Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
2.
Reprod Biol ; 23(3): 100787, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37467532

ABSTRACT

Polycystic ovary syndrome (PCOS) is a complex heterogeneous disorder with reproductive and metabolic consequences whose aetiology is still elusive. To understand the cellular mechanisms that potentially govern follicular defect in women with PCOS, we performed transcriptomic profiles of granulosa-lutein cells (GLCs) by RNA-Seq analysis. We found differential expression of 876 genes in GLCs between PCOS and controls that belonged to various processes such as cell cycle, extracellular matrix organization, angiogenesis, oxidative stress, metabolism, etc. that support folliculogenesis, oocyte development, and maturation. The cross-talk between oocyte and GLCs is a fundamental cornerstone in determining oocyte quality and highly interlinked pathways of metabolism and redox homeostasis may influence this. We found several genes involved in the metabolism of carbohydrates, nucleotides, cholesterol, and lipids were dysregulated, which may impair the supply of metabolites to the growing oocyte, affecting oocyte development and competence. Additionally, high metabolic activity during folliculogenesis may augment oxidative damage to cells and macromolecules if not counter-balanced. We observed dysregulation of redox homeostasis and AGE-RAGE signalling in the follicular environment. Among the validated genes, prokineticin-1 and growth differentiation factor-15 were found to be negatively regulated, while, S100, calcium-binding protein A9 and angiomotin-like-2 were positively regulated in GLCs of women with PCOS. Comparing our data with previously published relevant transcriptomic studies showed metabolic, cytokine-cytokine receptor interaction, IL-17, and chemokine signalling pathways were most commonly affected in PCOS. Overall, this data can provide insights into mechanisms contributing to PCOS pathophysiology and can be explored as potential indicators for oocyte/embryo quality in IVF settings.


Subject(s)
Luteal Cells , Polycystic Ovary Syndrome , Transcriptome , Female , Humans , Luteal Cells/metabolism , Oocytes/metabolism , Oogenesis , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , RNA-Seq
3.
Arch Gynecol Obstet ; 306(2): 547-555, 2022 08.
Article in English | MEDLINE | ID: mdl-35477803

ABSTRACT

PURPOSE: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is global pandemic with more than 5 million deaths so far. Female reproductive tract organs express coronavirus-associated receptors and factors (SCARFs), suggesting they may be susceptible to SARS-CoV-2 infection; however, the susceptibility of ovary/follicle/oocyte to the same is still elusive. Co-morbidities like obesity, type-2 diabetes mellitus, cardiovascular disease, etc. increase the risk of SARS-CoV-2 infection. These features are common in women with polycystic ovary syndrome (PCOS), warranting further scope to study SCARFs expression in ovary of these women. MATERIALS AND METHODS: SCARFs expression in ovary and ovarian tissues of women with PCOS and healthy women was explored by analyzing publically available microarray datasets. Transcript expressions of SCARFs were investigated in mural and cumulus granulosa cells (MGCs and CGCs) from control and PCOS women undergoing in vitro fertilization (IVF). RESULTS: Microarray data revealed that ovary expresses all genes necessary for SARS-CoV-2 infection. PCOS women mostly showed down-regulated/unchanged levels of SCARFs. MGCs and CGCs from PCOS women showed lower expression of receptors ACE2, BSG and DPP4 and protease CTSB than in controls. MGCs showed lower expression of protease CTSL in PCOS than in controls. Expression of TMPRSS2 was not detected in both cell types. CONCLUSION: Human ovarian follicle may be susceptible to SARS-CoV-2 infection. Lower expression of SCARFs in PCOS indicates that the risk of SARS-CoV-2 infection to the ovary may be lesser in these women than controls. This knowledge may help in safe practices at IVF settings in the current pandemic.


Subject(s)
COVID-19 , Polycystic Ovary Syndrome , Receptors, Virus , Female , Granulosa Cells/metabolism , Humans , Peptide Hydrolases/metabolism , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , Receptors, Virus/metabolism , SARS-CoV-2
4.
Mol Biol Rep ; 49(2): 1519-1528, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34988892

ABSTRACT

Polycystic Ovarian Syndrome (PCOS) is an endocrine disorder commonly affecting the reproductive capacity of women leading to infertility. PCOS-related infertility is majorly due to anovulation; however, it is not the only cause. The defective endometrium causing recurrent miscarriage and implantation failure can also be accountable for infertility in PCOS women. The unusual levels of hormones and their receptors in the PCOS endometrium have a hostile effect during WOI, making the microenvironment unfavorable for embryo implantation. To date, many studies have been performed to determine the role of candidate genes in endometrial receptivity but very limited data is available using whole genome approach. This review aims at summarizing the existing studies on the basic aspects of endometrial receptivity in PCOS. The review focuses on aberrant levels of hormones and their receptors in the endometrium, affecting the receptivity. Additionally, it explores the novel approach reviewing the effect on treatment options administered for ovulation induction in PCOS on their endometrial receptivity. Overall, this review will help us to understand the molecular milieu in PCOS endometrium and its effect on the receptivity potential. However, to have a thorough understanding of the mechanistic approach of hormonal imbalance in PCOS on endometrial receptivity, there is a need to give more weightage to genome-wide studies in the future. The current review will further guide us to formulate future studies using whole genome technologies for the assessment of endometrial receptivity in different cohorts of PCOS women, which may have future diagnostic implementations.


Subject(s)
Endometrium/pathology , Infertility, Female/etiology , Polycystic Ovary Syndrome/physiopathology , Embryo Implantation , Endometrium/metabolism , Female , Humans , Infertility, Female/physiopathology , Ovulation Induction
5.
Reprod Sci ; 29(3): 836-848, 2022 03.
Article in English | MEDLINE | ID: mdl-34748173

ABSTRACT

The cumulus-oocyte complex (COC) matrix plays a critical role in the ovulation and fertilization process and a major predictor of oocyte quality. Proteomics studies of follicular fluid showed differential expression of COC matrix proteins in women with polycystic ovary syndrome (PCOS), indicating altered COC matrix in these women. In the present study, we aimed to understand COC matrix gene induction in humans and its probable dysfunction in women with PCOS. Animal studies have shown that amphiregulin (AREG) and growth differentiation factor-9 (GDF-9) are important in the induction of COC matrix genes which are involved in cumulus expansion. The effects of AREG and GDF-9 on expression of tumor necrosis factor alpha induced protein 6 (TNFAIP6) and hyaluronan synthase 2 (HAS2) on human cumulus granulosa cells (CGCs) and murine COC expansion were evaluated. Further time-dependent effects of growth factor supplementation on these gene expressions in CGCs from PCOS and control women were compared. Follicular fluid from PCOS showed reduced COC matrix expansion capacity, using murine COCs. Expression of COC matrix genes TNFAIP6 and HAS2 were significantly reduced in CGCs of PCOS. Treatment of CGCs with AREG and GDF-9 together induced expression of both these genes in controls and could only restore HAS2 but not TNFAIP6 expression in PCOS. Our results suggest that the reduced potential of follicular fluid to support COC expansion, altered expression of structural constituents, and intrinsic defects in granulosa cells of women with PCOS may contribute to the aberrant COC organization and expansion in PCOS, thus affecting fertilization.


Subject(s)
Cumulus Cells/metabolism , Oocytes/metabolism , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , Adult , Amphiregulin/metabolism , Animals , Female , Fertilization in Vitro , Gene Expression , Growth Differentiation Factor 9/metabolism , Humans , Hyaluronan Synthases/metabolism , Mice
6.
Reprod Sci ; 29(2): 513-523, 2022 02.
Article in English | MEDLINE | ID: mdl-34410653

ABSTRACT

Recurrent implantation failure (RIF) is one of the major obstacles in IVF. Transcriptomic literature has revealed the various biological processes involved in endometrial receptivity (ER) under different physiological circumstances, especially in natural cycle. We intended to determine the function-specific ER profile under controlled ovarian stimulation (COS) cycle. This can help to back trace the genomic impairment in RIF patients during the IVF cycle and to validate the genes involved in enriched pathways. In our study, retrospective gene expression microarray dataset was reanalysed after the follow-up, in classic non pregnant RIF (cases) vs fertile women (controls) under COS (n = 5/group). Reanalysis of microarray revealed significant downregulation of cell adhesion function (P:3.11E-05) with the maximum gene count. For validation purpose, downregulation of eight genes (COMP, HABP2, ITGAD, CDH3, COL22A1, MFAP4, THBS1and CD300A) involved in enriched cell adhesion pathway having fold change > 3 were assessed by real-time PCR in independent cohorts of cases and controls (n = 24, each). Downregulation of six out of eight genes (COMP, HABP2, ITGAD, CDH3, MFAP4 and THBS1) were confirmed by real-time PCR (P < 0.05) with fold change > 2. This indicates the importance of analysed genes in the ER mechanism under COS, thus mimicking the fresh embryo transfer. The further analysis in larger cohorts would substantiate the study findings in RIF patients undergoing IVF cycle.


Subject(s)
Cell Adhesion Molecules/metabolism , Embryo Implantation , Endometrium/metabolism , Fertilization in Vitro , Adult , Case-Control Studies , Embryo Transfer , Female , Humans , Oligonucleotide Array Sequence Analysis , Ovulation Induction , Real-Time Polymerase Chain Reaction , Transcriptome , Treatment Failure
7.
J Assist Reprod Genet ; 38(10): 2609-2623, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34041658

ABSTRACT

BACKGROUND: Women with polycystic ovary syndrome (PCOS) are often infertile and opt for artificial reproductive techniques (ART) to conceive. Disrupted pro-/antioxidant balance in oocyte microenvironment may contribute towards sub-optimal oocyte/embryo quality and poor ART outcome in them. METHODS: Activities/levels of redox markers and their transcript expression were investigated in follicular fluid and granulosa cells respectively, in women with PCOS (n = 71) and controls (n = 50) undergoing in vitro fertilization (IVF). Correlation analysis of redox markers and IVF parameters was performed. RESULTS: Activities of superoxide dismutase, glutathione reductase, glutathione peroxidase, and paraoxonase1 were significantly lower in follicular fluid of PCOS women than in controls. Levels of lipid peroxidation, oxidative protein modification, and oxidized glutathione were higher, whereas those of total antioxidant capacity, total thiols, and reduced glutathione were lower in follicular fluid of PCOS women than in controls. Further, comparison of redox markers based on insulin resistance and BMI status of study participants showed similar trends, indicating that PCOS pathophysiology is a significant contributor to oxidative stress irrespective of insulin resistance and BMI. Transcript levels of antioxidant enzymes were lower in granulosa cells from PCOS women than in controls, and they accorded with their activities in follicular fluid. Moreover, few redox markers showed significant correlations with oocyte/embryo quality and pregnancy outcome. CONCLUSION: Our data indicates disrupted redox homeostasis in follicular environment in PCOS which may negatively influence oocyte/embryo quality. Further, granulosa cells may play crucial role in maintaining follicular redox homeostasis. Glutathione system and paraoxonase1 could be explored further as surrogates for IVF prognosis/outcome.


Subject(s)
Biomarkers/metabolism , Follicular Fluid/chemistry , Granulosa Cells/pathology , Infertility, Female/pathology , Ovarian Follicle/pathology , Polycystic Ovary Syndrome/physiopathology , Pregnancy Outcome , Adult , Case-Control Studies , Female , Fertilization in Vitro/methods , Follicular Fluid/metabolism , Granulosa Cells/metabolism , Humans , Infertility, Female/etiology , Infertility, Female/metabolism , Ovarian Follicle/metabolism , Oxidation-Reduction , Pregnancy
9.
Hum Reprod ; 36(4): 1052-1064, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33377483

ABSTRACT

STUDY QUESTION: Is angiogenic potential of follicular fluid (FF) and granulosa-lutein cells (GLCs) altered in polycystic ovary syndrome (PCOS) and does it play a role in corpus luteum (CL) defect observed in them? SUMMARY ANSWER: FF and GLCs of women with PCOS show reduced expression of pro-angiogenic factors compared to controls and exhibit a diminished capacity to induce angiogenesis. WHAT IS KNOWN ALREADY: In women with PCOS, CL insufficiency and frequent miscarriage are reported, which may be due to defect in CL. The development of new blood vessels is essential to promote ovarian folliculogenesis and functional CL formation. The vasculature formation in CL which is important for its function is still unexplored in these women. STUDY DESIGN, SIZE, DURATION: This case-control study was conducted in 30 healthy control women and 30 women with PCOS undergoing controlled ovarian hyperstimulation for IVF. The FF, GLCs and serum were collected from all participants during ovum pick up. PARTICIPANTS/MATERIALS, SETTING, METHODS: The capacity of FF to induce angiogenesis was assessed by measuring levels of pro-angiogenic factors vascular endothelial growth factor (VEGF) and fibroblast growth factor 2 (FGF2) and its tube formation and wound healing potential using human umbilical vein endothelial cells (HUVECs). We investigated the angiogenic potential and endothelial cell-like nature of GLCs using several approaches such as the expression of angiogenic genes by quantitative PCR, DiI-conjugated acetylated low-density lipoproteins (Dil-Ac-LDL) internalization assay, tube formation assay, expression of endothelial cell markers by immunofluorescence analysis. In addition, correlation of transcript levels of angiogenic genes with oocyte parameters was studied. MAIN RESULTS AND THE ROLE OF CHANCE: FF and serum levels of VEGF and FGF2 were significantly higher and lower, respectively, in PCOS compared to controls. The tube formation and wound healing capacity of HUVECs was found to be reduced when measured after supplementation with FF of women with PCOS compared to controls. This suggests a decreased angiogenic capacity of FF in women with PCOS. Tube formation (P = 0.003) and Dil-Ac-LDL internalization (P = 0.03) ability of GLCs were significantly reduced in women with PCOS compared to controls. Protein expression levels of endothelial markers, vascular endothelial growth factor A (VEGFA) (P = 0.004), vascular endothelial growth factor receptor 2 (VEGFR2) (P = 0.011), TEK Receptor Tyrosine Kinase (Tie-2) (P = 0.026), fibroblast growth factor receptor 1 (FGFR1) (P = 0.026) and CD31 (P = 0.035) and transcript levels of angiogenic genes VEGFA (P = 0.042), hypoxia inducing factor 1A (HIF1A) (P = 0.025), FGF2 (P = 0.038), angiopoietin 1 (ANGPT1) (P = 0.028), heparin sulfate proteoglycan 2 (HSPG2) (P = 0.016), ADAM metallopeptidase with thrombospondin type1 motif, 1 (ADAMTS1) (P = 0.027) and fibronectin 1 (FN1) (P = 0.016) were found to be low in GLCs of PCOS compared to controls. Thus, the findings of this study indicate that endothelial cell-like characteristics of GLCs were significantly decreased in PCOS. Furthermore, transcript levels of VEGFA (r = 0.46, P = 0.009), ADAMTS1 (r = 0.55, P = 0.001), FGF2 (r = 0.42, P = 0.022) and ANGPT2 (r = 0.47, P = 0.008) showed a positive correlation with oocyte fertilization rate. LIMITATIONS, REASONS FOR CAUTION: The vasculature formation in CL is not possible to study in women, but we explored the angiogenic characteristics of FF and GLC obtained from women with PCOS to speculate any vascularization defect of CL in these women. The FF and GLCs were obtained from the stimulated cycle during oocyte retrieval, which may not exactly mimic the in-vivo condition. The small sample size is another limitation of this study. Larger sample size and support by color Doppler studies on CL blood flow would help to strengthen our findings. WIDER IMPLICATIONS OF THE FINDINGS: Our findings suggest that the altered angiogenic potential of FF and GLCs may affect vasculature development required for CL formation and function in PCOS. These findings pave the way to devise therapeutic strategies to support angiogenesis process in follicle of women with PCOS, which may improve CL insufficiency, progesterone levels and prevent frequent miscarriages in these women. Furthermore, our study also hypothesizes that the vascularization around the ovarian follicles is also compromised which may lead to the growth arrest of the follicles in PCOS, however, this needs thorough investigations. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by Grant BT/PR16524/MED/97/346/2016 from the Department of Biotechnology, Government of India. The authors have no conflicts of interest to disclose. TRIAL REGISTRATION NUMBER: N/A.


Subject(s)
Luteal Cells , Polycystic Ovary Syndrome , Case-Control Studies , Endothelial Cells , Female , Follicular Fluid , Humans , India , Vascular Endothelial Growth Factor A
10.
Am J Reprod Immunol ; 83(1): e13196, 2020 01.
Article in English | MEDLINE | ID: mdl-31595580

ABSTRACT

PROBLEM: DNA methylation profile in mid-secretory phase of endometrium is reported to be varied from other phases in natural menstrual cycle. Therefore, we intended to study the impairment in endometrial receptivity by performing whole-genome methylation and gene expression profiling in endometrium of recurrent implantation failure patients (RIF) during IVF under controlled ovarian stimulation (COS). METHOD OF STUDY: Endometrial biopsies were collected from IVF-RIF patients (cases, n = 6) and healthy fertile oocyte donors (controls, n = 6) undergoing COS after 6/7th day of human chorionic gonadotropin administration. The whole-genome methylation and gene expression microarray were performed and analysed by GenomeStudio software (P < .05 by Illumina Custom Model), whereas the enrichment analysis was performed using "Database for Annotation, Visualization and Integrated Discovery" (DAVID, V6.8). Significant differentially methylated genes were correlated with dys-regulated genes using Pearson's correlation. RESULTS: Differential methylation in RIF patients revealed 448 CpG sites. The enrichment analysis showed aberrant methylation in genes involved in immunological response and G protein activity. Methylation in NLRP2 gene in inflammatory pathway had significant negative correlation with gene expression (P = .008), whereas SERPINA5 gene that is already known to be involved in endometrial receptivity was observed to be hypomethylated in promoter region with highest delta beta value and up-regulated in gene expression analysis. CONCLUSION: The aberrant methylation of genes involved in immunological functions and G protein activation was found to be prevalent which might suggest a role in endometrial receptivity. However, the findings need to be further validated on a larger cohort of IVF-RIF patients.


Subject(s)
DNA Methylation , Embryo Implantation/genetics , Embryo Implantation/immunology , Endometrium/physiology , Fertilization in Vitro , Adult , Chorionic Gonadotropin/therapeutic use , Female , Humans , Transcriptome , Treatment Failure , Young Adult
11.
J Assist Reprod Genet ; 36(8): 1759-1760, 2019 08.
Article in English | MEDLINE | ID: mdl-31273586

ABSTRACT

In the original Fig 1, we had inadvertently placed the DAPI channel image of the HSP90α as negative control.

12.
PLoS One ; 14(4): e0214742, 2019.
Article in English | MEDLINE | ID: mdl-30946770

ABSTRACT

Polycystic ovary syndrome (PCOS) is a complex endocrinopathy affecting women of reproductive age, and whose etiology is not well understood yet. In these women, the follicular growth is arrested at preantral stage leading to cyst formation, consequently resulting in anovulatory infertility in these women. As the follicular fluid provides the conducive microenvironment for the growth of oocytes, molecular profiling of the fluid may provide unique information about pathophysiology associated with follicular development in PCOS. Post-translational addition of oligosaccharide residues is one of the many modifications of secreted proteins influencing their functions. These glycoproteins play a significant role in disease pathology. Despite glycoproteins having such essential functions, very limited information is available on their profiling in human reproductive system, and glycoproteomic profile of follicular fluid of women with PCOS is yet unexplored. In the present study, we performed a comparative glycoproteomic analysis of follicular fluid between women with PCOS and controls undergoing in vitro fertilization, by enrichment of glycoproteins using three different lectins viz. concanavalin A, wheat germ agglutinin and Jacalin. Peptides generated by trypsin digestion were labeled with isobaric tags for relative and absolute quantification reagents and analyzed by liquid chromatography tandem mass spectrometry. We identified 10 differentially expressed glycoproteins, in the follicular fluid of women with PCOS compared to controls. Two important differentially expressed proteins- SERPINA1 and ITIH4, were consistently upregulated and downregulated respectively, upon validation by immunoblotting in follicular fluid and real-time polymerase chain reaction in granulosa cells. These proteins play a role in angiogenesis and extracellular matrix stabilization, vital for follicle maturation. In conclusion, a comparative glycoproteomic profiling of follicular fluid from women with PCOS and controls revealed an altered expression of proteins which may contribute to the defects in follicle development in PCOS pathophysiology.


Subject(s)
Follicular Fluid/metabolism , Glycoproteins/metabolism , Polycystic Ovary Syndrome/metabolism , Adult , Female , Gene Expression Profiling , Humans , Mass Spectrometry/methods , Ovarian Follicle/metabolism , Proteomics
13.
Indian J Med Res ; 147(6): 552-559, 2018 06.
Article in English | MEDLINE | ID: mdl-30168486

ABSTRACT

Background & objectives: CD9 and CD146 are important adhesion molecules that play a role in the implantation of an embryo. This study was undertaken to correlate the expression of these markers in fertile and infertile women's endometrial stromal cells. Methods: Human endometrial stromal cell culture from endometrial biopsies of fertile (n=50) and infertile females (n=50) was performed and primary cell lines were established. Expression of CD9 and CD146 was studied for all the 100 cell lines with the help of flow cytometry. Gene expression of CD9 and CD146 was performed by real-time polymerase chain reaction. Results: There was a significant difference in endometrial stromal cells of fertile and infertile females. Flow cytometric results revealed significantly lower expression of CD9 (P=0.0126) and CD146 (P=0.0006) in the infertile endometrial stromal cells as compared to fertile endometrial stromal cells. These results were comparable with real-time data. Interpretation & conclusions: This study showed that endometrial stromal cells from infertile females had lower expression of adhesion molecules, CD9 and CD146. Our findings suggest that CD9 and CD146 may have a role in infertility. Infertile female's endometrial stromal cells have decreased expression of CD9 and CD146 which can be the cause of infertility related to implantation failure.


Subject(s)
CD146 Antigen/analysis , Infertility, Female/diagnosis , Stromal Cells/immunology , Tetraspanin 29/analysis , Adult , Animals , Endometrium/immunology , Female , Fertility , Humans , India , Mice , Pregnancy , Young Adult
14.
Am J Reprod Immunol ; 78(1)2017 07.
Article in English | MEDLINE | ID: mdl-28370781

ABSTRACT

PROBLEM: Implantation failure (IF) even after the good-quality embryo transfer (ET) is main obstacle in in vitro fertilization (IVF). We aim to study the genomics of endometrial receptivity in IF patients under controlled ovarian stimulation (COS) during which ET is generally practised in IVF. METHOD OF STUDY: Endometrial gene expression profiling in IF patients (n=10) and oocyte donors (n=8) were compared during window of implantation under COS by microarray. Enrichment analysis of microarray data was performed to determine dysregulated pathways. Microarray results were validated by real-time PCR. Localization of genes related to immune response (progestagen-associated endometrial protein (PAEP), leukaemia inhibitory factor (LIF), interleukin-6 signal transducer (IL6ST) was detected by immunohistochemistry. RESULTS: The gene ontology, pathway analysis and enrichment mapping revealed significant downregulation in activation and regulation of immune and inflammation response in IF patients under COS. The lower expression of PAEP, LIF and IL6ST in cases compared to controls by real time and immunohistochemistry suggests the functional importance of these genes. CONCLUSION: Importance of immune and inflammatory response in endometrial receptivity adds on to the current knowledge of gene expression profile in IF under COS. The panel of genes involved in these pathways would be useful in determining further line of treatment for IF during IVF.


Subject(s)
Embryo Implantation/genetics , Embryo Implantation/immunology , Fertilization in Vitro , Adult , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/immunology , Cytokine Receptor gp130/metabolism , Down-Regulation , Endometrium/immunology , Endometrium/metabolism , Female , Gene Expression Profiling , Glycodelin/genetics , Glycodelin/immunology , Glycodelin/metabolism , Humans , Inflammation/genetics , Inflammation/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-6/metabolism , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/immunology , Leukemia Inhibitory Factor/metabolism , Oocytes/immunology , Oocytes/metabolism , Ovary/immunology , Ovary/metabolism , Young Adult
15.
J Assist Reprod Genet ; 34(4): 495-503, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28236106

ABSTRACT

PURPOSE: The aims of this paper were to study whether heat shock protein 90 (HSP90) is a regulator of sperm functions and to determine its association with oligoasthenozoospermia. METHODS: The levels of HSP90 in sperm lysates were measured by ELISA. Localization of HSP90 and its isoforms was evaluated by immunofluorescence. Sperm motility and kinetics were assessed by computer-assisted sperm analysis. Acrosome reaction was determined by lectin staining. RESULTS: The levels of HSP90 were lower in oligoasthenozoospermic men and correlated positively with the number of motile spermatozoa. In capacitated human spermatozoa, HSP90α was mostly found in residual nuclear envelope, and the HSP90ß isoform was higher in the flagella. Inhibition of HSP90 by geldanamycin or 17-AAG did not affect basal motility, but suppressed progesterone-mediated forward progressive motility, hyperactivation and acrosome reaction. Progesterone treatment dephosphorylated both HSP90α and HSP90ß at Ser/Thr-Pro residues, but not Tyr residues. CONCLUSION: HSP90 levels are downregulated in oligoasthenozoospermia, and its functional inhibition attenuates progesterone-mediated sperm motility and acrosome reaction.


Subject(s)
HSP90 Heat-Shock Proteins/genetics , Oligospermia/genetics , Progesterone/metabolism , Spermatozoa/growth & development , Acrosome Reaction/genetics , Benzoquinones/pharmacology , Gene Expression Regulation, Developmental/drug effects , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Lactams, Macrocyclic/pharmacology , Male , Oligospermia/physiopathology , Progesterone/pharmacology , Semen Analysis , Sperm Capacitation/genetics , Sperm Motility/drug effects , Sperm Motility/genetics , Spermatozoa/pathology
16.
J Clin Diagn Res ; 10(9): LC06-LC11, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27790471

ABSTRACT

INTRODUCTION: India is the 2nd most populated country in the world. Population of India is increasing at a tremendous rate. Proportionately, the numbers of people seeking health care are increasing. In that ratio the quantities of hospital wastes, in wider terms, healthcare wastes that are getting generated is also increasing. Current methods for the safe disposal of healthcare wastes are not able to cope up with the rate of generation of healthcare wastes and moreover are not eco-friendly at all. Due to this, the current rules and regulations regarding the safe disposal of healthcare wastes are getting violated, ultimately leading to improper management of healthcare wastes, posing a serious threat to the environment and to the community. AIM: To develop a novel, sustainable and beneficial system for the systematic management of healthcare wastes utilizing the strategies of waste reduction, waste segregation and recycling of Non Hazardous Hospital Wastes (NHHWs). MATERIALS AND METHODS: Firstly a detailed study of the Healthcare Waste Management System (HCWMS) operational at the Jaslok Hospital and Research Centre was done. A pilot study was then performed. After that, data regarding the generation and management of healthcare wastes in the other healthcare settings was collected and analyzed. Considering all this, a novel, sustainable and beneficial template system for the systematic management of healthcare wastes was proposed. Lastly the possible positive impacts from the implementation of HCWMSs designed using proposed template HCWMS in significant numbers of healthcare establishments was gauged. RESULTS: The healthcare waste management system operational at the Jaslok Hospital and Research Centre was found to be very efficient and provided vital inputs about developing the novel HCWMS. The pilot study was successfully completed generating significant revenue from the hospital's own NHHWs while managing them in an eco-friendly way. The total healthcare waste generation in Maharashtra was approximately estimated at about 2,89,200kg/day of which about 43,380kg/day was Bio-Medical Wastes (BMWs) while about 2,45,820kg/day were the NHHWs. This stresses the need of implementing HCWMSs in Healthcare Establishments (HCEs) based on the proposed novel template of HCWMS. CONCLUSION: The novel template system is proposed in a detailed manner under various heads in the form of a handbook which is scalable upwards or downwards as per the requirement of a HCE. The enormous economic and environmental positive impacts from the implementation of the HCWMSs based on the proposed HCWMS in significant numbers of HCEs were presented numerically, putting light on the necessity and tremendous potential of this field of research.

17.
Stem Cells Int ; 2016: 5096596, 2016.
Article in English | MEDLINE | ID: mdl-26635884

ABSTRACT

Adult mammalian ovary has been under the scanner for more than a decade now since it was proposed to harbor stem cells that undergo postnatal oogenesis during reproductive period like spermatogenesis in testis. Stem cells are located in the ovary surface epithelium and exist in adult and menopausal ovary as well as in ovary with premature failure. Stem cells comprise two distinct populations including spherical, very small embryonic-like stem cells (VSELs which express nuclear OCT-4 and other pluripotent and primordial germ cells specific markers) and slightly bigger ovarian germ stem cells (OGSCs with cytoplasmic OCT-4 which are equivalent to spermatogonial stem cells in the testes). These stem cells have the ability to spontaneously differentiate into oocyte-like structures in vitro and on exposure to a younger healthy niche. Bone marrow may be an alternative source of these stem cells. The stem cells express FSHR and respond to FSH by undergoing self-renewal, clonal expansion, and initiating neo-oogenesis and primordial follicle assembly. VSELs are relatively quiescent and were recently reported to survive chemotherapy and initiate oogenesis in mice when exposed to FSH. This emerging understanding and further research in the field will help evolving novel strategies to manage ovarian pathologies and also towards oncofertility.

18.
Reprod Biomed Online ; 32(2): 218-24, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26655651

ABSTRACT

The AZFc locus on the human Y chromosome harbours several multicopy genes, some of which are required for spermatogenesis. It is believed that deletion of one or more copies of these genes is a cause of infertility in some men. GOLGA2LY is one of the genes in the AZFc locus and it exists in two copies, GOLGA2P2Y and GOLGA2P3Y. The involvement of GOLGA2LY gene copy deletions in male infertility, however, is unknown. This study aimed to investigate the association of deletions of GOLGA2P2Y and GOLGA2P3Y gene copies with male infertility and with sperm concentration and motility. The frequency of GOLGA2P3Y deletion was significantly higher in oligozoospermic men compared with normozoospermic men (7.7% versus 1.2%; P = 0.0001), whereas the frequency of GOLGA2P2Y deletion was comparable between oligozoospermic and normozoospermic men (10.3% versus 11.3%). The deletion of GOLGA2P3Y but not GOLGA2P2Y was significantly higher (P = 0.03) in men with gr/gr rearrangements, indicating that GOLGA2P3Y deletions increase the susceptibility of men with gr/gr rearrangements to oligozoospermia. Furthermore, men with GOLGA2P3Y deletion had reduced sperm concentration and motility compared with men without deletion or with deletion of GOLGA2P2Y. These findings indicate GOLGA2P3Y gene copy may be candidate AZFc gene for male infertility.


Subject(s)
Autoantigens/genetics , Membrane Proteins/genetics , Oligospermia/genetics , Autoantigens/physiology , Chromosomes, Human, Y , Gene Deletion , Gene Rearrangement , Genetic Predisposition to Disease/genetics , Humans , Infertility, Male/genetics , Male , Membrane Proteins/physiology , Oligospermia/physiopathology , Risk Factors , Sequence Deletion , Sperm Count , Sperm Motility , Spermatogenesis , Spermatozoa/physiology , Temperature
19.
J Clin Endocrinol Metab ; 100(2): 744-53, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25393639

ABSTRACT

CONTEXT: Polycystic ovary syndrome (PCOS), a major cause of anovulatory infertility, is characterized by arrested follicular growth. Altered protein levels in the follicular fluid surrounding the ovum may reflect the molecular defects of folliculogenesis in these women. OBJECTIVE: To identify differentially regulated proteins in PCOS by comparing the follicular fluid protein repertoire of PCOS with healthy women. METHODS: The follicular fluid samples were collected from PCOS and normo-ovulatory women undergoing in vitro fertilization. Follicular fluid proteins were subjected to digestion using trypsin, and resultant peptides were labeled with isobaric tags for relative and absolute quantification reagents and analyzed by liquid chromatography tandem mass spectrometry. Differential abundance of selected proteins was confirmed by ELISA. RESULTS: A total of 770 proteins were identified, of which 186 showed differential abundance between controls and women with PCOS. Proteins involved in various processes of follicular development including amphiregulin; heparan sulfate proteoglycan 2; tumor necrosis factor, α-induced protein 6; plasminogen; and lymphatic vessel endothelial hyaluronan receptor 1 were found to be deregulated in PCOS. We also identified a number of new proteins from follicular fluid, whose function in the ovary is not yet clearly established. These include suprabasin; S100 calcium binding protein A7; and helicase with zinc finger 2, transcriptional coactivator. CONCLUSIONS: Proteins indispensable for follicular growth were found to be differentially expressed in follicular fluid of women with PCOS, which may in part explain the aberrant folliculogenesis observed in these women.


Subject(s)
Follicular Fluid/metabolism , Infertility, Female/metabolism , Ovarian Follicle/metabolism , Polycystic Ovary Syndrome/metabolism , Adult , Female , Fertilization in Vitro , Humans , Ovarian Follicle/growth & development , Proteomics
20.
Reprod Biol Endocrinol ; 12: 114, 2014 Nov 24.
Article in English | MEDLINE | ID: mdl-25421462

ABSTRACT

The urge to have one's own biological child supersedes any desire in life. Several options have been used to obtain gametes including pluripotent stem cells (embryonic ES and induced pluripotent iPS stem cells); gonadal stem cells (spermatogonial SSCs, ovarian OSCs stem cells), bone marrow, mesenchymal cells and fetal skin. However, the field poses a huge challenge including inefficient existing protocols for differentiation, epigenetic and genetic changes associated with extensive in vitro manipulation and also ethical/regulatory constraints. A tremendous leap in the field occurred using mouse ES and iPS cells wherein they were first differentiated into epiblast-like cells and then primordial germ cell-like cells. These on further development produced sperm, oocytes and live offspring (had associated genetic problems). Evidently differentiating pluripotent stem cells into primordial germ cells (PGCs) remains a major bottleneck. Against this backdrop, we propose that a novel population of pluripotent stem cells termed very small embryonic-like stem cells (VSELs) may serve as an alternative, potential source of autologus gametes, keeping in mind that they are indeed PGCs surviving in adult mammalian ovaries and testes. Both VSELs and PGCs are pluripotent, relatively quiescent because of epigenetic modifications of parentally imprinted genes loci like Igf2-H19 and KCNQ1p57, share several markers like Stella, Fragilis, Mvh, Dppa2, Dppa4, Sall4, Blimp1 and functional receptors. VSELs are localized in the basement membrane of seminiferous tubules in testis and in the ovary surface epithelium. Ovarian stem cells from mouse, rabbit, sheep, marmoset and humans (menopausal women and those with premature ovarian failure) spontaneously differentiate into oocyte-like structures in vitro with no additional requirement of growth factors. Thus a more pragmatic option to obtain autologus gametes may be the pluripotent VSELs and if we could manipulate them in vivo - existing ethical and epigenetic/genetic concerns associated with in vitro culture may also be minimized. The field of oncofertility may undergo a sea-change and existing strategies of cryopreservation of gametes and gonadal tissue for fertility preservation in cancer patients will necessitate a revision. However, first the scientific community needs to arrive at a consensus about VSELs in the gonads and then work towards exploiting their potential.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Germ Cells/cytology , Induced Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/cytology , Adult , Animals , Callithrix , Embryonic Stem Cells/metabolism , Female , Germ Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Meiosis/genetics , Mice , Models, Biological , Ovary/cytology , Ovary/metabolism , Pluripotent Stem Cells/metabolism , Rabbits , Sheep , Stem Cells/cytology , Stem Cells/metabolism , Testis/cytology , Testis/metabolism , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...