is poorly defined. Here, we identified the estrogen receptor ERα as a negative regulator of HYAL1 expression in breast cancer cells. Integrative data mining using METABRIC dataset revealed a significant inverse correlation between ERα and HYAL1 gene expression in human breast tumors. ChIP-Seq analysis identified several ERα binding sites within the 3p21.3 locus, supporting the role of estrogen as an upstream signal that diversely regulates the expression of 3p21.3 genes at both proximal and distal locations. Of these, HYAL1 was repressed by estrogen through ERα binding to a consensus estrogen response element (ERE) located in the proximal promoter of HYAL1 and flanked by an Sp1 binding site, required to achieve optimal estrogen repression. The repressive chromatin mark H3K27me3 was increased at the proximal HYAL1 ERE but not at other EREs contained in the cluster, providing a mechanism to selectively downregulate HYAL1. The HYAL1 repression was also specific to ERα and not to ERß, whose expression did not correlate with HYAL1 in human breast tumors. This study identifies HYAL1 as an ERα target gene and provides a functional framework for the direct effect of estrogen on 3p21.3 genes in breast cancer cells.


Subject(s)
Breast Neoplasms/genetics , Down-Regulation , Estrogen Receptor alpha/metabolism , Hyaluronoglucosaminidase/genetics , Sp1 Transcription Factor/metabolism , Binding Sites , Breast Neoplasms/metabolism , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Chromosomes, Human, Pair 3/chemistry , Chromosomes, Human, Pair 3/genetics , Databases, Genetic , Estradiol/pharmacology , Female , GPI-Linked Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Hyaluronoglucosaminidase/chemistry , MCF-7 Cells , Multigene Family , Promoter Regions, Genetic
15.
Diabetes ; 65(12): 3705-3717, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27585521

ABSTRACT

Poor glycemic control profoundly affects protein expression and the cell signaling action that contributes to glycemic memory and irreversible progression of diabetic nephropathy (DN). We demonstrate that SHP-1 is elevated in podocytes of diabetic mice, causing insulin unresponsiveness and DN. Thus, sustained SHP-1 expression caused by hyperglycemia despite systemic glucose normalization could contribute to the glycemic memory effect in DN. Microalbuminuria, glomerular filtration rate, mesangial cell expansion, and collagen type IV and transforming growth factor-ß expression were significantly increased in diabetic Ins2+/C96Y mice compared with nondiabetic Ins2+/+ mice and remained elevated despite glucose normalization with insulin implants. A persistent increase of SHP-1 expression in podocytes despite normalization of systemic glucose levels was associated with sustained inhibition of the insulin signaling pathways. In cultured podocytes, high glucose levels increased mRNA, protein expression, and phosphatase activity of SHP-1, which remained elevated despite glucose concentration returning to normal, causing persistent insulin receptor-ß inhibition. Histone posttranslational modification analysis showed that the promoter region of SHP-1 was enriched with H3K4me1 and H3K9/14ac in diabetic glomeruli and podocytes, which remained elevated despite glucose level normalization. Hyperglycemia induces SHP-1 promoter epigenetic modifications, causing its persistent expression and activity and leading to insulin resistance, podocyte dysfunction, and DN.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Epigenesis, Genetic/genetics , Podocytes/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Animals , Cell Line , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Glomerular Filtration Rate/physiology , Hyperglycemia/genetics , Hyperglycemia/metabolism , Immunohistochemistry , Insulin Resistance/genetics , Insulin Resistance/physiology , Kidney Glomerulus/metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Promoter Regions, Genetic/genetics , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Signal Transduction/genetics , Signal Transduction/physiology
16.
Methods Mol Biol ; 1334: 261-72, 2015.
Article in English | MEDLINE | ID: mdl-26404156

ABSTRACT

Genetic information is organized in a complex structure composed of DNA and proteins together designated chromatin. Chromatin plays a dynamic role in transcriptional processes in that alteration of the interaction between its components results in the deregulation of cellular transcriptional program. Modification of epigenetic marks, variation in the precise positioning of nucleosomes, and consequent mobilization of nucleosomes regulate the access of various transcriptional factors to its underlying DNA template. Nucleosome-depleted regions, also designated open chromatin domains, are associated with active DNA regulatory elements, including promoters, enhancers, silencers, and insulators. Here, we describe the protocol of a rapid and simple technique entitled FAIRE (formaldehyde-assisted isolation of regulatory elements). Combined with high-throughput sequencing (FAIRE-seq), this procedure allows isolation of nucleosome-free regions and their mapping along the genome, thereby providing a global view of cell-specific regulatory elements.


Subject(s)
Chromatin/genetics , DNA/genetics , High-Throughput Nucleotide Sequencing/methods , Proteins/genetics , Regulatory Sequences, Nucleic Acid/genetics , Chromatin/chemistry , DNA/chemistry , Enhancer Elements, Genetic , Formaldehyde/chemistry , Insulator Elements/genetics , Promoter Regions, Genetic/genetics , Proteins/chemistry , Silencer Elements, Transcriptional/genetics
17.
Nucleic Acids Res ; 43(20): 9742-56, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26319018

ABSTRACT

Gene regulatory programs in different cell types are largely defined through cell-specific enhancers activity. The histone variant H2A.Z has been shown to play important roles in transcription mainly by controlling proximal promoters, but its effect on enhancer functions remains unclear. Here, we demonstrate by genome-wide approaches that H2A.Z is present at a subset of active enhancers bound by the estrogen receptor alpha (ERα). We also determine that H2A.Z does not influence the local nucleosome positioning around ERα enhancers using ChIP sequencing at nucleosomal resolution and unsupervised pattern discovery. We further highlight that H2A.Z-enriched enhancers are associated with chromatin accessibility, H3K122ac enrichment and hypomethylated DNA. Moreover, upon estrogen stimulation, the enhancers occupied by H2A.Z produce enhancer RNAs (eRNAs), and recruit RNA polymerase II as well as RAD21, a member of the cohesin complex involved in chromatin interactions between enhancers and promoters. Importantly, their recruitment and eRNAs production are abolished by H2A.Z depletion, thereby revealing a novel functional link between H2A.Z occupancy and enhancer activity. Taken together, our findings suggest that H2A.Z acts as an important player for enhancer functions by establishing and maintaining a chromatin environment required for RNA polymerase II recruitment, eRNAs transcription and enhancer-promoters interactions, all essential attributes of enhancer activity.


Subject(s)
Enhancer Elements, Genetic , Histones/metabolism , Transcriptional Activation , Cell Cycle Proteins/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA Methylation , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Genomics , Histone Code , MCF-7 Cells , Nucleosomes/metabolism , RNA Polymerase II/metabolism , Cohesins
18.
PLoS One ; 10(3): e0119387, 2015.
Article in English | MEDLINE | ID: mdl-25803847

ABSTRACT

Mitogen-activated protein kinase 3/1 (Mapk3/1) pathway is critical for LH signal transduction during ovulation. However, the mechanisms remain incompletely understood. We hypothesized that Mapk pathway regulates ovulation through transcriptional regulation of ovulatory genes. To test this hypothesis we used immature mice superovulated with equine and human chorionic gonadotropins (eCG and hCG) and PD0325901, to inhibit hCG-induced Mapk3/1 activity. Mice received either the inhibitor PD0325901 (25 µg/g, i.p.) or vehicle at 2h before hCG stimulation. Administration of the inhibitor abolished Mapk3/1 phosphorylation in granulosa cells. While vehicle-treated mice ovulated normally, there were no ovulations in inhibitor-treated mice. First, we analyzed gene expression in granulosa cells at 0h, 1h and 4h post-hCG. There was expected hCG-driven increase in mRNA abundance of many ovulation-related genes including Ptgs2 in vehicle-treated granulosa cells, but not (P<0.05) in inhibitor-treated group. There was also reduced mRNA and protein abundance of the transcription factor, early growth response 1 (Egr1) in inhibitor-treated granulosa cells. We then used GRMO2 cell-line to test if Egr1 is recruited to promoter of Ptgs2 followed by chromatin immunoprecipitation with either Egr1 or control antibody. Enrichment of the promoter regions in immunoprecipitants of Egr1 antibody indicated that Egr1 binds to the Ptgs2 promoter. We then knocked down Egr1 expression in mouse primary granulosa cells using siRNA technology. Treatment with Egr1-siRNA inhibited Egr1 transcript accumulation, which was associated with reduced expression of Ptgs2 when compared to control-siRNA treated granulosa cells. These data demonstrate that transient inhibition of LH-stimulated MAPK3/1 activity abrogates ovulation in mice. We conclude that Mapk3/1 regulates ovulation, at least in part, through Egr1 and its target gene, Ptgs2 in granulosa cells of ovulating follicles in mice.


Subject(s)
MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Ovulation/drug effects , Protein Kinase Inhibitors/pharmacology , Animals , Benzamides/pharmacology , Cyclooxygenase 2/metabolism , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Early Growth Response Protein 1/metabolism , Female , Gonadotropins/pharmacology , Granulosa Cells/drug effects , Granulosa Cells/enzymology , Horses , Humans , MAP Kinase Signaling System/physiology , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Ovarian Follicle/drug effects , Ovarian Follicle/enzymology , Ovulation/physiology , Primary Cell Culture , Superovulation/drug effects
19.
Nucleic Acids Res ; 42(18): 11339-48, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25223786

ABSTRACT

Chromatin constitutes a repressive barrier to the process of ligand-dependent transcriptional activity of nuclear receptors. Nucleosomes prevent the binding of estrogen receptor α (ERα) in absence of ligand and thus represent an important level of transcriptional regulation. Here, we show that in breast cancer MCF-7 cells, TLE3, a co-repressor of the Groucho/Grg/TLE family, interacts with FoxA1 and is detected at regulatory elements of ERα target genes in absence of estrogen. As a result, the chromatin is maintained in a basal state of acetylation, thus preventing ligand-independent activation of transcription. In absence of TLE3, the basal expression of ERα target genes induced by E2 is increased. At the TFF1 gene, the recruitment of TLE3 to the chromatin is FoxA1-dependent and prevents ERα and RNA polymerase II recruitment to TFF1 gene regulatory elements. Moreover, the interaction of TLE3 with HDAC2 results in the maintenance of acetylation at a basal level. We also provide evidence that TLE3 is recruited at several other regulatory elements of ERα target genes and is probably an important co-regulator of the E2 signaling pathway. In sum, our results describe a mechanism by which TLE3 affects ligand dependency in ERα-regulated gene expression via its binding restricting function and its role in gene regulation by histone acetylation.


Subject(s)
Co-Repressor Proteins/metabolism , Estrogen Receptor alpha/metabolism , Gene Expression Regulation , Cell Line , Chromatin/metabolism , Co-Repressor Proteins/physiology , Hepatocyte Nuclear Factor 3-alpha/physiology , Histone Deacetylases/metabolism , Histones/metabolism , Humans , MCF-7 Cells , Regulatory Elements, Transcriptional , Signal Transduction , Transcription, Genetic
20.
Cancer Res ; 74(7): 2015-25, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24520076

ABSTRACT

Tumor characteristics are decisive in the determination of treatment strategy for patients with breast cancer. Patients with estrogen receptor α (ERα)-positive breast cancer can benefit from long-term hormonal treatment. Nonetheless, the majority of patients will develop resistance to these therapies. Here, we investigated the role of the nuclear receptor liver receptor homolog-1 (LRH-1, NR5A2) in antiestrogen-sensitive and -resistant breast cancer cells. We identified genome-wide LRH-1-binding sites using ChIP-seq (chromatin immunoprecipitation sequencing), uncovering preferential binding to regions distal to transcriptional start sites. We further characterized these LRH-1-binding sites by integrating overlapping layers of specific chromatin marks, revealing that many LRH-1-binding sites are active and could be involved in long-range enhancer-promoter looping. Combined with transcriptome analysis of LRH-1-depleted cells, these results show that LRH-1 regulates specific subsets of genes involved in cell proliferation in antiestrogen-sensitive and antiestrogen-resistant breast cancer cells. Furthermore, the LRH-1 transcriptional program is highly associated with a signature of poor outcome and high-grade breast cancer tumors in vivo. Herein, we report the genome-wide location and molecular function of LRH-1 in breast cancer cells and reveal its therapeutic potential for the treatment of breast cancers, notably for tumors resistant to treatments currently used in therapies.


Subject(s)
Breast Neoplasms/genetics , Estrogen Antagonists/therapeutic use , Receptors, Cytoplasmic and Nuclear/physiology , Transcription, Genetic , Binding Sites , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Proliferation , Chromatin/physiology , Cyclin D1/genetics , Drug Resistance, Neoplasm , Estrogen Receptor alpha/physiology , Humans , MCF-7 Cells , Receptors, Estrogen/physiology , Receptors, G-Protein-Coupled/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...