Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 165
Filter
1.
Am J Reprod Immunol ; 91(5): e13854, 2024 May.
Article in English | MEDLINE | ID: mdl-38716832

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) is a common endocrine-metabolic disorder characterized by oligo-anovulation, hyperandrogenism, and polycystic ovaries, with hyperandrogenism being the most prominent feature of PCOS patients. However, whether excessive androgens also exist in the ovarian microenvironment of patients with PCOS, and their modulatory role on ovarian immune homeostasis and ovarian function, is not clear. METHODS: Follicular fluid samples from patients participating in their first in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) treatment were collected. Androgen concentration of follicular fluid was assayed by chemiluminescence, and the macrophage M1:M2 ratio was detected by flow cytometry. In an in vitro model, we examined the regulatory effects of different concentrations of androgen on macrophage differentiation and glucose metabolism levels using qRT-PCR, Simple Western and multi-factor flow cytometry assay. In a co-culture model, we assessed the effect of a hyperandrogenic environment in the presence or absence of macrophages on the function of granulosa cells using qRT-PCR, Simple Western, EdU assay, cell cycle assay, and multi-factor flow cytometry assay. RESULTS: The results showed that a significantly higher androgen level and M1:M2 ratio in the follicular fluid of PCOS patients with hyperandrogenism. The hyperandrogenic environment promoted the expression of pro-inflammatory and glycolysis-related molecules and inhibited the expression of anti-inflammatory and oxidative phosphorylation-related molecules in macrophages. In the presence of macrophages, a hyperandrogenic environment significantly downregulated the function of granulosa cells. CONCLUSION: There is a hyperandrogenic microenvironment in the ovary of PCOS patients with hyperandrogenism. Hyperandrogenic microenvironment can promote the activation of ovarian macrophages to M1, which may be associated with the reprogramming of macrophage glucose metabolism. The increased secretion of pro-inflammatory cytokines by macrophages in the hyperandrogenic microenvironment would impair the normal function of granulosa cells and interfere with normal ovarian follicle growth and development.


Subject(s)
Androgens , Follicular Fluid , Granulosa Cells , Hyperandrogenism , Macrophages , Polycystic Ovary Syndrome , Humans , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/immunology , Female , Granulosa Cells/metabolism , Macrophages/immunology , Macrophages/metabolism , Hyperandrogenism/metabolism , Adult , Follicular Fluid/metabolism , Androgens/metabolism , Cells, Cultured , Macrophage Activation , Cellular Microenvironment , Coculture Techniques , Cell Differentiation
2.
Am J Reprod Immunol ; 91(4): e13847, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38661639

ABSTRACT

PROBLEM: Polycystic ovary syndrome (PCOS), a prevalent endocrine-metabolic disorder, presents considerable therapeutic challenges due to its complex and elusive pathophysiology. METHOD OF STUDY: We employed three machine learning algorithms to identify potential biomarkers within a training dataset, comprising GSE138518, GSE155489, and GSE193123. The diagnostic accuracy of these biomarkers was rigorously evaluated using a validation dataset using area under the curve (AUC) metrics. Further validation in clinical samples was conducted using PCR and immunofluorescence techniques. Additionally, we investigate the complex interplay among immune cells in PCOS using CIBERSORT to uncover the relationships between the identified biomarkers and various immune cell types. RESULTS: Our analysis identified ACSS2, LPIN1, and NR4A1 as key mitochondria-related biomarkers associated with PCOS. A notable difference was observed in the immune microenvironment between PCOS patients and healthy controls. In particular, LPIN1 exhibited a positive correlation with resting mast cells, whereas NR4A1 demonstrated a negative correlation with monocytes in PCOS patients. CONCLUSION: ACSS2, LPIN1, and NR4A1 emerge as PCOS-related diagnostic biomarkers and potential intervention targets, opening new avenues for the diagnosis and management of PCOS.


Subject(s)
Biomarkers , Mitochondria , Nuclear Receptor Subfamily 4, Group A, Member 1 , Polycystic Ovary Syndrome , Humans , Polycystic Ovary Syndrome/immunology , Polycystic Ovary Syndrome/metabolism , Female , Biomarkers/metabolism , Mitochondria/metabolism , Machine Learning , Adult , Mast Cells/immunology , Mast Cells/metabolism
3.
J Ovarian Res ; 15(1): 126, 2022 Dec 03.
Article in English | MEDLINE | ID: mdl-36463191

ABSTRACT

The survival of ovary granulosa cells (GC) is critical in the initiation and progression of polycystic ovary syndrome (PCOS) in females. Here, we found that the PCOS process is accompanied by massive GC pyroptosis resulting from Caspase-1 inflammasome activation. Administration of plumbagin, an effective compound isolated from plant medicine, can prevent the pyroptosis of GC and the onset of PCOS. Mechanistic study indicates the over-activation of the inflammasome in GC is due to the upregulation of WTAP, a key regulator of the RNA N6-methylase complex. WTAP mediates the mRNA N6-methylation of NLRP3 inflammasome component ASC and enhances ASC RNA stability, which results in the overactivation of the inflammasome in GCs from the PCOS model. Plumbagin treatment suppresses the WTAP-mediated N6-methylation of ASC mRNA and reduces the pyroptosis of GCs. This study supports the profound potential of plumbagin in PCOS treatment.


Subject(s)
Granulosa Cells , Naphthoquinones , Polycystic Ovary Syndrome , Pyroptosis , Female , Humans , Cell Cycle Proteins/genetics , Cell Cycle Proteins/immunology , Granulosa Cells/drug effects , Granulosa Cells/immunology , Inflammasomes/genetics , Inflammasomes/immunology , Methylation/drug effects , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/immunology , Pyroptosis/drug effects , Pyroptosis/genetics , Pyroptosis/immunology , RNA Splicing Factors/genetics , RNA Splicing Factors/immunology , RNA, Messenger , Naphthoquinones/immunology , Naphthoquinones/pharmacology , Naphthoquinones/therapeutic use
4.
J Ovarian Res ; 15(1): 30, 2022 Feb 28.
Article in English | MEDLINE | ID: mdl-35227296

ABSTRACT

BACKGROUND: Accumulating evidence suggests a strong association between polycystic ovary syndrome (PCOS) and ovarian cancer (OC), but the potential molecular mechanism remains unclear. In this study, we identified previously unrecognized genes that are significantly correlated with PCOS and OC via bioinformatics. MATERIALS AND METHODS: Multiple bioinformatic analyses, such as differential expression analysis, univariate Cox analysis, functional and pathway enrichment analysis, protein-protein interaction (PPI) network construction, survival analysis, and immune infiltration analysis, were utilized. We further evaluated the effect of OGN on FSHR expression via immunofluorescence. RESULTS: TCGA-OC, GSE140082 (for OC) and GSE34526 (for PCOS) datasets were downloaded. Twelve genes, including RNF144B, LPAR3, CRISPLD2, JCHAIN, OR7E14P, IL27RA, PTPRD, STAT1, NR4A1, OGN, GALNT6 and CXCL11, were identified as signature genes. Drug sensitivity analysis showed that OGN might represent a hub gene in the progression of PCOS and OC. Experimental analysis found that OGN could increase FSHR expression, indicating that OGN could regulate the hormonal response in PCOS and OC. Furthermore, correlation analysis indicated that OGN function might be closely related to m6A and ferroptosis. CONCLUSIONS: Our study identified a 12-gene signature that might be involved in the prognostic significance of OC. Furthermore, the hub gene OGN represent a significant gene involved in OC and PCOS progression by regulating the hormonal response.


Subject(s)
Intercellular Signaling Peptides and Proteins , Ovarian Neoplasms , Polycystic Ovary Syndrome , Female , Humans , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Chemokine CXCL11/genetics , Computational Biology , Dendritic Cells , Disease Progression , Drug Resistance, Neoplasm/genetics , Ferroptosis/genetics , Intercellular Signaling Peptides and Proteins/genetics , Interferon Regulatory Factors/genetics , Membrane Glycoproteins/metabolism , Methylation , N-Acetylgalactosaminyltransferases/genetics , Nerve Tissue Proteins/metabolism , Neutrophils , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/immunology , Polycystic Ovary Syndrome/pathology , Prognosis , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Receptors, FSH/genetics , Receptors, Interleukin/genetics , Receptors, Lysophosphatidic Acid/genetics , RNA, Messenger/metabolism , STAT1 Transcription Factor/genetics , Transcriptome , Ubiquitin-Protein Ligases/genetics , Polypeptide N-acetylgalactosaminyltransferase
5.
J Clin Endocrinol Metab ; 107(1): 39-52, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34529073

ABSTRACT

CONTEXT: Polycystic ovary syndrome (PCOS) is a common endocrine disorder associated with low-grade inflammation and increased incidence of pregnancy complications, but its influence on the maternal immune system in pregnancy is unknown. Longitudinal serum cytokine profiling is a sensitive measure of the complex immunological dynamics of pregnancy. OBJECTIVE: This work aimed to determine the immunological dynamics of serum cytokines throughout pregnancy in women with PCOS and compare it to pregnancy in women without PCOS. METHODS: A post hoc analysis was conducted of longitudinal serum samples from 2 randomized, placebo-controlled multicenter studies of pregnant women with PCOS and 2 studies of pregnant women without PCOS. Pregnant women with PCOS (n = 358) and without PCOS (n = 258, controls) provided 1752 serum samples from 4 time points in pregnancy (weeks 10, 19, 32, and 36). Main outcome measures included maternal serum levels of 22 cytokines and C-reactive protein (CRP) at 4 time points in pregnancy. RESULTS: Women with PCOS showed marked immunological changes in serum cytokines throughout pregnancy. Compared to controls, women with PCOS showed higher levels of 17 cytokines and CRP at week 10 of pregnancy and a distinct cytokine development throughout pregnancy. The immunological dynamics in women with PCOS was significantly affected by maternal body mass index, smoking, and fetal sex. CONCLUSION: Pregnancy in women with PCOS was associated with a strong early mobilization of inflammatory and other serum cytokines persisting throughout pregnancy, indicating a more activated immune status. These findings provide a novel basis for further study of PCOS and pregnancy complications.


Subject(s)
Cytokines/blood , Polycystic Ovary Syndrome/immunology , Pregnancy Complications/immunology , Adult , C-Reactive Protein/analysis , Case-Control Studies , Cytokines/immunology , Female , Humans , Longitudinal Studies , Polycystic Ovary Syndrome/blood , Pregnancy , Pregnancy Complications/blood , Young Adult
6.
J Ethnopharmacol ; 282: 114594, 2022 Jan 10.
Article in English | MEDLINE | ID: mdl-34480994

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Saffron petal has traditionally been used to treat a variety of diseases, such as gynecological disease such as primary dysmenorrhea and premenstrual tension. Polycystic Ovary Syndrome (PCOS) is a form of gynecological disease that causes amenorrhea, infertility, menopausal and urogenital disorders. This disease may be treated with saffron petals. AIM OF THE STUDY: In this study, the effects of saffron petal extract (SPE) and saffron petal anthocyanins (SPA) on ovarian hormones, steroidogenic enzymes, ovarian dysfunction, regulation of anti-inflammatory genes, and antioxidant factors in female PCOS mice were studied. METHODS AND RESULTS: The PCOS mouse model was induced by testosterone enanthate (TE), and an in vivo evaluation of whether the dietary consumption of SPE and SPA improved the PCOS-like symptoms was conducted. The luteinizing hormone (LH), testosterone, and estrogen levels increased in PCOS mice, but decreased following SPE and SPA treatment. In the PCOS mice, the reduced follicular-stimulating hormone (FSH) progesterone levels were restored to that of normal controls with SPE and SPA treatment in serum. The transcription level(s) of gonadotropin receptors (Fshr and Lhr), steroid receptors (Pgr, and Esr1), inflammatory markers (TNFα, IL1ß, IL6 and IL18), inflammatory-related factors (NF-κB, NF-κB p65, IκB) and antioxidant enzymes (GPx, SOD, CAT, GST, and GSH) changed under the PCOS condition. Moreover, they were regulated by SPE and SPA treatment in PCOS mice ovaries. The reproductive tissues of TE induced PCOS mice were restored into estrogenic conditions from androgen environments. The study of antioxidant activity of SPE and SPA using FRAP and DPPH tests showed high antioxidant activity. CONCLUSION: These results suggest that SPE and SPA ameliorates symptoms of PCOS by improving dysregulation of ovarian steroids, steroidogenic, antioxidant enzymes and inflammatory markers in PCOS mice.


Subject(s)
Crocus , Estrogens/blood , Luteinizing Hormone/blood , Ovary , Plant Extracts , Polycystic Ovary Syndrome , Testosterone/blood , Animals , Anthocyanins/pharmacology , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Disease Models, Animal , Drug Monitoring , Ethnopharmacology/methods , Female , Mice , Ovary/drug effects , Ovary/metabolism , Plant Extracts/chemistry , Plant Extracts/pharmacology , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/immunology , Polycystic Ovary Syndrome/metabolism
7.
Int J Mol Sci ; 22(24)2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34948471

ABSTRACT

Hyperandrogenemia and ovulatory dysfunction are hallmarks of polycystic ovary syndrome (PCOS), pointing to a deranged hypothalamus-pituitary-ovarian (HPO) axis. An autoimmune etiology of PCOS is suspected in a subset of patients due to the relatively high concordance of PCOS with common autoimmune diseases. For this reason, we tested the hypothesis that natural autoantibodies (aAb) to the follicle-stimulating hormone receptor (FSHR) or luteinizing hormone receptor (LHR) are prevalent in PCOS. To this end, new luminometric assays for quantifying aAb to the FSHR (FSHR-aAb) or LHR (LHR-aAb) were developed using full-length recombinant human receptors as fusion proteins with luciferase as reporter. Prevalence of FSHR-aAb and LHR-aAb was determined in serum samples from healthy controls and PCOS patients. Steroid hormone profiles were compared between patients with and without FSHR-aAb or LHR-aAb. Signal linearity and detection ranges were characterized and both methods passed basic performance quality checks. The analysis revealed a relatively low prevalence, with 4 out of 430 samples positive for FSHR-aAb in the control versus 11 out of 550 samples in the PCOS group, i.e., 0.9% versus 2.0%, respectively. Similarly, there were only 5 samples positive for LHR-aAb in the control versus 2 samples in the PCOS group, i.e., 1.2% versus 0.4%, respectively. Samples positive for FSHR-aAb displayed steroid hormones in the typical range of PCOS patients, whereas the two samples positive for LHR-aAb showed relatively elevated free testosterone in relation to total testosterone concentrations with unclear significance. We conclude that the FSHR and LHR constitute potential autoantigens in human subjects. However, the prevalence of specific autoantibodies to these receptors is relatively low, both in control subjects and in women with PCOS. It is therefore unlikely that autoimmunity to the LHR or FSHR constitutes a frequent cause of hyperandrogenemia or ovulatory dysfunction in PCOS.


Subject(s)
Autoantibodies/blood , Polycystic Ovary Syndrome/immunology , Receptors, FSH/immunology , Receptors, LH/immunology , Case-Control Studies , Female , Humans , Luciferases/genetics , Luciferases/metabolism , Polycystic Ovary Syndrome/blood , Prevalence , Receptors, FSH/genetics , Receptors, LH/genetics , Recombinant Proteins/immunology , Testosterone
8.
Reprod Biol Endocrinol ; 19(1): 175, 2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34847942

ABSTRACT

BACKGROUND: Immune dysfunction is one of the mechanisms to promote polycystic ovary syndrome (PCOS). Various immune cells have been reported to be involved in the development of PCOS. Meanwhile, the disturbance of metabolism is closely related to PCOS. The aim of this study is to explore the association of mucosal-associated invariant T (MAIT) cells and myeloid-derived suppressor cells (MDSCs) with the metabolic dysfunction in PCOS. METHODS: 68 PCOS patients and 40 controls were recruited in this study and we collected the peripheral blood of participants' during their follicular phase. The frequencies of MAIT cells and MDSCs were determined by flow cytometry after being stained with different monoclonal antibodies. And the concentrations of cytokines were determined by ELISA. RESULTS: Compared to controls with normal metabolism, the frequency of MDSCs, CD8+MAIT cells and CD38+CD8+MAIT cells were significantly decreased in PCOS patients with normal metabolism, however, proportion of CD4+MAIT cells exhibited a noticeable increase. Similar results of CD8+MAIT, CD38+CD8+MAIT cells and reduced expression of IL-17 were observed in PCOS patients with metabolic dysfunction as compared to controls with metabolic disorders. PCOS patients with excessive testosterone levels displayed significantly decreased levels of CD8+MAIT, CD38+CD8+MAIT cells, MDSCs and Mo-MDSCs as compared to PCOS patients with normal testosterone concentrations. PCOS patients with abnormal weight showed a lower level and activation of CD8+MAIT cells. On the contrary, they displayed an enrichment of CD4+MAIT cells. PCOS patients with glucose metabolic disorder displayed a remarkable dysregulation of MDSCs and Mo-MDSCs. MDSCs were positively correlated with MAIT cells. Negative correlations between the frequency of CD8+MAIT cells, CD38+CD8+MAIT cells and body mass index were revealed. CD4+MAIT cells positively correlated with BMI. Mo-MDSCs were found to be negatively related to the levels of 2hour plasma glucose and HOMA-IR index. CONCLUSION: The impairment of CD8+MAIT cells and MDSCs is involved in the metabolic dysfunction of PCOS.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Mucosal-Associated Invariant T Cells/immunology , Myeloid-Derived Suppressor Cells/immunology , Polycystic Ovary Syndrome/immunology , Adult , CD8-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Female , Humans , Mucosal-Associated Invariant T Cells/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Polycystic Ovary Syndrome/metabolism , Young Adult
9.
J Ovarian Res ; 14(1): 155, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34781996

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) is a disease with chronic nonspecific low-grade inflammation. The imbalance of immune cells exists in PCOS. Several studies have found that heat shock protein 70 (HSP70) may be involved in the immunological pathogenesis of PCOS, but the relationship between HSP70 and Regulatory T cell (Treg)/T helper cell 17(Th17) ratio remains unclear. This study aims to explore the correlation between HSP70 and Treg/Th17 ratio and to provide evidence for the role of HSP70 in the immunological etiology of PCOS. RESULTS: There was no significant difference in age and body mass index (BMI) between the two groups. The concentrations of basal estradiol (E2), basal follicle-stimulating hormone (FSH) did not show a significant difference between the two groups. The concentrations of basal luteinizing hormone (LH) (P < 0.01), testosterone (T) (P < 0.01), glucose (P < 0.001) and insulin (P < 0.001) in PCOS patients were significantly higher than those in the control group. The protein levels of HSP70 were significantly higher in serum in the PCOS group (P < 0.001). The percentage of Treg cells was significantly lower (P < 0.01), while the percentage of the Th17 cells of the PCOS group was significantly higher than that of the control group (P < 0.05). The ratio of Treg/Th17 in the PCOS group was significantly lower (P < 0.001). The concentrations of Interleukin (IL)-6, IL-17, and IL-23 were significantly higher, while the levels of IL-10 and Transforming growth factor-ß (TGF-ß) were significantly lower in the PCOS group (P < 0.001). Spearman rank correlation analysis showed a strong negative correlation of serum HSP70 levels with Treg/Th17 ratio, IL-10, and TGF-ß levels. In contrast, HSP70 levels were significantly positively correlated with IL-6, IL-17, IL-23, LH, insulin, and glucose levels. CONCLUSION: The abnormal level of HSP70 is correlated with Treg/Th17 imbalance and corresponding cytokines, which indicates that HSP70 may play an important role in PCOS immunologic pathogenesis.


Subject(s)
HSP70 Heat-Shock Proteins/immunology , Polycystic Ovary Syndrome/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Adult , Blood Glucose/metabolism , Female , HSP70 Heat-Shock Proteins/blood , HSP70 Heat-Shock Proteins/metabolism , Humans , Insulin/blood , Interleukin-10/blood , Interleukin-10/immunology , Interleukin-17/blood , Interleukin-17/immunology , Interleukin-23/blood , Interleukin-23/immunology , Interleukin-6/blood , Interleukin-6/immunology , Luteinizing Hormone/blood , Lymphocyte Count , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/metabolism , Testosterone/blood , Transforming Growth Factor beta/blood , Transforming Growth Factor beta/immunology , Young Adult
10.
Front Endocrinol (Lausanne) ; 12: 686054, 2021.
Article in English | MEDLINE | ID: mdl-34248847

ABSTRACT

Background: rs13405728 was identified as one of the most prevalent susceptibility loci for polycystic ovary syndrome (PCOS) in Han Chinese and Caucasian women. However, the target genes and potential mechanisms of the rs13405728 locus remain to be determined. Methods: Three-dimensional (3D) genome interactions from the ovary tissue were characterized via high-through chromosome conformation capture (Hi-C) and Capture Hi-C technologies to identify putative targets at the rs13405728 locus. Combined analyses of eQTL, RNA-Seq, DNase-Seq, ChIP-Seq, and sing-cell sequencing were performed to explore the molecular roles of these target genes in PCOS. PCOS-like mice were applied to verify the expression patterns. Results: Generally, STON1 and FSHR were identified as potential targets of the rs13405728 locus in 3D genomic interactions with epigenomic regulatory peaks, with STON1 (P=0.0423) and FSHR (P=0.0013) being highly expressed in PCOS patients. STON1 co-expressed genes were associated with metabolic processes (P=0.0008) in adipocytes (P=0.0001), which was validated in the fat tissue (P<0.0001) and ovary (P=0.0035) from fat-diet mice. The immune system process (GO:0002376) was enriched in FSHR co-expressed genes (P=0.0002) and PCOS patients (P=0.0002), with CD4 high expression in PCOS patients (P=0.0316) and PCOS-like models (P=0.0079). Meanwhile, FSHR expression was positively correlated with CD4 expression in PCOS patients (P=0.0252) and PCOS-like models (P=0.0178). Furthermore, androgen receptor (AR) was identified as the common transcription factor for STON1 and FSHR and positively correlated with the expression of STON1 (P=0.039) and FSHR (P=4e-06) in ovary tissues and PCOS-like mice. Conclusion: Overall, we identified STON1 and FSHR as potential targets for the rs13405728 locus and their roles in the processes of adipocyte metabolism and CD4 immune expression in PCOS, which provides 3D genomic insight into the pathogenesis of PCOS.


Subject(s)
Membrane Proteins/genetics , Polycystic Ovary Syndrome/genetics , Receptors, FSH/genetics , Transcription Factors, General/genetics , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , CD4 Antigens/immunology , Female , Gene Expression , Genetic Loci , Genome , Humans , Membrane Proteins/metabolism , Mice, Inbred C57BL , Ovary/metabolism , Polycystic Ovary Syndrome/immunology , Polycystic Ovary Syndrome/metabolism , Receptors, Androgen/genetics , Receptors, FSH/immunology , Transcription Factors, General/metabolism
11.
Fertil Steril ; 116(4): 1139-1146, 2021 10.
Article in English | MEDLINE | ID: mdl-34119324

ABSTRACT

OBJECTIVE: To study the relationship between circulating chemokine cysteine-cysteine motif ligand (CCL) 5 levels and cysteine-cysteine chemokine receptor type 5 (CCR5) expression in peripheral blood mononuclear cells (PBMCs) and adipose tissue with hyperandrogenism and insulin resistance in patients with polycystic ovary syndrome (PCOS). DESIGN: Case-control study. SETTING: University teaching hospital. PATIENT(S): Fifteen women with PCOS and 15 controls matched for body mass index and age were enrolled in this study. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Plasma levels of CCL3, CCL4, and CCL5 were determined using enzyme-linked immunosorbent assay kits, and omental adipose tissue and PBMCs were analyzed using real-time polymerase chain reaction to determine the expression level of CCR5 in participants. RESULT(S): Levels of CCL5 were significantly higher in women with PCOS. Expression of CCR5 in adipose tissue and PBMCs was significantly higher in women with PCOS compared with that in women in the control group. Cysteine-cysteine chemokine receptor type 5 expression also was upregulated in THP-1 cells after chronic exposure to testosterone. Levels of CCL5 had a significant positive correlation with testosterone levels in women with PCOS. Moreover, CCR5 showed a positive correlation with fasting glucose levels, homeostasis model insulin resistance index, and C-reactive protein. CONCLUSION(S): Increased levels of CCL5 and overexpression of CCR5 in PBMCs and adipose tissue are associated with hyperandrogenism and insulin resistance in women with PCOS. Additionally, CCR5 and CCL5 may be used as biomarkers in the pathogenesis of PCOS.


Subject(s)
Abdominal Fat/metabolism , Chemokine CCL5/metabolism , Hyperandrogenism/metabolism , Insulin Resistance , Leukocytes, Mononuclear/metabolism , Polycystic Ovary Syndrome/metabolism , Receptors, CCR5/metabolism , T-Lymphocytes/metabolism , Testosterone/blood , Abdominal Fat/immunology , Abdominal Fat/physiopathology , Adult , Blood Glucose/metabolism , C-Reactive Protein/analysis , Case-Control Studies , Female , Humans , Hyperandrogenism/diagnosis , Hyperandrogenism/immunology , Hyperandrogenism/physiopathology , Insulin/blood , Leukocytes, Mononuclear/immunology , Lymphocyte Activation , Omentum , Polycystic Ovary Syndrome/diagnosis , Polycystic Ovary Syndrome/immunology , Polycystic Ovary Syndrome/physiopathology , Receptors, CCR5/genetics , T-Lymphocytes/immunology , THP-1 Cells , Up-Regulation , Young Adult
12.
J Reprod Immunol ; 146: 103345, 2021 08.
Article in English | MEDLINE | ID: mdl-34116484

ABSTRACT

Polycystic Ovary Syndrome (PCOS), a major endocrine disorder, affects the reproductive function of a woman, along with an association with metabolic conditions like insulin resistance and inflammation. The inflammatory nature of PCOS is much debated over, owing to numerous cases of elevation in cytokine levels. Studies have shown the beneficiary effect of Gamma-Linolenic acid (GLA) in reducing inflammation related to many conditions such as atopic dermatitis, rheumatoid arthritis, arterial disease, obesity, and even PCOS. The study aims at assessing the expression of inflammatory cytokines in the ovary and Peri-ovarian adipose tissue (POAT) of the Dehydroepiandrosterone (DHEA) induced PCOS rat model. Further, this study also evaluates the effect of γ-linolenic Acid (GLA) on these cytokines in POAT. Female Wistar rats were subcutaneously injected with 60 mg/kg DHEA daily for 28 days. These PCOS-induced rats were then orally administered with 50 mg/kg GLA for 14 days. The gene expression of cytokines was assessed by Real Time-PCR. The study showed an increase in the expression of cytokines in the ovary and POAT of the DHEA group. This suggests the role of ovarian adipose in adding to the pro-inflammatory state of PCOS. Moreover, the administration of GLA to the PCOS-induced rats resulted in a reduction of cytokine expression from the POAT, indicating that the compound was successful in reducing the associated inflammation. The study throws light on the possibility of using GLA as a supplementary or naturalistic alternative in ameliorating ovarian adipose-associated inflammation that accompanies PCOS.


Subject(s)
Adipose Tissue/drug effects , Cytokines/metabolism , Polycystic Ovary Syndrome/immunology , gamma-Linolenic Acid/pharmacology , Adipose Tissue/immunology , Adipose Tissue/pathology , Animals , Disease Models, Animal , Female , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Ovary/drug effects , Ovary/immunology , Ovary/pathology , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/pathology , Rats , gamma-Linolenic Acid/therapeutic use
13.
Biol Reprod ; 105(2): 427-438, 2021 08 03.
Article in English | MEDLINE | ID: mdl-33959757

ABSTRACT

Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disease. It has been reported that chronic low-grade inflammation might participate in its pathogenesis. C1q and TNF related 6 (C1QTNF6) is a newly identified adiponectin paralog associated with inflammation. The aim of the present study was to investigate the role of C1QTNF6 in the development of chronic inflammation in PCOS and the underlying molecular mechanism. After analyzing the expression of C1QTNF6 in the serum and granulosa cells (GCs) of PCOS patients and healthy controls, we verified the roles of C1QTNF6 in inflammation through dehydroepiandrosterone-induced PCOS mouse models and cell models of lipopolysaccharide (LPS)-induced inflammation. The results demonstrated that C1QTNF6 expression in the serum and GCs of patients with PCOS was significantly elevated compared with those of the controls, and similar results were observed in the serum and ovary of PCOS mouse models. In PCOS mice and C1QTNF6-overexpressing PCOS mice, serum levels of pro-inflammatory factors including C-reactive protein (CRP), interleukin 6 (IL6), and tumor necrosis factor-α (TNFα) were increased, while the opposite effects were observed when C1QTNF6 was down-regulated in PCOS mice. Furthermore, C1QTNF6 overexpression up-regulated the levels of TNFα, IL6, and CRP and activated the AKT/NF-κB pathway in LPS-treated KGN cells, whereas C1QTNF6 knockdown and BAY-117082 (an NF-κB inhibitor) treatment resulted in the opposite effects. Taken together, our results indicate that C1QTNF6 is involved in the pathogenesis of PCOS by affecting the inflammatory response via the AKT/NF-κB signaling pathway.


Subject(s)
Collagen/genetics , Inflammation/genetics , Polycystic Ovary Syndrome/immunology , Animals , Collagen/metabolism , Female , Granulosa Cells/pathology , Humans , Mice , Polycystic Ovary Syndrome/genetics
14.
Nutrients ; 13(3)2021 Mar 10.
Article in English | MEDLINE | ID: mdl-33801995

ABSTRACT

The impact of diet on inflammation and oxidative stress (OS) in girls with polycystic ovary syndrome (PCOS) is unknown. Therefore, our study aimed to investigate, in PCOS girls, whether certain macronutrient intakes can be associated with these disturbances. For this purpose, 59 PCOS participants (aged 14-18 years) were recruited to this study and divided into two subgroups: overweight/obese-Ov/Ob group (n = 22) and normal weight-N group (n = 37). Nutrition was assessed using a 3-day food record. The studied markers were total antioxidant capacity (TAC), malondialdehyde (MDA), C-reactive protein (CRP), tumor necrosis factor α (TNF-α), and interleukins 1 and 6 (IL-1 and IL-6). We found plant protein intake inversely correlated with IL-6 (p = 0.007; r = -0.557), TNF-α (p = 0.006; r = -0.564), MDA (p = 0.01; r = -0.539) in the Ov/Ob group and with TAC (p = 0.021; r = -0.38) in the N group. Inverse correlations in the Ov/Ob group were observed between protein intake and IL-6 (p = 0.031; r = -0.461), TNF- α (p = 0.043; r = -0.435); carbohydrates and IL-6 (p = 0.037; r = -0.448), MDA (p = 0.045; r = -0.431); fiber and IL-6 (p = 0.025; r = -0.475). A positive relationship between cholesterol intake and CRP concentration (p = 0.038; r = 0.342) was also found in the N group. These findings revealed that inflammation and OS are increased in Ov/Ob girls with decreased plant protein intake and low carbohydrates in the diet. Moreover, inflammation may be increased by cholesterol intake in slim PCOS girls. On the other hand, decreased intake of fiber and total protein intake increased inflammation. ClinicalTrials.gov Identifier: NCT04738409.


Subject(s)
Eating , Inflammation/complications , Obesity/complications , Overweight/complications , Oxidative Stress , Polycystic Ovary Syndrome/physiopathology , Adolescent , Antioxidants/analysis , Body Mass Index , C-Reactive Protein/analysis , Cytokines/blood , Diet Records , Dietary Fats/administration & dosage , Dietary Proteins/administration & dosage , Female , Humans , Malondialdehyde/blood , Obesity/physiopathology , Overweight/physiopathology , Plant Proteins, Dietary/administration & dosage , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/immunology
15.
PLoS One ; 16(4): e0249639, 2021.
Article in English | MEDLINE | ID: mdl-33798258

ABSTRACT

CONTEXT: Polycystic ovarian syndrome (PCOS) is a complex disease with different subtypes and unclear etiology. Among the frequent comorbidities are autoimmune diseases, suggesting that autoantibodies (aAb) may be involved in PCOS pathogenesis. OBJECTIVE: As the gonadal axis often is dysregulated, we tested the hypothesis that aAb to the gonadotropin-releasing hormone receptor (GnRH-R) are of diagnostic value in PCOS. DESIGN: An in vitro assay for quantifying aAb to the GnRH-R (GnRH-R-aAb) was established by using a recombinant fusion protein of full-length human GnRH-R and firefly luciferase. A commercial rabbit antiserum to human GnRH-R was used for standardization. Serum samples of control subjects and different cohorts of European PCOS patients (n = 1051) were analyzed. RESULTS: The novel GnRH-R-aAb assay was sensitive, and signals were linear on dilution when tested with the commercial GnRH-R antiserum. Natural GnRH-R-aAb were detected in one control (0.25%) and two PCOS samples (0.31%), and 12 samples were slightly above the threshold of positivity. The identification of samples with positive GnRH-R-aAb was reproducible and the signals showed no matrix interferences. CONCLUSION: Natural GnRH-R-aAb are present in a very small fraction of adult control and PCOS subjects of European decent. Our results do not support the hypothesis that the GnRH-R constitutes a relevant autoantigen in PCOS.


Subject(s)
Autoantibodies/blood , Biomarkers/blood , Polycystic Ovary Syndrome/diagnosis , Receptors, LHRH/immunology , Adult , Autoantibodies/immunology , Case-Control Studies , Female , Humans , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/immunology , Young Adult
16.
Front Endocrinol (Lausanne) ; 12: 638621, 2021.
Article in English | MEDLINE | ID: mdl-33716989

ABSTRACT

Background: Women with polycystic ovary syndrome (PCOS) often have vitamin D deficiency, a known risk factor for severe COVID-19 disease. Alveolar macrophage-derived cytokines contribute to the inflammation underlying pulmonary disease in COVID-19. We sought to determine if basal macrophage activation, as a risk factor for COVID-19 infection, was present in PCOS and, if so, was further enhanced by vitamin D deficiency. Methods: A cross-sectional study in 99 PCOS and 68 control women who presented sequentially. Plasma levels of a macrophage-derived cytokine panel were determined by Slow Off-rate Modified Aptamer (SOMA)-scan plasma protein measurement. Vitamin D was measured by tandem mass spectroscopy. Results: Vitamin D was lower in PCOS women (p<0.0001) and correlated negatively with body mass index (BMI) in PCOS (r=0.28, p=0.0046). Basal macrophage activation markers CXCL5, CD163 and MMP9 were elevated, whilst protective CD200 was decreased (p<0.05); changes in these variables were related to, and fully accounted for, by BMI. PCOS and control women were then stratified according to vitamin D concentration. Vitamin D deficiency was associated with decreased CD80 and IFN-γ in PCOS and IL-12 in both groups (p<0.05). These factors, important in initiating and maintaining the immune response, were again accounted for by BMI. Conclusion: Basal macrophage activation was higher in PCOS with macrophage changes related with increased infection risk associating with vitamin D; all changes were BMI dependent, suggesting that obese PCOS with vitamin D deficiency may be at greater risk of more severe COVID-19 infection, but that it is obesity-related rather than an independent PCOS factor.


Subject(s)
COVID-19/epidemiology , Cytokines/metabolism , Macrophages/metabolism , Polycystic Ovary Syndrome/epidemiology , Vitamin D Deficiency/epidemiology , Adult , Biomarkers/analysis , Blood Proteins/analysis , Body Mass Index , COVID-19/complications , COVID-19/immunology , Cross-Sectional Studies , Female , Humans , Macrophage Activation , Macrophages/chemistry , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/immunology , Risk Assessment , Tandem Mass Spectrometry , Vitamin D/blood , Vitamin D Deficiency/complications
17.
J Reprod Immunol ; 144: 103289, 2021 04.
Article in English | MEDLINE | ID: mdl-33610928

ABSTRACT

Polycystic ovary syndrome (PCOS) is the most common cause of ovulatory infertility. Inflammation may be involved in the pathogenesis and development of PCOS. We investigated the anti-inflammatory effect of minocycline on TNF-α, TNFR2, and TLR4 expression levels and the key features of PCOS in a mouse model. Molecular docking was performed by Molecular Operating Environment software. PCOS was induced by estradiol valerate injection (EV) (2 mg/kg/day) in 40 mice. After 28 days, the mice were divided into five groups, including control, PCOS, minocycline control, minocycline PCOS model (50 mg/kg), and letrozole PCOS (0.5 mg/kg). The Levels of FSH, LH, E2, and testosterone were determined by ELISA. H&E staining was used for histological analysis in the ovarian tissues. Docking scores were -10.35, -10.57, and -12.45 kcal/mol for TNFα, TLR-4, and TNFR2, respectively. The expression levels of TNF-α, TNFR2, and TLR4 were detected by Real-Time PCR. PCOS models exhibited acyclicity, a significant increase in E2 levels (P < 0.01), and no difference in FSH, LH, and testosterone. The expression levels of TNF-α, TNFR2, and TLR-4 significantly increased in PCOS (2.70, 7.90, and 14.83-fold, respectively). EV treatment significantly increased graafian follicles (P < 0.001) and decreased corpus luteum (CL) (P < 0.01). Minocycline treatment in PCOS led to a significant decrease in E2 (P < 0.01) and graafian follicles (P < 0.001) and a significant increase in the CL numbers (P < 0.05). Our findings showed the positive effects of minocycline on estradiol level, CL and graafian follicles counts, suggesting that minocycline might inhibit these proteins and improve ovulation in our mouse model of PCOS.


Subject(s)
Minocycline/pharmacology , Ovulation/drug effects , Polycystic Ovary Syndrome/drug therapy , Animals , Disease Models, Animal , Estradiol/administration & dosage , Estradiol/toxicity , Female , Humans , Letrozole/pharmacology , Letrozole/therapeutic use , Mice , Minocycline/therapeutic use , Molecular Docking Simulation , Ovary/immunology , Ovary/pathology , Ovulation/immunology , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/immunology , Polycystic Ovary Syndrome/pathology , Receptors, Tumor Necrosis Factor, Type II/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Type II/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
18.
J Reprod Immunol ; 144: 103282, 2021 04.
Article in English | MEDLINE | ID: mdl-33607547

ABSTRACT

Polycystic ovary syndrome (PCOS) is described as a low-grade chronic inflammatory state. However, there are limited studies on the specific endometrial immune status of PCOS patients. Whether this endometrial immune cell pattern is intrinsic to PCOS or the consequence of PCOS-associated obesity is a subject of debate. This study retrospectively included one hundred women diagnosed with PCOS and ninety-five normal fertile controls, which further divided into four groups (normoweight PCOS; overweight PCOS; normoweight control; overweight control) based on body mass index. The percentages of endometrial CD68+ macrophages (1.97 % vs. 1.17 %; P < 0.001), CD163+ M2 macrophages (2.30 % vs. 1.83 %; P = 0.001), CD1a+ iDCs (0.044 % vs. 0.029 %; P = 0.002), CD83+ mDCs (1.72 % vs. 1.07 %; P < 0.001) and CD8+ T cells (2.82 % vs. 1.95 %; P < 0.001) were significantly higher in normoweight PCOS women than normoweight controls. The percentage of CD68+ macrophages (2.09 % vs. 1.15 %; P < 0.001) was significantly higher in overweight PCOS women compared with overweight controls. In multivariant linear regression analysis, participants' PCOS status was the main predictors of endometrial CD68+ macrophages, CD163+ M2 macrophages, CD1a+ iDCs, CD83+ mDCs and CD8+ T cells in the whole study population. Additionally, in PCOS group, positive correlations were found between endometrial CD56+ NK, CD163+ M2 macrophages and QUICKI, indicating there was an association between endometrial immune cells and insulin resistance in PCOS women. Our study suggests that women with PCOS have altered endometrial immune cells, which may reflect a state of chronic low grade inflammation. The chronic inflammation, independent of obesity, may help understand the pathophysiologic mechanisms of intrinsic PCOS.


Subject(s)
Endometrium/immunology , Hyperandrogenism/immunology , Insulin Resistance/immunology , Polycystic Ovary Syndrome/complications , Adult , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , CD56 Antigen/metabolism , Case-Control Studies , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endometrium/cytology , Endometrium/pathology , Female , Healthy Volunteers , Humans , Hyperandrogenism/blood , Hyperandrogenism/diagnosis , Immunoglobulins/metabolism , Insulin/blood , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Macrophages/immunology , Macrophages/metabolism , Membrane Glycoproteins/metabolism , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/immunology , Polycystic Ovary Syndrome/pathology , Receptors, Cell Surface/metabolism , Retrospective Studies , Testosterone/blood , CD83 Antigen
19.
Article in English | MEDLINE | ID: mdl-32849259

ABSTRACT

Objective: It has been shown that women with polycystic ovary syndrome (PCOS), as well as Hashimoto's thyroiditis (HT), are characterized by increased incidence of infertility. Serum anti-Müllerian hormone (AMH), which reflects ovarian reserve, is elevated in PCOS women and is decreased in women with HT. The Rotterdam criteria recognize four clinical PCOS phenotypes, i.e., phenotypes A, B, C, and D. The aim of the present study was to investigate the relation between serum concentrations of thyroid peroxidase antibodies (TPOAbs) and ovarian reserve in different PCOS phenotypes. Patients and methods: We examined 141 women with PCOS [phenotype A was diagnosed in 67 (47.5%) women, phenotype B in 30 (21.3%), phenotype C in 28 (19.9%), and phenotype D in 16 (11.3%)] and 88 control subjects of similar age; all women were euthyroid. Serum concentrations of AMH, thyroid-stimulating hormone (TSH), thyroid hormones, and TPOAbs were assessed. Results: We observed positive serum TPOAbs in 21.9% women with PCOS and in 23.9% controls (p = 0.07). We did not find differences in the frequency of detection of positive serum TPOAbs between phenotypes A, B, and C and the control group (p > 0.05). We did not observe a difference in AMH levels between TPOAbs-positive and TPOAbs-negative women, both in the control group and the PCOS women (all p > 0.05). However, serum AMH concentration was markedly higher in the whole PCOS group (p < 0.01) and in phenotype A (p < 0.01) vs. controls when the serum concentration of TPOAbs was negative. In the groups with positive serum levels of TPOAbs, serum concentration of AMH did not differ between PCOS phenotypes and controls (p = 0.23). Additionally, we observed that serum AMH concentration was related to the level of TPOAbs in the PCOS group (r = -0.4, p = 0.02). Conclusions: The frequency of serum detection of positive TPOAbs did not differ between PCOS phenotypes with clinical/biochemical hyperandrogenism and the control group. The observation of the difference in serum AMH between the PCOS and control groups only in TPOAbs negative women together with the inverse relation of TPOAbs with serum AMH only in the PCOS group might suggest that ovarian reserve is influenced by TPOAbs in PCOS.


Subject(s)
Autoantibodies/blood , Iodide Peroxidase/immunology , Ovarian Reserve , Polycystic Ovary Syndrome/pathology , Thyroid Gland/pathology , Adult , Autoantibodies/immunology , Case-Control Studies , Cross-Sectional Studies , Female , Humans , Phenotype , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/immunology , Prospective Studies , Thyroid Gland/immunology , Thyroid Gland/metabolism , Young Adult
20.
Cytokine ; 134: 155187, 2020 10.
Article in English | MEDLINE | ID: mdl-32645538

ABSTRACT

Polycystic Ovary Syndrome (PCOS) is a heterogeneous endocrinopathy considered to be the most common metabolic disorder in women of reproductive age. Women with PCOS present with an increased risk of noncommunicable diseases (NCDs), especially low-grade chronic inflammation mediated by proinflammatory cytokines, and insulin resistance. This study aimed to investigate cytokine levels and their ratios in PCOS women compared to a healthy control group. This study evaluated 97 women with PCOS and 99 healthy women as controls. The PCOS diagnosis was performed according to ESHRE/ASRM. Plasma cytokines were evaluated by flow cytometry. We observed lower TNF levels, and decreased TNF/IL-6, TNF/IL-2, and TNF/IL-4 ratios in PCOS patients compared to the control group (p < 0.05). These results indicate an imbalance between pro- and anti-inflammatory cytokines, with prominent counter-regulatory cytokine production. These changes may be important in explaining the phenotypes present in PCOS and to direct better interventions for patients with this syndrome.


Subject(s)
Cytokines/blood , Polycystic Ovary Syndrome/immunology , Adolescent , Adult , Biomarkers , Case-Control Studies , Female , Humans , Inflammation Mediators/blood , Middle Aged , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...