Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Ovarian Res ; 14(1): 178, 2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34930403

ABSTRACT

Maternal metabolic disorders such as obesity and diabetes are detrimental factors that compromise fertility and the success rates of medically assisted procreation procedures. During metabolic stress, adipose tissue is more likely to release free fatty acids (FFA) in the serum resulting in an increase of FFA levels not only in blood, but also in follicular fluid (FF). In humans, high concentrations of palmitic acid and stearic acid reduced granulosa cell survival and were associated with poor cumulus-oocyte complex (COC) morphology. Obesity and high levels of circulating FFA were also causatively linked to hampered insulin sensitivity in cells and compensatory hyperinsulinemia. To provide a global picture of the principal upstream signaling pathways and genomic mechanisms involved in this metabolic context, human granulosa-like tumor cells (KGN) were treated with a combination of palmitic acid, oleic acid, and stearic acid at the higher physiological concentrations found in the follicular fluid of women with a higher body mass index (BMI) (≥ 30.0 kg/m2). We also tested a high concentration of insulin alone and in combination with high concentrations of fatty acids. Transcription analysis by RNA-seq with a cut off for fold change of 1.5 and p-value 0.05 resulted in thousands of differentially expressed genes for each treatment. Using analysis software such as Ingenuity Pathway Analysis (IPA), we were able to establish that high concentrations of FFA affected the expression of genes mainly related to glucose and insulin homoeostasis, fatty acid metabolism, as well as steroidogenesis and granulosa cell differentiation processes. The combination of insulin and high concentrations of FFA affected signaling pathways related to apoptosis, inflammation, and oxidative stress. Taken together, our results provided new information on the mechanisms that might be involved in human granulosa cells exposed to high concentrations of FFA and insulin in the contexts of metabolism disorders.


Subject(s)
Fatty Acids, Nonesterified/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Granulosa Cell Tumor/genetics , Insulin/pharmacology , Apoptosis/genetics , Cell Line, Tumor , Energy Metabolism/genetics , Female , Humans , Inflammation/genetics , Mitochondria/genetics , Oxidative Stress/genetics
2.
Biol Reprod ; 103(3): 583-598, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32427331

ABSTRACT

The female reproductive function largely depends on timing and coordination between follicle-stimulating hormone (FSH) and luteinizing hormone. Even though it was suggested that these hormones act on granulosa cells via shared signaling pathways, mainly protein kinases A, B, and C (PKA, PKB, and PKC), there is still very little information available on how these signaling pathways are regulated by each hormone to provide such differences in gene expression throughout folliculogenesis. To obtain a global picture of the principal upstream factors involved in PKA, PKB, and PKC signaling in granulosa cells, human granulosa-like tumor cells (KGN) were treated with FSH or specific activators (forskolin, SC79, and phorbol 12-myristate 13-acetate) for each pathway to analyze gene expression with RNA-seq technology. Normalization and cutoffs (FC 1.5, P ≤ 0.05) revealed 3864 differentially expressed genes between treatments. Analysis of major upstream regulators showed that PKA is a master kinase of early cell differentiation as its activation resulted in the gene expression profile that accompanies granulosa cell differentiation. Our data also revealed that the activation of PKC in granulosa cells is also a strong differentiation signal that could control "advanced" differentiation in granulosa cells and the inflammatory cascade that occurs in the dominant follicle. According to our results, PKB activation provides support for PKA-stimulated gene expression and is also involved in granulosa cell survival throughout follicular development. Taken together, our results provide new information on PKA, PKB, and PKC signaling pathways and their roles in stimulating a follicle at the crossroad between maturation/ovulation and atresia.


Subject(s)
Gonadotropins/physiology , Granulosa Cell Tumor/genetics , Ovarian Neoplasms/genetics , Protein Kinases/genetics , Protein Kinases/physiology , Signal Transduction/genetics , Cell Survival , Female , Follicle Stimulating Hormone/pharmacology , Gene Expression Regulation, Developmental , Granulosa Cell Tumor/physiopathology , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Humans , Inflammation/genetics , Inflammation/physiopathology , Ovarian Neoplasms/physiopathology , Transcriptome
3.
Biol Reprod ; 96(4): 855-865, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28383663

ABSTRACT

The developmental competence of an oocyte is its capacity to resume maturation, undergo successful fertilization, and reach the blastocyst stage. This competence is acquired through interaction with somatic cells of the follicle. Cumulus and granulosa cells support oocyte development, while the oocyte influences follicular cell growth and differentiation. Studies suggest that follicle-stimulating hormone and luteinizing hormone play an essential role in oocyte competence acquisition through signaling initiated by protein kinases A and C (PKA and PKC) in granulosa cells. Using a microarray and RT-qPCR, the transcriptome of human granulosa-like tumor cells (KGN) treated for 24 h with forskolin (FSK) or phorbol 12-myristate 13-acetate (PMA) was analyzed to determine the effects of PKA and PKC stimulation on gene expression. Protein-kinase-driven signaling appeared to involve five major upstream regulators, namely epidermal growth factor (EGF), transforming growth factor beta 1 (TGFß1), vascular endothelial growth factor (VEGF), basic fibroblast growth factor (FGF2), and hepatocyte growth factor (HGF). Gene associations with seven major ovarian functions were identified: Prostaglandin- endoperoxide synthase 2 (PTGS2), interleukin 8 (IL8), and interleukin 6 (IL6) with inflammation; Steroidogenic acute regulatory protein (STAR), cytochrome P450scc (CYP11A1), and cytochrome P450 family 19 subfamily A member 1 (CYP19A1) with steroidogenesis; Vascular endothelial growth factor C (VEGFC), Vascular endothelial growth factor A (VEGFA), and C-X-C chemokine receptor type 4 (CXCR4) with angiogenesis; Amphiregulin (AREG), epidermal growth factor receptor (EGFR), and sprouty RTK signaling antagonist 2 (SPRY2) with differentiation, BCL2 associated X (BAX), BCL2 like 12 (BCL2L12), and caspase 1(CASP1) with apoptosis, Cyclin D1 (CCND1), cyclin B1 (CCNB1), and cyclin B2 (CCNB2) with division; and Matrix metalloproteinase-1 (MMP1), Matrix metallopeptidase 9 (MMP9), and TIMP metallopeptidase inhibitor 1 (TIMP1) with ovulation. Overall, these results indicate that signaling via both PKA and PKC potentiates gene regulation of functions such as inflammation and apoptosis, while functions such as differentiation, ovulation and angiogenesis are partial to one kinase or the other. These results improve understanding of the pathways underlying the most important changes that occur in the follicle prior to ovulation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation, Enzymologic/physiology , Granulosa Cell Tumor/metabolism , Transcriptome , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/genetics , Estradiol/biosynthesis , Humans , Protein Array Analysis , Protein Kinase C/genetics , Protein Kinase C/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...