Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters

Therapeutic Methods and Therapies TCIM
Country/Region as subject
Publication year range
1.
Taiwan J Obstet Gynecol ; 61(2): 223-229, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35361380

ABSTRACT

OBJECTIVE: Poor ovarian responder (POR) poses a significant challenge for in vitro fertilization (IVF). Previous studies have suggested that dehydroepiandrosterone (DHEA) may improve IVF outcomes in POR. The current study attempts to investigate the clinical benefits of DHEA in POR and the possible mechanism of DHEA on cumulus cells (CCs). MATERIALS AND METHODS: A total of 60 women who underwent IVF treatment participated, including 22 normal ovarian responders (NORs) and 38 PORs. PORs were assigned to receive DHEA supplementation (n = 18) or not (n = 20) before IVF cycles. For all patients, CCs were obtained after oocyte retrieval. In the CCs, mRNA expression of mitochondrial dynamics relataed genes were measured. RESULTS: Supplementation of DHEA in POR reduced mitochondrial fission in CCs and decreased the expression of PGAM5 in CCs. CONCLUSION: The benefit of DHEA supplementation on IVF outcomes in POR is significant, and this effect may be mediated in part through improved mitochondrial dynamics in CC.


Subject(s)
Cumulus Cells , Dehydroepiandrosterone , Mitochondrial Dynamics , Mitochondrial Proteins , Ovulation Induction , Phosphoprotein Phosphatases , Cumulus Cells/cytology , Cumulus Cells/drug effects , Dehydroepiandrosterone/pharmacology , Female , Fertilization in Vitro , Humans , Mitochondrial Dynamics/drug effects , Mitochondrial Proteins/genetics , Ovary , Phosphoprotein Phosphatases/genetics
2.
J Tissue Eng Regen Med ; 15(10): 807-817, 2021 10.
Article in English | MEDLINE | ID: mdl-34310055

ABSTRACT

Here, we present a novel in vitro maturation (IVM) system comprising an agarose matrix supplemented with extracellular matrix (ECM) proteins for enhanced maturation of immature oocytes within cumulus-oocyte complexes (COCs) derived from porcine medium antral follicles (MAFs). Immunocytochemical analyses of integrin subunit α2 , α5 , α6 , ß1 , and ß4 expression suggested that integrin α2 ß1 , α5 ß1 , α6 ß1 , and α6 ß4 play pivotal roles in IVM of porcine immature oocytes. Combinatorial supplementation of fibronectin interacting with integrin α5 ß1 , collagen interacting with integrin α2 ß1 , and laminin interacting with integrin α6 ß1 and α6 ß4 to the agarose matrix had no significant effect on nuclear maturation. However, the number of parthenogenetic embryos that developed into blastocysts increased when oocytes were matured using agarose IVM matrices supplemented with fibronectin, collagen, or laminin. Furthermore, significant increases in cytoplasmic maturation-related parameters (BMP15 level, cumulus cell expansion score, intra-oocyte ATP level, and index of cortical granule distribution) were observed in COCs matured in vitro using ECM protein-incorporated agarose matrices. Our data suggest that mature porcine oocytes with enhanced developmental competence and high-quality cytoplasm can be generated via IVM using agarose matrices supplemented with fibronectin, collagen, or laminin.


Subject(s)
Cytoplasm/metabolism , Extracellular Matrix Proteins/metabolism , Oocytes/cytology , Sepharose/pharmacology , Adenosine Triphosphate/metabolism , Animals , Blastocyst/drug effects , Bone Morphogenetic Protein 15 , Cumulus Cells/cytology , Cumulus Cells/drug effects , Cumulus Cells/metabolism , Cytoplasm/drug effects , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , In Vitro Oocyte Maturation Techniques , Integrins/metabolism , Oocytes/drug effects , Oocytes/metabolism , Parthenogenesis/drug effects , Protein Subunits/metabolism , Swine
3.
Naunyn Schmiedebergs Arch Pharmacol ; 394(7): 1487-1495, 2021 07.
Article in English | MEDLINE | ID: mdl-33683419

ABSTRACT

This study aims to evaluate the effect of melatonin supplementation on the outcomes of in vitro fertilization (IVF) and mitochondrial adenosine triphosphate production (MT-ATP6) gene expression in Iranian infertile couples. A single-blind nonrandomized controlled trial was conducted, recruiting 90 infertile couples who underwent IVF at an infertility center in Tehran, Iran. Patients who were assigned to the intervention group received melatonin as a supplementation to the standard controlled ovarian stimulation (COS). The control group received a COS protocol only. Primary outcome was the mRNA level of the MT-ATP6 gene in cumulus cells of ovarian follicles. Secondary outcomes were the mean number of mature oocytes retrieved, the embryo quality, and biochemical and clinical pregnancy rates. The mRNA level of the MT-ATP6 gene in cumulus cells between intervention and control groups was not statistically different (0.931 vs.1; P Ëƒ 0.05). The mean number of poor-quality embryos was significantly lower in the intervention group than that in the control group (0.27 vs. 0.80; P = 0.028). The biochemical and clinical pregnancy rates were higher in the intervention group (24% vs. 14%, P = 0.089, and 14% vs. 7%, P = 0.302, respectively); however, the difference was not significant. Melatonin supplementation did not increase the odds of clinical pregnancy and the number of mature oocytes retrieved, but significantly reduced the number of low-quality embryos. More extensive studies focusing on the level of MT-ATP6 gene expression in the oocyte or blastomere cells may further elucidate the effect of supplementation with melatonin in infertile couples who have poor clinical outcomes. Trial registration: Current Controlled Trials: IRCT2015042912307N4.


Subject(s)
Fertilization in Vitro/trends , Infertility/metabolism , Infertility/therapy , Melatonin/administration & dosage , Mitochondrial Proton-Translocating ATPases/biosynthesis , Pregnancy Rate/trends , Administration, Oral , Adult , Antioxidants/administration & dosage , Cumulus Cells/drug effects , Cumulus Cells/metabolism , Female , Fertilization in Vitro/methods , Gene Expression , Humans , Infertility/epidemiology , Iran/epidemiology , Male , Mitochondrial Proton-Translocating ATPases/genetics , Pregnancy , Single-Blind Method , Treatment Outcome
4.
Mol Reprod Dev ; 87(5): 607-619, 2020 05.
Article in English | MEDLINE | ID: mdl-32270588

ABSTRACT

Here, we investigated the biological effects of arachidonic acid (AA) in human cumulus granulosa cells (CGCs) after exposure to ASA. Cells were isolated from the follicular fluid and incubated with 0.5 mM acetylsalicylic acid (ASA) and 50 µM AA. Cell viability was analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. E2 and P4 levels were measured by chemiluminescence assay. Expression of genes including CYP19A1, FACN, and SCD1 was measured by real-time polymerase chain reaction assay. Oxidative status was analyzed by monitoring glutathione peroxidase activity. The fatty acid profile was analyzed by the gas chromatography technique. Enzyme-linked immunosorbent assay was used to measure prostaglandin E2 (PGE2 ) in CGCs after exposure to ASA and AA. Protein levels of the estrogen receptor were studied by immunofluorescence staining. Ultrastructural changes were evaluated by transmission electron microscopy imaging. ASA treatment reduced E2 production, Cyp19a1 expression, glutathione peroxidase (GPx) activity, and estradiol receptor expression in CGCs. The addition of AA prevented the ASA-induced E2 reduction (p < .05) and expression of Cyp19a1. Moreover, AA increased the antioxidant capacity of CGCs exposed to ASA by promoting GPx activity (p < .05). AA increased monounsaturated fatty acid/saturated fatty acid ratio compared with the ASA group (p < .05). AA supplementation triggered the synthesis and secretion of PGE2 in ASA-treated CGCS (p < .05). Cytoplasmic vacuolation observed in the ASA group and treatment with AA intensified vacuolation rate. The expression of the estrogen receptor was increased after AA supplementation. Data demonstrated that AA decreased the detrimental effects of ASA on human CGCs after 72 hr.


Subject(s)
Arachidonic Acid/pharmacology , Aspirin/adverse effects , Cumulus Cells/drug effects , Aspirin/pharmacology , Cell Survival/drug effects , Cells, Cultured , Cumulus Cells/cytology , Cumulus Cells/physiology , Dinoprostone/metabolism , Drug Interactions , Fatty Acids/metabolism , Female , Gene Expression/drug effects , Humans , Lipid Metabolism/drug effects , Receptors, Estrogen/drug effects , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism
5.
J Cell Physiol ; 234(11): 19574-19581, 2019 11.
Article in English | MEDLINE | ID: mdl-30980384

ABSTRACT

Juglone, a naphthoquinone isolated from many species of the Juglandaceae family, has been used in traditional Chinese medicine for centuries because of its antiviral, antibacterial, and antitumor activities. However, the toxicity of juglone has also been demonstrated. Here, we used porcine oocytes as a model to explore the effects of juglone on oocyte maturation and studied the impact of vitamin C (VC) administration on juglone exposure-induced meiosis defects. Exposure to juglone significantly restricted cumulus cell expansion and decreased the first polar body extrusion. In addition, juglone exposure disturbed spindle organization, actin assembly, and the distribution of mitochondria during oocyte meiosis, while the acetylation level of α-tubulin was also reduced. These defects were all ameliorated by VC administration. Our findings indicate that juglone exposure induced meiotic failure in porcine oocytes, while VC protected against these defects during porcine oocyte maturation by ameliorating the organization of the cytoskeleton and mitochondrial distribution.


Subject(s)
Ascorbic Acid/pharmacology , Meiosis/drug effects , Naphthoquinones/adverse effects , Oocytes/drug effects , Acetylation/drug effects , Animals , Cumulus Cells/drug effects , Cumulus Cells/pathology , Cytoskeleton/drug effects , Cytoskeleton/pathology , Female , In Vitro Oocyte Maturation Techniques , Mitochondria/drug effects , Mitochondria/pathology , Naphthoquinones/pharmacology , Oocytes/pathology , Polar Bodies/drug effects , Polar Bodies/pathology , Swine , Tubulin/genetics
6.
J Reprod Dev ; 65(3): 215-221, 2019 Jun 14.
Article in English | MEDLINE | ID: mdl-30760649

ABSTRACT

All-trans retinoic acid (RA) is a metabolite of vitamin A and has pleiotropic actions on many different biological processes, including cell growth and differentiation, and is involved in different aspects of fertility and developmental biology. In the current study, we investigated the effects of RA on camel (Camelus dromedarius) cumulus-oocyte complex in vitro maturation (IVM). IVM medium was supplemented with 0, 10, 20, and 40 µM RA. Application of 20 µM RA significantly reduced the proportion of degenerated oocytes and significantly improved oocyte meiosis and first polar body extrusion compared to the control and other experimental groups. Retinoic acid significantly reduced the mRNA transcript levels of apoptosis-related genes, including BAX and P53, and reduced the BAX/BCL2 ratio. In addition, RA significantly reduced the expression of the Transforming growth factor beta (TGFß) pathway-related transcripts associated with the actin cytoskeleton, ACTA2 and TAGLN; however, RA increased TGFß expression in cumulus cells. The small molecule SB-431542 inhibits the TGFß pathway by inhibiting the activity of activin receptor-like kinases (ALK-4, ALK-5, and ALK-7); however, combined supplementation with RA during IVM compensated for the inhibitory effect of SB-431542 on cumulus expansion, oocyte meiosis I, and first polar body extrusion in activated oocytes. The current study shows the beneficial effects of RA on camel oocyte IVM and provides a model to study the multifunctional mechanisms involved in cumulus expansion and oocyte meiosis, particularly those involved in the TGFß pathway.


Subject(s)
Cumulus Cells/cytology , In Vitro Oocyte Maturation Techniques , Oocytes/cytology , Tretinoin/pharmacology , Animals , Apoptosis/drug effects , Benzamides/pharmacology , Blastocyst/cytology , Camelus , Culture Media , Cumulus Cells/drug effects , Dioxoles/pharmacology , Embryo Culture Techniques , Female , Fertility , Nuclear Transfer Techniques , Oocytes/drug effects , Transforming Growth Factor beta/metabolism
7.
J Pineal Res ; 66(4): e12543, 2019 May.
Article in English | MEDLINE | ID: mdl-30584671

ABSTRACT

Aflatoxin B1 (AFB1) is a major food and feed contaminant that threaten public health. Previous studies indicate that AFB1 exposure disrupted oocyte maturation. However, an effective and feasible method is unavailable for protecting oocytes against toxicity of AFB1. In the present study, using in vitro matured porcine oocytes and parthenogenetic embryos as model, we confirmed that AFB1 exposure during in vitro oocyte maturation (IVM) significantly impaired both nuclear and cytoplasmic maturation in a dose- and time-dependent manner. The different concentrations of melatonin were also tested for their protective effects on oocytes against the AFB1-induced toxicity. Our results showed that supplementation of a relative high concentration of melatonin (10-3 mol/L) during IVM efficiently reversed the impaired development rate and blastocyst quality, to the levels comparable to those of the control group. Further analysis indicated that melatonin application efficiently alleviated reactive oxygen species accumulation and initiation of apoptosis induced by AFB1 exposure. In addition, disrupted GSH/GPX system, as well as inhibited mitochondrial DNA (mtDNA) replication and mitochondrial biogenesis in AFB1-treated oocytes, can be notably reversed by melatonin application. Furthermore, cumulus cells may be important in mediating the toxicity of AFB1 to oocytes, and the metabolism of AFB1 in cumulus cells can be depressed by melatonin. To the best of our knowledge, this is the first report to confirm that melatonin application can efficiently protect oocytes from AFB1-induced toxicity. Our study provides a promising and practical strategy for alleviating or reversing AFB1-induced female reproductive toxicity in both clinical treatment and domestic reproductive management.


Subject(s)
Aflatoxin B1/pharmacology , In Vitro Oocyte Maturation Techniques , Melatonin/pharmacology , Oocytes/cytology , Oocytes/drug effects , Animals , Apoptosis/drug effects , Cells, Cultured , Cumulus Cells/cytology , Cumulus Cells/drug effects , Cumulus Cells/metabolism , DNA Copy Number Variations/genetics , DNA Copy Number Variations/physiology , DNA, Mitochondrial/drug effects , Female , Glutathione/metabolism , In Situ Nick-End Labeling , Oocytes/metabolism , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Swine
8.
Theriogenology ; 118: 80-89, 2018 Sep 15.
Article in English | MEDLINE | ID: mdl-29885644

ABSTRACT

Oocyte quality is one of the important factors in female fertility, in vitro maturation (IVM), and subsequent embryonic development. In the present study, we assessed whether acetyl-l-carnitine (ALC) supplementation during in vitro maturation of buffalo oocytes could improve oocyte quality and subsequent embryonic development. To determine the optimal level of ALC supplementation, we matured cumulus-oocyte complexes in maturation medium supplemented with 0, 2.5, and 5 mM ALC. The oocytes with a polar body were selected for parthenogenetic activation (PA) and in vitro fertilization (IVF). We found that oocytes matured in 2.5 mM ALC had significantly higher PA blastocyst rate (P < 0.05) and blastocyst cell number than those of unsupplemented oocytes (P < 0.05) and a significantly higher IVF blastocyst rate than that of oocytes matured in 5 mM ALC (P < 0.05). In all further experiments, we supplemented the maturation medium with 2.5 mM ALC. We then tested whether ALC supplementation could improve various markers of oocytes and cumulus cells. We compared cell proliferation; concentrations of reactive oxygen species (ROS), intracellular ATP, estradiol, and progesterone; mitochondrial distribution; mitochondrial DNA copy number (mtDNA); and expression levels of four genes encoding oocyte-derived factors (GDF9, BMP15) and steroid hormones (StAR, P450scc) between the supplemented and unsupplemented oocytes and cumulus cells. Cumulus cells matured with ALC supplementation were more prolific than those matured without ALC supplementation (P < 0.05). Oocytes treated with ALC had lower concentrations of intracellular ROS (P < 0.05) and a higher rate of diffuse mitochondrial distributions (P < 0.05) than those of untreated oocytes. Additionally, the mtDNA was higher in the ALC-treated oocytes (P < 0.05) and cumulus cells (P < 0.05) than that in the untreated cells. The ALC-treated maturation medium had a higher postmaturation concentration of estradiol than that of the untreated medium (P < 0.05). Finally, the gene expression levels of P450scc and GDF9 were greater in ALC-treated oocytes and cumulus cells than those in untreated cells (P < 0.05). Therefore, in buffalo, our results suggest that ALC affects mitochondrial function, regulates oocyte-derived paracrine factors, and increases the production of steroid hormones, leading to increased quality of matured oocytes and improved embryonic development in vitro.


Subject(s)
Acetylcarnitine/pharmacology , Buffaloes , Embryonic Development/drug effects , In Vitro Oocyte Maturation Techniques/veterinary , Oocytes/drug effects , Acetylcarnitine/administration & dosage , Animals , Blastocyst/physiology , Cell Proliferation/drug effects , Culture Media , Cumulus Cells/drug effects , Cumulus Cells/physiology , DNA, Mitochondrial/analysis , Embryonic Development/physiology , Estradiol/analysis , Female , Fertilization in Vitro/veterinary , In Vitro Oocyte Maturation Techniques/methods , Oocytes/chemistry , Oocytes/physiology , Reactive Oxygen Species/analysis
9.
Reprod Fertil Dev ; 30(8): 1109-1115, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29448974

ABSTRACT

Phyto-oestrogens such as isoflavones are natural compounds that can profoundly affect reproductive function. In the present study, we tested whether including isoflavone compounds (genistein, biochanin A, formononetin) in the maturation medium would affect the outcomes for ovine oocytes in vitro. Each isoflavone compound was evaluated at five concentrations (0, 2.5, 5, 10, 25µgmL-1) and the entire protocol was repeated four times. Cumulus-oocyte complexes were randomly allocated to the treatments, then fertilised and cultured in vitro. Compared with control (0µgmL-1), the lower concentrations of isoflavone (2.5, 5 and 10µgmL-1) had no detectable effect on the rates of cleavage or embryo development, or on embryo total cell counts (TCC). However, the highest concentration (25µgmL-1) of all three isoflavones exerted a variety of effects (P<0.05): genistein decreased cleavage rate, blastocyst rate and blastocyst efficiency (blastocysts produced per 100 oocytes); biochanin A decreased cleavage rate and blastocyst efficiency; and formononetin decreased blastocyst rate and blastocyst efficiency. Biochanin A (25µgmL-1) reduced embryo TCC specifically at the hatched blastocyst stage (P<0.05). We conclude that the presence of isoflavones at 25µgmL-1 during IVM decreases the cleavage rate and inhibits blastocyst hatching.


Subject(s)
Cumulus Cells/drug effects , Embryonic Development/drug effects , Fertilization/drug effects , Oocytes/drug effects , Phytoestrogens/pharmacology , Animals , Dose-Response Relationship, Drug , Female , Genistein/pharmacology , In Vitro Oocyte Maturation Techniques , Isoflavones/pharmacology , Oocytes/growth & development , Sheep
10.
Biol Reprod ; 98(5): 644-653, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29385411

ABSTRACT

Low oocyte quality is a possible causal factor of obesity-induced infertility. High palmitic acid (PA) concentration in follicular fluid is a crucial feature noted in obese women. This study examined how high PA concentration reduced mitochondrial quality in oocytes and investigated a possible countermeasure against mitochondrial dysfunction. Cumulus cell-oocyte complexes were obtained from the ovaries of gilts, and incubated in medium containing PA (0.5 mM) or vehicle (BSA) for 44 h. Culturing oocytes at high PA concentration induced mitochondrial dysfunction determined by high reactive oxygen species and low ATP content in oocytes. Furthermore, high PA levels increased mitochondrial acetylation levels determined by a high degree of co-localization of TOMM20 and acetylated-lysine. In addition, high PA levels reduced the expression of Sirtuin 3 (SIRT3) and phosphorylated AMP-activated protein kinase (AMPK), while the AMPK activator, AICAR, restored mitochondrial function as well as oocyte ability and reduced the acetylation of mitochondrial protein. Supplementation of culture medium with dorsomorphin dihydrochloride (an AMPK inhibitor) reduced mitochondrial function and increased mitochondrial protein acetylation. Treatment of oocytes with LB100 (an inhibitor of AMPK dephosphorylation) reduced mitochondrial acetylation levels and restored mitochondrial function. Furthermore, high PA levels increased ceramide accumulation in oocytes, and addition of ceramide to the culture medium also induced mitochondrial dysfunction and increased mitochondrial acetylation. This detrimental effect of ceramide was diminished by AICAR treatment of oocytes. Our results indicated that PA induces ceramide accumulation and downregulates the AMPK/SIRT3 pathway causing mitochondrial protein hyperacetylation and dysfunction in oocytes.


Subject(s)
Ceramides/metabolism , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Oocytes/drug effects , Palmitic Acid/pharmacology , AMP-Activated Protein Kinases/metabolism , Acetylation , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Cumulus Cells/drug effects , Cumulus Cells/metabolism , Female , Mitochondria/metabolism , Oocytes/metabolism , Oxidative Stress/drug effects , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Sirtuin 3/metabolism , Swine
11.
Reprod Toxicol ; 75: 86-95, 2018 01.
Article in English | MEDLINE | ID: mdl-29247838

ABSTRACT

Here we report a retrospective analysis of negative effects of routine enrofloxacin treatment of recurrent diarrhea on the ovary and the developing oocytes of the common marmoset, a small New World primate. The most deleterious effect on oocytes was observed about two months post treatment suggesting that the enrofloxacin effect is on early growing follicles. Manifestations of toxicity included decreased numbers of growing follicles and recovered culturable oocytes, as well as signs of early atresia of granulosa cells. In addition, increased amounts of holed stroma after treatment strongly suggested increased death of the early growing follicles. Of the oocytes judged to be of adequate quality for culture, maturation rates were not affected but fertilization of in vitro matured MII oocytes and subsequent cleavage rates were severely reduced in the enrofloxacin treated animals. Further, the arrested oocytes, which failed to mature or fertilize, showed obvious meiotic spindle abnormalities.


Subject(s)
Anti-Bacterial Agents/toxicity , Fluoroquinolones/toxicity , Oocytes/drug effects , Administration, Oral , Animals , Callithrix , Cumulus Cells/cytology , Cumulus Cells/drug effects , Enrofloxacin , Estrogens/blood , Female , Fertilization/drug effects , Oocytes/metabolism , Oocytes/ultrastructure , Ovariectomy , Spindle Apparatus/drug effects
12.
Reprod Sci ; 25(6): 844-853, 2018 06.
Article in English | MEDLINE | ID: mdl-28345489

ABSTRACT

Stearoyl-coenzyme A desaturase 1 (SCD1) is a key enzyme in lipid metabolism and is expressed in cumulus cells. The objective of the present study was to evaluate the effect of SCD1 inhibition in human cumulus cells on triglyceride content, steroidogenesis, and oocyte in vitro maturation. Human cumulus cells were exposed to SCD1 inhibitor CAY10566 (SCDinhib) alone or in combination with oleic acid in primary culture. The SCDinhib markedly suppressed triglyceride accumulation (-47%, P = .01), aromatase gene expression (-36%, P = .02), and estradiol production (-49%, P = .01) even at a dose not affecting cell viability and apoptosis. Human immature oocytes at the germinal vesicle (GV) stage were cocultured with pretreated cumulus cells. The rate of oocytes reaching the metaphase II stage was significantly lower in coculture with SCDinhib-treated cumulus cells than with control cumulus cells (-18%, P < .01), which recovered by oleic acid supplementation. This finding on in vitro maturation rate was also reproducible with mouse GV oocytes. The results suggest that SCD1 activity is required for cumulus cell lipid storage and steroidogenesis. In addition, oocyte maturation is negatively affected by SCD1 inhibition in cumulus cells, possibly due to a deficient lipid-mediated paracrine support.


Subject(s)
Cumulus Cells/enzymology , In Vitro Oocyte Maturation Techniques , Stearoyl-CoA Desaturase/metabolism , Steroids/metabolism , Adult , Animals , Apoptosis , Aromatase/metabolism , Cell Survival , Coculture Techniques , Cumulus Cells/drug effects , Estradiol/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Oleic Acid/administration & dosage , Primary Cell Culture , RNA, Messenger/metabolism , Stearoyl-CoA Desaturase/antagonists & inhibitors , Triglycerides/metabolism , Young Adult
13.
Reprod Fertil Dev ; 30(3): 417-429, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28822459

ABSTRACT

Dietary supplementation with propylene glycol (PG) increases in vitro production of high-quality embryos in feed-restricted heifers. The aim of the present study was to evaluate the effects of PG in feed-restricted heifers on follicular fluid insulin and insulin-like growth factor (IGF) 1 concentrations, expression of IGF system genes in oocytes and cumulus cells and the expression of selected genes in blastocysts. Feed-restricted (R) heifers were drenched with water or PG during induced oestrous cycles (400mL of PG or water/drench, daily drenching at 1600 hours for the first 9 days of the oestrous cycle). Ovum pick-up (OPU) was performed after superovulation to produce in vitro embryos and without superovulation to recover oocytes, cumulus cells and follicular fluid. OPU was also performed in a control group (not feed restricted and no drenching). Follicular fluid IGF1 concentrations were reduced by R, and PG restored IGF1 concentrations to those seen in the control group. In cumulus cells, expression of IGF1, IGF1 receptor (IGF1R) and IGF binding protein 4 (IGFBP4) was decreased in the R group, and fully (IGF1 and IGF1R) or partially (IGFBP4) restored to control levels by PG. Blastocyst perilipin 2 (PLIN2; also known as adipophilin), Bcl-2-associated X protein (BAX), SCL2A1 (facilitated glucose/fructose transporter GLUT1), aquaporin 3 (AQP3), DNA (cytosine-5)-methyltransferase 3A (DNMT3A) and heat shock 70-kDa protein 9 (HSPA9B) expression were decreased in R heifers; PG restored the expression of the last four genes to control levels. In conclusion, these results suggest that, during follicular growth, PG exerts epigenetic regulatory effects on gene expression in blastocyst stage embryos.


Subject(s)
Blastocyst/drug effects , Caloric Restriction/veterinary , Cumulus Cells/drug effects , Dairying , Dietary Supplements , Fertilization in Vitro/veterinary , Follicular Fluid/drug effects , Oocytes/drug effects , Propylene Glycol/administration & dosage , Transcriptome/drug effects , Administration, Oral , Animals , Blastocyst/metabolism , Cattle , Cumulus Cells/metabolism , Epigenesis, Genetic/drug effects , Female , Follicular Fluid/metabolism , Gene Expression Profiling/veterinary , Gene Expression Regulation, Developmental/drug effects , Insulin/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Nutritional Status , Oocytes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors
14.
J Ovarian Res ; 10(1): 74, 2017 Nov 09.
Article in English | MEDLINE | ID: mdl-29122003

ABSTRACT

BACKGROUND: Supplementation of bovine oocyte-cumulus complexes during in vitro maturation (IVM) with 1 µM of docosahexaenoic acid (DHA), C22:6 n-3 polyunsaturated fatty acid, was reported to improve in vitro embryo development. The objective of this paper was to decipher the mechanisms of DHA action. RESULTS: Transcriptomic analysis of 1 µM DHA-treated and control cumulus cells after 4 h IVM showed no significant difference in gene expression. MALDI-TOF mass spectrometry analysis of lipid profiles in DHA-treated and control oocytes and cumulus cells after IVM showed variations of only 3 out of 700 molecular species in oocytes and 7 out of 698 species in cumulus cells (p < 0.01). We showed expression of free fatty acid receptor FFAR4 in both oocytes and cumulus cells, this receptor is known to be activated by binding to DHA. FFAR4 protein was localized close to the cellular membrane by immunofluorescence. Functional studies demonstrated that supplementation with FFAR4 agonist TUG-891 (1 µM or 5 µM) during IVM led to an increased blastocyst rate (39.5% ± 4.1%, 41.3% ± 4.1%), similar to DHA 1 µM treatment (39.2% ± 4.1%) as compared to control (25.2% ± 3.6%). FFAR4 activation via TUG-891 led to beneficial effect on oocyte developmental competence and might explain in part similar effects of DHA. CONCLUSIONS: In conclusion, we suggested that low dose of DHA (1 µM) during IVM might activate regulatory mechanisms without evident effect on gene expression and lipid content in oocyte-cumulus complexes, likely through signaling pathways which need to be elucidated in further studies.


Subject(s)
Cumulus Cells/drug effects , Cumulus Cells/metabolism , Docosahexaenoic Acids/pharmacology , Oocytes/drug effects , Oocytes/metabolism , Animals , Cattle , Embryonic Development/drug effects , Embryonic Development/genetics , Female , Fertilization in Vitro , Gene Expression Profiling , Gene Expression Regulation/drug effects , Immunohistochemistry , In Vitro Oocyte Maturation Techniques , Lipid Metabolism/drug effects , Lipids , Mitogen-Activated Protein Kinase 3/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
15.
J Obstet Gynaecol Res ; 43(12): 1855-1862, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28892223

ABSTRACT

AIM: Ovarian aging, which leads to diminished ovarian reserve and decreased oocyte quality, is highly associated with poor reproductive outcomes. It has been suggested that dehydroepiandrosterone (DHEA) might be able to temporarily slow down the aging process. This study attempted to investigate the clinical benefits of DHEA in older patients and the anti-senescence effect of DHEA on cumulus cells (CC) and human ovarian granulosa cells (HO23 cell line). METHODS: This prospective study enrolled 88 patients who underwent in vitro fertilization (IVF), including 30 younger patients (aged ≤ 37 years) and 58 older patients (aged > 37 years). Older patients were assigned to receive DHEA treatment or not prior to the IVF cycle. CC were obtained from all patients after oocyte retrieval and the HO23 granulosa cell line was used for in vitro studies. Senescence-associated ß-galactosidase (SA-ß-gal) was used as a biomarker of senescence. RESULTS: In older patients, following DHEA supplementation, a greater number of transferred embryos and a higher fertilization rate were observed compared with those in patients without DHEA supplementation. However, the clinical pregnancy rate was not significantly increased following DHEA supplementation. Additionally, treatment with DHEA resulted in significantly reduced SA-ß-gal staining in both CC and HO23 cells. CONCLUSION: DHEA supplementation ameliorated IVF outcomes but without a consequence on pregnancy rate in older patients and decreased SA-ß-gal activity in CC and HO23 cells, suggesting that DHEA might be used as a possible intervention to slow down ovarian aging.


Subject(s)
Dehydroepiandrosterone/pharmacology , Ovary/drug effects , Ovary/physiology , Adult , Aging , Cell Line , Cellular Senescence/drug effects , Cohort Studies , Cumulus Cells/drug effects , Female , Fertilization in Vitro , Granulosa Cells/drug effects , Humans , Oocytes/drug effects , Oocytes/physiology , Prospective Studies , Treatment Outcome
16.
Int J Med Sci ; 14(6): 585-594, 2017.
Article in English | MEDLINE | ID: mdl-28638275

ABSTRACT

Poor ovarian responders (PORs) pose a great challenge for in vitro fertilization (IVF). Previous studies have suggested that dehydroepiandrosterone (DHEA) may improve IVF outcomes in PORs. The current study attempted to investigate the clinical benefits of DHEA in PORs and the possible mechanisms of DHEA on cumulus cells (CCs). This was a prospective study performed at one tertiary center from January 2015 to March 2016. A total of 131 women who underwent IVF treatment participated, including 59 normal ovarian responders (NORs) and 72 PORs. PORs were assigned to receive DHEA supplementation or not before the IVF cycle. For all patients, CCs were obtained after oocyte retrieval. In the CCs, mRNA expression of apoptosis-related genes and mitochondrial transcription factor A (TFAM) gene, terminal deoxynucleotidyl transferase dUTP nick end labeling assay, mitochondrial dehydrogenase activity and mitochondrial mass were measured. The results indicated that PORs with DHEA supplementation produces a great number of top-quality embryos at day 3 and increased the number of transferred embryos and fertilization rate compared with those without DHEA supplementation. Additionally, supplementation with DHEA in PORs decreased DNA damage and apoptosis in CCs while enhancing the mitochondrial mass, mitochondrial dehydrogenase activity and TFAM expression in CCs. In conclusion, our results showed that the benefits of DHEA supplementation on IVF outcomes in PORs were significant, and the effects may be partially mediated by improving mitochondrial function and reducing apoptosis in CCs.


Subject(s)
Cumulus Cells/drug effects , Dehydroepiandrosterone/administration & dosage , Ovary/drug effects , Ovulation Induction , Adult , Apoptosis/drug effects , Female , Fertilization in Vitro , Humans , Mitochondria/drug effects , Ovary/growth & development
17.
Biol Reprod ; 96(6): 1181-1196, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28520897

ABSTRACT

Elevated concentrations of free fatty acids (FFAs), predominantly palmitic, stearic, and oleic acids (PSO), exert detrimental effects on oocyte developmental competence. This study examined the effects of omega-3 alpha-linolenic acid (ALA) during in vitro oocyte maturation (IVM) in the presence of PSO on subsequent embryo development and quality, and the cellular mechanisms that might be involved. Bovine cumulus-oocyte complexes (COCs) were supplemented during IVM with ALA (50 µM), PSO (425 µM), or PSO+ALA. Compared with FFA-free controls (P < 0.05), PSO increased embryo fragmentation and decreased good quality embryos on day 2 postfertilization. Day 7 blastocyst rate was also reduced. Day 8 blastocysts had lower cell counts and higher apoptosis but normal metabolic profile. In the PSO group, cumulus cell (CC) expansion was inhibited with an increased CC apoptosis while COC metabolism was not affected. Mitochondrial inner membrane potential (MMP; JC-1 staining) was reduced in the CCs and oocytes. Heat shock protein 70 (HSP70) but not glucose-regulated protein 78 kDa (GRP78, known as BiP; an endoplasmic reticulum stress marker) was upregulated in the CCs. Higher reactive oxygen species levels (DCHFDA staining) were detected in the oocytes. In contrast, adding ALA in the presence of PSO normalized embryo fragmentation, cleavage, blastocyst rates, and blastocyst quality compared to controls (P > 0.05). Combined treatment with ALA also reduced CC apoptosis, partially recovered CC expansion, abrogated the reduction in MMP in the CCs but not in the oocytes, and reduced BiP and HSP70 expression in CCs, compared with PSO only (P < 0.05). In conclusion, ALA supplementation protected oocyte developmental capacity under lipotoxic conditions mainly by protecting cumulus cell viability.


Subject(s)
Cattle/physiology , Cumulus Cells/drug effects , In Vitro Oocyte Maturation Techniques/veterinary , Oocytes/drug effects , alpha-Linolenic Acid/pharmacology , Animals , Biomarkers , Blastocyst/drug effects , Blastocyst/physiology , Cumulus Cells/physiology , Mitochondria/physiology , Oocytes/physiology , Stress, Physiological/physiology
18.
Int J Mol Sci ; 18(4)2017 Apr 17.
Article in English | MEDLINE | ID: mdl-28420163

ABSTRACT

(1) Background: The binding sites of melatonin, as a multifunctional molecule, have been identified in human, porcine, and bovine samples. However, the binding sites and mechanisms of melatonin have not been reported in sheep; (2) Methods: Cumulus-oocyte complexes (COCs) were cultured in TCM-199 supplemented with melatonin at concentrations of 0, 10-3, 10-5, 10-7, 10-9, and 10-11 M. Melatonin receptors (MT1 and MT2) were evaluated via immunofluorescence and Western blot. The effects of melatonin on cumulus cell expansion, nuclear maturation, embryo development, and related gene (GDF9, DNMT1, PTX3, HAS2, and EGFR) expression were investigated. The level of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) were evaluated in oocytes and cumulus, respectively; (3) Results: Both MT1 and MT2 were expressed in oocytes, cumulus cells, and granulosa cells. Melatonin with a concentration of 10-7 M significantly enhanced the rates of nuclear maturation, cumulus cells expansion, cleavage, and blastocyst. Melatonin enhanced the expression of BMP15 in oocytes and of PTX3, HAS2, and EGFR in cumulus cells. Melatonin decreased the cAMP level of oocytes but enhanced the cGMP level in oocytes and cumulus cells; (4) Conclusion: The higher presence of MT1 in GV cumulus cells and the beneficial effects of melatonin indicated that its roles in regulating sheep oocyte maturation may be mediated mainly by the MT1 receptor.


Subject(s)
Cell Differentiation/drug effects , Melatonin/metabolism , Melatonin/pharmacology , Oocytes/cytology , Oocytes/metabolism , Receptors, Melatonin/metabolism , Animals , Cumulus Cells/drug effects , Cumulus Cells/metabolism , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Embryonic Development/genetics , Female , Gene Expression , Gene Expression Regulation, Developmental , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Sheep
19.
Reprod Fertil Dev ; 29(11): 2217-2224, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28356185

ABSTRACT

In vivo, oocyte maturation is triggered by the ovulatory LH surge, whereas in vitro it is precociously induced when the cumulus-oocyte complex is removed from the follicle. Natriuretic peptide C (NPPC) delays germinal vesicle breakdown (GVBD) while increasing oocyte-cumulus communication during in vitro maturation (IVM) in cattle. In the present study we first tested the hypothesis that steroids secreted by the follicle (17ß-oestradiol, progesterone and androstenedione) interact with NPPC to delay GVBD and to maintain oocyte-cumulus communication as assessed by transfer of a dye (Lucifer Yellow) from the oocyte to cumulus cells. Then, we assessed the effects of steroid hormones and NPPC, alone and in combination in a pre-IVM culture, on embryo production. The combination of NPPC with steroids delayed GVDB, increased natriuretic peptide receptor 2 (NPR2) mRNA abundance in cumulus cells during culture, and maintained oocyte-cumulus communication at levels not different from non-cultured controls. The addition of steroids and/or NPPC to a pre-IVM culture did not alter blastocyst rates after IVF, but supplementation with steroids increased blastocyst total cell number. The present study provides evidence, for the first time in cattle, that steroids interact with NPPC to regulate oocyte nuclear maturation and oocyte-cumulus communication, and improve oocyte developmental competence.


Subject(s)
Androstenedione/pharmacology , Cumulus Cells/metabolism , Estradiol/pharmacology , In Vitro Oocyte Maturation Techniques/veterinary , Natriuretic Peptide, C-Type/pharmacology , Oocytes/metabolism , Progesterone/pharmacology , Animals , Cattle , Cumulus Cells/drug effects , Female , Oocytes/drug effects , Oogenesis/drug effects , Ovarian Follicle/metabolism
20.
Biol Trace Elem Res ; 176(2): 374-383, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27604975

ABSTRACT

Progesterone production is upregulated in granulosa cells (cumulus and mural) after the LH surge, but the intra-follicular mechanisms regulating this transition are not completely known. Recent findings show that the transition metal chelator, N,N,N',N'-tetrakis-(2-pyridylmethyl)-ethylenediamine (TPEN), impairs ovarian function. In this study, we provide evidence that chelating transition metals, including zinc, enhances progesterone production. The findings show that TPEN (transition metal chelator) increases abundance of Cyp11a1 and Star messenger RNA (mRNA) between 8- and 20-fold and progesterone production more than 3-fold in cultured cumulus-oocyte complexes (COC). Feeding a zinc-deficient diet for 10 days, but not 3 days, increased Star, Hsd3b, and prostaglandin F2 alpha receptor (Ptgfr) mRNA ~2.5-fold, suggesting that the effect of TPEN is through modulation of zinc availability. Progesterone from cumulus cells promotes oocyte developmental potential. Blocking progesterone production with epostane during maturation reduced subsequent blastocyst formation from 89 % in control to 18 % in epostane-treated complexes, but supplementation with progesterone restored blastocyst developmental potential to 94 %. Feeding a zinc-deficient diet for 5 days before ovulation did not affect the number of CL, STAR protein, or serum progesterone. However, incubating luteal tissue with TPEN increased abundance of Star, Hsd3b, and Ptgfr mRNA 2-3-fold and increased progesterone production 3-fold. TPEN is known to abolish SMAD2/3 signaling in cumulus cells. However, treatment of COC with the SMAD2/3 phosphorylation inhibitor, SB421542, did not by itself induce steroidogenic transcripts but did potentiate EGF-induced Star mRNA expression. Collectively, the results show that depletion of transition metals with TPEN acutely enhances progesterone biosynthesis in COC and luteal tissue.


Subject(s)
Chelating Agents/pharmacology , Corpus Luteum/drug effects , Corpus Luteum/metabolism , Cumulus Cells/drug effects , Cumulus Cells/metabolism , Progesterone/biosynthesis , Zinc/pharmacology , Animals , Cells, Cultured , Chelating Agents/administration & dosage , Diet , Female , Mice , Zinc/administration & dosage , Zinc/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL