transforming-growth-factor-beta and Primary-Ovarian-Insufficiency

transforming-growth-factor-beta has been researched along with Primary-Ovarian-Insufficiency* in 21 studies

Reviews

3 review(s) available for transforming-growth-factor-beta and Primary-Ovarian-Insufficiency

ArticleYear
Genetic defects of ovarian TGF-β-like factors and premature ovarian failure.
    Journal of endocrinological investigation, 2011, Volume: 34, Issue:3

    Premature ovarian failure (POF) is an ovarian defect characterized by the premature depletion of ovarian follicles; POF affects approximately 1-2% of women under the age of 40 yr, thus representing one major cause of female infertility. POF relevance is continuously growing because women tend to conceive always more frequently beyond 30 yr. Frequently, POF is the end-stage of an occult process [primary ovarian insufficiency (POI)]. POI is a heterogeneous disease caused by a variety of mechanisms. Though the underlying cause remains unexplained in the majority of cases, several data indicate that POI has a strong genetic component. These data include the existence of several causal genetic defects in human, experimental, and natural models, as well as the frequent familiarity. The candidate genes are numerous, but POF remains unexplained in most of the cases. Several recent evidences have driven the attention of researchers on the possible involvement of various elements belonging to the transforming growth factor β family, which includes bone morphogenetic proteins, growth/differentiation factors, and inhibins. These peptides are produced by either the oocyte or granulosa cells to constitute a complex paracrine network within the ovarian follicle. Here, we review the studies reporting the genetic alterations of these factors in human and animal defects of ovarian folliculogenesis which support the fundamental roles played by these signals in ovarian morphogenesis and function.

    Topics: Animals; Bone Morphogenetic Protein 15; Bone Morphogenetic Protein Receptors, Type I; Female; Growth Differentiation Factor 9; Humans; Infertility, Female; Inhibins; Ovarian Follicle; Primary Ovarian Insufficiency; Transforming Growth Factor beta

2011
[Genetic analysis of premature ovarian failure: role of forkhead and TGF-beta genes].
    Gynecologie, obstetrique & fertilite, 2008, Volume: 36, Issue:9

    Premature ovarian failure is a common pathology affecting 1% of women. Although multiple etiologies have been described the majority of cases are idiopathic. Forkhead transcription factors as FOXL2 and FOXO3A are of particular interest in the research of genetic factors related with the pathology as they are present in diverse developmental pathways and ovarian physiology. Similarly, some TGF-beta factors (i.e. BMP 15 and GDF-9) have been demonstrated to play a key role in the regulation, at ovarian level, of female reproduction. In recent years numerous studies have been performed in order to elucidate the implication of these factors in the ovarian physiopathology. The aim of this manuscript is to describe some of these advances in the context of premature ovarian failure.

    Topics: Female; Forkhead Transcription Factors; Humans; Mutation; Primary Ovarian Insufficiency; Transforming Growth Factor beta

2008
Recent advances in the study of genes involved in non-syndromic premature ovarian failure.
    Molecular and cellular endocrinology, 2008, Jan-30, Volume: 282, Issue:1-2

    Premature ovarian failure (POF) is a common pathology leading to infertility affecting about 1% of women under 40 years old. In POF patients, the ovarian dysfunction is characterized by the lack of the ovarian response to close a negative feedback loop on the synthesis of pituitary gonadotropins. Although the majority of cases are considered as idiopathic, diverse aetiologies have been associated, including genetic factors. Up to now, the potential genetic causes of non-syndromic POF have been established mainly by genetic linkage analysis of familial cases or by the screening of mutations in candidate genes based on animal models. Here, we review recent advances in the study of candidate genes.

    Topics: Bone Morphogenetic Protein 15; Female; Forkhead Transcription Factors; Growth Differentiation Factor 9; Humans; Inhibins; Intercellular Signaling Peptides and Proteins; Mutation; Primary Ovarian Insufficiency; Transforming Growth Factor beta

2008

Other Studies

18 other study(ies) available for transforming-growth-factor-beta and Primary-Ovarian-Insufficiency

ArticleYear
Regulatory T Cells Overexpressing Peli1 Show Better Efficacy in Repairing Ovarian Endocrine Function in Autoimmune Premature Ovarian Insufficiency.
    Laboratory investigation; a journal of technical methods and pathology, 2023, Volume: 103, Issue:2

    Regulatory T (Treg) cell dysfunction is involved in the pathogenesis of autoimmune premature ovarian insufficiency (POI). Adoptive transfer of Treg cells has been shown to be effective in the treatment of autoimmune POI in mice. However, the therapeutic effect of Treg cell therapy is limited because the phenotype and function of Treg cells is not properly maintained when they are reinfused in an inflammatory environment. Therefore, enhancing the function of Treg cells using genetic engineering is of great significance for improving the efficacy of Treg cells in the treatment of immune diseases. In this study, we investigated the role of the E3 ubiquitinated ligase Pellino 1 (Peli1) in the proliferation and immunosuppressive function of Treg cells and the therapeutic effect of Treg cells overexpressing Peli1 on autoimmune POI. The results showed that the overexpression of Peli1 promoted cell proliferation and enhanced the immunosuppressive function of Treg cells in vitro. After the adoptive transfer of Treg cells overexpressing Peli1 in autoimmune POI mice, the apoptosis rate of ovarian granulosa cells declined. The levels of the inflammatory inhibitors interleukin 10 and transforming growth factor-β as well as the ovarian hormone estradiol were elevated. The number of primordial, primary, secondary, and mature follicles was restored to a certain extent compared with those in control subjects. These results revealed that the adoptive transfer of Treg cells overexpressing Peli1 promoted its efficacy against zona pellucida protein 3 peptide-induced POI, which provides new insights into the treatment of autoimmune POI.

    Topics: Animals; Estradiol; Female; Humans; Mice; Nuclear Proteins; Primary Ovarian Insufficiency; T-Lymphocytes, Regulatory; Transforming Growth Factor beta; Ubiquitin-Protein Ligases

2023
Amniotic fluid-derived exosomes attenuated fibrotic changes in POI rats through modulation of the TGF-β/Smads signaling pathway.
    Journal of ovarian research, 2023, Jun-27, Volume: 16, Issue:1

    In the current study, we investigated the regenerative effects of amniotic fluid exosomes (AF-Exos) in a rat model for premature ovarian insufficiency (POI). POI is a condition characterized by a decrease in ovarian function that can lead to infertility. We induced POI by administering cyclophosphamide (CTX) for 15 consecutive days, and then transplanted AF-Exos directly into both ovarian tissues. Four weeks later, we measured the serum levels of follicle-stimulating hormone (FSH), luteinizing hormone (LH), and estradiol (E2), and performed histopathological evaluations using H & E and Masson's trichrome staining. We also monitored the expression of genes related to the TGF-β signaling pathway using real-time PCR and examined the fertility rate of POI rats after AF-Exos therapy. Histological analysis showed an increase in atretic follicles and a decrease in healthy follicle count after POI induction. Four weeks post-AF-Exos intervention, the healthy follicle count increased (p < 0.01) while the atretic follicle count decreased (p < 0.001). In parallel, the deposition of collagen fibers also decreased following AF-Exos transplantation. The concentrations of FSH and LH hormones in sera remained unchanged after injection of AF-Exos, while E2 levels increased (p < 0.05). The expression of Smad-4 (p < 0.01) and Smad-6 (p < 0.05) was upregulated in POI rats that received AF-Exos, while Smad-2, TGF-β1, TNF-α, and IL-10 remained statistically unchanged. Our records showed a notable increase in litter number after AF-Exos compared to the non-treated POI rats. These results suggest that AF-Exos transplantation has the potential to restore ovarian function through the TGF-β/Smads signaling pathway in POI rats.

    Topics: Amniotic Fluid; Animals; Exosomes; Female; Follicle Stimulating Hormone; Menopause, Premature; Primary Ovarian Insufficiency; Rats; Signal Transduction; Transforming Growth Factor beta

2023
Effects of umbilical cord mesenchymal stem cells on expression of CYR61, FSH and AMH in mice with premature ovarian failure.
    Cellular and molecular biology (Noisy-le-Grand, France), 2022, Jan-02, Volume: 67, Issue:4

    The aim of this study was to objective to investigate the effects of umbilical cord mesenchymal stem cells on the expression of CYR61, FSH and AMH in mice with premature ovarian failure. For this purpose, thirty SPF female SD mice were selected as the research object, 10 of which were control group, namely group α, and 20 mice with premature ovarian failure model were established by cyclophosphamide. The mice were divided into model group, namely β group and the umbilical cord mesenchymal stem cell transplantation group (γ group), with 10 mice in each group. ELSA method was used to determine the levels of follicle-stimulating hormone (FSH), Luteinizing hormone (LH), estradiol (Estradiol) in serum. The changes of E2, Antimullerian hormone (AMH) and cysteine-rich protein 61 in ovarian tissues were determined by the protein imprinting method. Connective tissue growth factor (CTGF) and caspase-3 protein expression. Results showed that in fertility rate, γ group > α group > β group, the difference was statistically significant (P<0.05), in litter size, α group > γ group > β group, the difference was statistically significant (P<0.05). The levels of serum E2 and AMH in α group > γ group > β group, and the levels of serum FSH and LH in β group > γ group > α group were statistically significant (P<0.05). The growth follicles were α group > γ group > β group, and the atresia follicles were β group > γ group > α group, and there was a statistical difference among all groups (P<0.05). There was no difference in luteal number among the three groups (P>0.05). In terms of CYR61 and CTGF protein expression, α group > γ group > β group, and in terms of caspase-3, β group > γ group > α group had statistical significance (P<0.05). It is concluded that intervention with umbilical cord mesenchymal stem cells can significantly improve the expression levels of CYR61 and AMH, reduce the level of FSH, promote cell survival, improve the reproductive quality of mice, and restore the physiological function of the ovary. It is feasible to treat premature ovarian failure with umbilical cord mesenchymal stem cells.

    Topics: Animals; Anti-Mullerian Hormone; Caspase 3; Estradiol; Female; Follicle Stimulating Hormone; Humans; Luteinizing Hormone; Mesenchymal Stem Cells; Mice; Primary Ovarian Insufficiency; Transforming Growth Factor beta; Umbilical Cord

2022
Bone Marrow-Derived Mesenchymal Stem Cells Reverse Radiotherapy-Induced Premature Ovarian Failure: Emphasis on Signal Integration of TGF-β, Wnt/β-Catenin and Hippo Pathways.
    Stem cell reviews and reports, 2021, Volume: 17, Issue:4

    Topics: Animals; beta Catenin; Bone Marrow; Female; Hippo Signaling Pathway; Mesenchymal Stem Cells; Primary Ovarian Insufficiency; Radiation Injuries, Experimental; Radiotherapy; Rats; Transforming Growth Factor beta; Wnt Signaling Pathway

2021
A homozygous truncating variant in GDF9 in siblings with primary ovarian insufficiency.
    Journal of assisted reproduction and genetics, 2021, Volume: 38, Issue:6

    Premature or primary ovarian insufficiency (POI) affects approximately 1% of women and can be due to a variety of causes. Genetic causes include syndromic and non-syndromic POI. There are several promising candidate genes for whom a clear Mendelian association with non-syndromic POI has not yet been conclusively established, including GDF9. GDF9 is an oocyte-secreted factor and is part of the TGF-beta superfamily of morphogens. It has an important role in follicular development and granulosa cell maturation. We report the case of two siblings with primary ovarian insufficiency (POI) and a homozygous truncating variant in GDF9 (c.604C>T; p.(Gln202*). This report helps establish a clear gene-disease association between GDF9 and POI and argues for routine evaluation for GDF9 variants in patients undergoing genomic investigation for POI.

    Topics: Adolescent; Adult; Female; Genetic Predisposition to Disease; Granulosa Cells; Growth Differentiation Factor 9; Humans; Morphogenesis; Oocytes; Ovarian Follicle; Primary Ovarian Insufficiency; Siblings; Transforming Growth Factor beta; Young Adult

2021
Novel variants in women with premature ovarian function decline identified via whole-exome sequencing.
    Journal of assisted reproduction and genetics, 2020, Volume: 37, Issue:10

    To investigate the potential etiologies of premature ovarian insufficiency (POI) and diminished ovarian reserve (DOR).. Fourteen women with sporadic POI and 6 women with DOR were enrolled. We used whole-exome sequencing (WES) and bioinformatics analysis to identify variants in a subset of 599 selected POI candidate genes. The identified genes were subjected to gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment and protein-protein interaction (PPI) network analyses to uncover key genes and pathways.. Among the 20 patients, 79 heterozygous variants were detected in 49 genes, which were classified as "likely pathogenic" or "variants of uncertain significance" according to the guidelines of the American College of Medical Genetics and Genomics. Most patients (17/20) carried two or more variants. Monoacylglycerol O-acyltransferase 1 mutations were found in six patients, and cytochrome P450 family 26 subfamily B member 1 and Bardet-Biedl syndrome 9 mutations were each found in four patients. Some variants were shared between DOR and POI. Enrichment analyses showed that the identified genes participate in key ovarian processes, such as follicular development, gonadal development, meiosis, Fanconi anemia, homologous recombination, and transforming growth factor β signaling. A PPI network revealed interactions between these proteins.. Premature ovarian function decline may be polygenic, and overlap exists between the genetic backgrounds of DOR and POI. WES and in silico analyses may be a useful clinical tool for etiological diagnosis and risk prediction for high-risk women in the future.

    Topics: Adult; Computational Biology; Exome Sequencing; Female; Genetic Variation; Genomics; Humans; Mutation; Ovarian Diseases; Ovarian Reserve; Ovary; Primary Ovarian Insufficiency; Transforming Growth Factor beta

2020
Mechanistic approach of the inhibitory effect of chrysin on inflammatory and apoptotic events implicated in radiation-induced premature ovarian failure: Emphasis on TGF-β/MAPKs signaling pathway.
    Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie, 2019, Volume: 109

    Radiotherapy is one of the most relevant treatment modalities for various types of malignancies. However, it causes premature ovarian failure (POF) and subsequent infertility in women of reproductive age; hence urging the development of effective radioprotective agents. Chrysin, a natural flavone, possesses several pharmacological activities owing to its antioxidant, anti-inflammatory and anti-apoptotic properties. Therefore, the aim of this study was to investigate the efficacy of chrysin in limiting γ-radiation-mediated POF and to elucidate the underlying molecular mechanisms. Immature female Sprague-Dawley rats were subjected to a single dose of γ-radiation (3.2 Gy) and/or treated with chrysin (50 mg/kg) once daily for two weeks before and three days post-irradiation. Chrysin prevented the radiation-induced ovarian dysfunction by restoring estradiol levels, preserving the normal ovarian histoarchitecture and combating the follicular loss. Eelectron microscopic analysis showed that the disruption of ultrastructure components due to radiation exposure was hampered by chrysin administration. Mechanistically, chrsyin was able to reduce the levels of the inflammatory markers NF-κB, TNF-α, iNOS and COX-2 in radiation-induced ovarian damage. Chrysin also exhibited potent anti-apoptotic effects against radiation-induced cell death by downregulating the expression of cytochrome c and caspase 3. Radiation obviously induced upregulation of TGF-β protein with subsequent phospholyration and hence activation of downstream mitogen-activated protein kinases (MAPKs); p38 and JNK. Notably, administration of chrysin successfully counteracted these effects. These findings revealed that chrysin may be beneficial in ameliorating radiation-induced POF, predominantly via downregulating TGF-β/MAPK signaling pathways leading subsequently to hindering inflammatory and apoptotic signal transduction pathways implicated in POF.

    Topics: Animals; Apoptosis; Cytoprotection; Female; Flavonoids; Inflammation Mediators; MAP Kinase Signaling System; Primary Ovarian Insufficiency; Radiation Injuries, Experimental; Rats; Rats, Sprague-Dawley; Signal Transduction; Transforming Growth Factor beta; Treatment Outcome

2019
Immunomodulatory effect of human amniotic epithelial cells on restoration of ovarian function in mice with autoimmune ovarian disease.
    Acta biochimica et biophysica Sinica, 2019, Aug-05, Volume: 51, Issue:8

    Autoimmune ovarian disease (AOD) is considered to be a major cause of premature ovarian failure (POF). The immunomodulatory properties of human amniotic epithelial cells (hAECs) have been studied in many disease models. We previously reported that hAECs restored ovarian function in chemotherapy-induced POF mice, but the immunomodulatory mechanism of hAECs is still unclear. To investigate the effect of hAECs on recipient mice, especially on regulatory Treg cells, hAECs and hAEC-conditioned medium (hAEC-CM) were intravenously injected into AOD mice immunized with zona pellucida protein 3 peptides (pZP3). Ovarian function was evaluated through estrous cycle, hormone secretion, follicle development, and cell apoptosis analysis. Immune cells including CD3, CD4, CD8 and Treg cells in the spleens were tested by flow cytometry. To elucidate the effect of hAEC-CM on macrophage function, inflammation model in vitro was established in RAW264.7 cells induced by lipopolysaccharide (LPS). hAECs and hAEC-CM regulated estrous cycles, promoted follicle development, ameliorated cell apoptosis and fibrosis in ovaries of AOD mice. In addition, hAECs significantly reversed the decrease of pZP3-induced Treg cells in the spleens. In vitro, hAEC-CM significantly inhibited the inflammatory reaction induced by LPS in RAW264.7 cells via up-regulating the expression of M2 macrophage genes. Further study demonstrated that hAEC-secreted transforming growth factor-beta and macrophage inhibitory factor played important roles in the macrophage polarization and migration under inflammatory stimulation. Taken together, hAECs restored ovarian function by up-regulating Treg cells in the spleens and reduced the inflammatory reaction via modulating the activated macrophage function in a paracrine manner in the ovaries of AOD mice.

    Topics: Amnion; Animals; Apoptosis; Autoimmune Diseases; Cell Movement; Culture Media, Conditioned; Disease Models, Animal; Epithelial Cells; Female; Granulosa Cells; Humans; Immunohistochemistry; Inflammation; Intramolecular Oxidoreductases; Lipopolysaccharides; Macrophage Migration-Inhibitory Factors; Macrophages; Mice; Ovarian Diseases; Primary Ovarian Insufficiency; RAW 264.7 Cells; Spleen; Transforming Growth Factor beta; Zona Pellucida Glycoproteins

2019
Paracrine effects of human amniotic epithelial cells protect against chemotherapy-induced ovarian damage.
    Stem cell research & therapy, 2017, Nov-28, Volume: 8, Issue:1

    Human amniotic epithelial cells (hAECs) are attractive candidates for regenerative medical therapy, with the potential to replace deficient cells and improve functional recovery after injury. Previous studies have demonstrated that transplantation of hAECs effectively alleviate chemotherapy-induced ovarian damage via inhibiting granulose cells apoptosis in animal models of premature ovarian failure/insufficiency (POF/POI). However, the underlying molecular mechanism accounting for hAECs-mediated ovarian function recovery is not fully understood.. To investigate whether hAECs-secreting cytokines act as molecular basis to attenuate chemotherapy-induced ovarian injury, hAECs or hAEC-conditioned medium (hAEC-CM) was injected into the unilateral ovary of POF/POI mouse. Follicle development was evaluated by H&E staining at 1, 2 months after hAECs or hAEC-CM treatment. In addition, we performed a cytokine array containing 507 human cytokines on hAECs-derived serum-free conditioned medium. Finally, we further investigated whether hAECs could affect chemotherapy-induced apoptosis in primary human granulosa-lutein (hGL) cells and the tube formation of human umbilical vein endothelial cells (hUVECs) via a co-culture system in vitro.. We observed the existence of healthy and mature follicles in ovaries treated with hAECs or hAEC-CM, whereas seriously fibrosis and many atretic follicles were found in the contralateral untreated ovaries of the same mouse. To distinguish cytokines involved in the process of hAECs-restored ovarian function, hAEC-CM was analyzed with a human cytokines array. Results revealed that 109 cytokines in hAEC-CM might participate in a variety of biological processes including apoptosis, angiogenesis, cell cycle and immune response. In vitro experiments, hAECs significantly inhibited chemotherapy-induced apoptosis and activated TGF-β/Smad signaling pathway within primary granulosa-lutein cells in paracrine manner. Furthermore, hAEC-CM was shown to promote angiogenesis in the injured ovaries and enhance the tube formation of human umbilical vein endothelial cells (hUVECs) in co-culture system.. These findings demonstrated that paracrine might be a key pathway in the process of hAECs-mediating ovarian function recovery in animal models of premature ovarian failure/insufficiency (POF/POI).

    Topics: Amnion; Animals; Busulfan; Coculture Techniques; Culture Media, Conditioned; Cyclophosphamide; Epithelial Cells; Female; Gene Expression Profiling; Gene Expression Regulation; Granulosa Cells; Human Umbilical Vein Endothelial Cells; Humans; Mice; Mice, Inbred C57BL; Paracrine Communication; Primary Cell Culture; Primary Ovarian Insufficiency; Protein Array Analysis; Signal Transduction; Smad Proteins; Transforming Growth Factor beta

2017
Human umbilical cord blood-mesenchymal stem cells transplantation renovates the ovarian surface epithelium in a rat model of premature ovarian failure: Possible direct and indirect effects.
    Tissue & cell, 2016, Volume: 48, Issue:4

    This study aimed to isolate mesenchymal stem cells (MSC) from human umbilical cord blood (HCB) and to explore their influence on the ovarian epithelium after paclitaxel-induced ovarian failure. Ninety-five rats were divided into 6 groups: control, paclitaxel, paclitaxel and saline, HCB-MSC-treated for 2 weeks, HCB-MSC-treated for 4 weeks, and HCB-MSC-treated for 6 weeks. HCB cells were studied for CD34, CD44, and Oct ¾ using flow cytometry. Serum levels of FSH and E2 were measured using ELISA, RT-PCR analysis for human gene; beta-actin (ACTB), immunohistochemical analysis for CK 8/18, TGF-ß, PCNA and CASP-3 were performed. We found that ACTB gene was expressed in all rats' ovaries received HCB-MSC. After 4 weeks of transplantation, there was significant reduction in FSH, elevation in E2 levels, stabilization of the surface epithelium morphostasis, an increase in the antral follicle count and increase in integrated densities (ID) of CK 8/18, TGF-ß, and PCNA expressions and decrease in ID of CASP-3 expression. We concluded that HCB-MSC can restore the ovarian function after paclitaxel injection through a direct triggering effect on the ovarian epithelium and/or indirect enrichment of ovarian niche through regulating tissue expression of CK 8/18, TGF-ß and PCNA. These molecules are crucial in regulating folliculogenesis and suppressing CASP-3-induced apoptosis.

    Topics: 12E7 Antigen; Actins; Animals; Apoptosis; Caspase 3; Cord Blood Stem Cell Transplantation; Female; Follicle Stimulating Hormone; Gene Expression Regulation; Humans; Mesenchymal Stem Cell Transplantation; Mesenchymal Stem Cells; Ovary; Paclitaxel; Primary Ovarian Insufficiency; Rats; Transforming Growth Factor beta

2016
Calreticulin is required for development of the cumulus oocyte complex and female fertility.
    Scientific reports, 2015, Sep-21, Volume: 5

    Calnexin (CANX) and calreticulin (CALR) chaperones mediate nascent glycoprotein folding in the endoplasmic reticulum. Here we report that these chaperones have distinct roles in male and female fertility. Canx null mice are growth retarded but fertile. Calr null mice die during embryonic development, rendering indeterminate any effect on reproduction. Therefore, we conditionally ablated Calr in male and female germ cells using Stra8 (mcKO) and Zp3 (fcKO) promoter-driven Cre recombinase, respectively. Calr mcKO male mice were fertile, but fcKO female mice were sterile despite normal mating behavior. Strikingly, we found that Calr fcKO female mice had impaired folliculogenesis and decreased ovulatory rates due to defective proliferation of cuboidal granulosa cells. Oocyte-derived, TGF-beta family proteins play a major role in follicular development and molecular analysis revealed that the normal processing of GDF9 and BMP15 was defective in Calr fcKO oocytes. These findings highlight the importance of CALR in female reproduction and demonstrate that compromised CALR function leads to ovarian insufficiency and female infertility.

    Topics: Animals; Bone Morphogenetic Protein 15; Calnexin; Calreticulin; Cell Differentiation; Cell Proliferation; Cells, Cultured; Cumulus Cells; Endoplasmic Reticulum; Female; Fertility; Growth Differentiation Factor 9; Infertility, Female; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Oocytes; Organ Culture Techniques; Ovarian Follicle; Ovulation; Primary Ovarian Insufficiency; Protein Folding; Transforming Growth Factor beta

2015
Skin-derived mesenchymal stem cells help restore function to ovaries in a premature ovarian failure mouse model.
    PloS one, 2014, Volume: 9, Issue:5

    Skin-derived mesenchymal stem cells (SMSCs) can differentiate into the three embryonic germ layers. For this reason, they are considered a powerful tool for therapeutic cloning and offer new possibilities for tissue therapy. Recent studies showed that skin-derived stem cells can differentiate into cells expressing germ-cell specific markers in vitro and form oocytes in vivo. The idea that SMSCs may be suitable for the treatment of intractable diseases or traumatic tissue damage has attracted attention. To determine the ability of SMSCs to reactivate injured ovaries, a mouse model with ovaries damaged by busulfan and cyclophosphamide was developed and is described here. Female skin-derived mesenchymal stem cells (F-SMSCs) and male skin-derived mesenchymal stem cells (M-SMSCs) from red fluorescence protein (RFP) transgenic adult mice were used to investigate the restorative effects of SMSCs on ovarian function. Significant increases in total body weight and the weight of reproductive organs were observed in the treated animals. Both F-SMSCs and M-SMSCs were shown to be capable of partially restoring fertility in chemotherapy-treated females. Immunostaining with RFP and anti-Müllerian hormone (AMH) antibodies demonstrated that the grafted SMSCs survived, migrated to the recipient ovaries. After SMSCs were administered to the treated mice, real-time PCR showed that the expression levels of pro-inflammatory cytokines TNF-α, TGF-β, IL-8, IL-6, IL-1β, and IFNγ were significantly lower in the ovaries than in the untreated controls. Consistent with this observation, expression of oogenesis marker genes Nobox, Nanos3, and Lhx8 increased in ovaries of SMSCs-treated mice. These findings suggest that SMSCs may play a role within the ovarian follicle microenvironment in restoring the function of damaged ovaries and could be useful in reproductive health.

    Topics: Animals; Disease Models, Animal; Female; Homeodomain Proteins; Interferon-gamma; Interleukin-1beta; Interleukin-6; Interleukin-8; LIM-Homeodomain Proteins; Male; Mesenchymal Stem Cells; Mice; Mice, Transgenic; Oogenesis; Ovarian Follicle; Ovary; Primary Ovarian Insufficiency; RNA-Binding Proteins; Skin; Transcription Factors; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha

2014
The protective effect of granulocyte colony-stimulating factor on endometrium and ovary in a rat model of diabetes mellitus.
    Gynecologic and obstetric investigation, 2014, Volume: 78, Issue:2

    To evaluate the effect of granulocyte colony-stimulating factor (G-CSF) on the endometrium and ovaries in an experimental diabetes mellitus (DM) rat model.. A total of 18 female Sprague-Dawley albino mature rats (8 weeks, 200-220 g) were used in this study. Diabetes was induced by intraperitoneal (i.p.) injection of streptozocin randomly in 12 rats. No drug was administered to the remainder of the rats (control group, group 1, n = 6). The other 12 rats were randomly divided into 2 groups; 1 ml/kg i.p. saline was given as vehicle to group 2 (diabetic nontreated control group, n = 6) and 100 µg/kg/day of i.p. G-CSF was given to group 3 (G-CSF-treated group, n = 6) for 4 weeks. After 4 weeks, blood samples were collected and hysterectomy with bilateral oophorectomy was performed for histopathological examination.. The mean endometrial gland degeneration and stromal fibrosis scores were significantly higher in group 2 compared with groups 1 and 3. Ovarian follicle degeneration, stromal degeneration and stromal fibrosis scores were significantly higher in group 2 compared with groups 1 and 3. Plasma TGF-β and malondialdehyde levels were significantly lower in groups 1 and 3 compared with group 2. Antimüllerian hormone levels were significantly lower in group 2 compared with groups 1 and 3.. Glucose toxicity occurred severely in the ovaries and endometrium of the DM rats. After G-CSF treatment, ovarian and endometrial injury and fibrosis scores decreased significantly. The effects of G-CSF in rat models give hope to improved treatment of human DM complications such as premature ovarian failure and endometrial dysfunction.

    Topics: Animals; Diabetes Mellitus, Experimental; Endometrium; Female; Granulocyte Colony-Stimulating Factor; Malondialdehyde; Ovarian Diseases; Ovary; Primary Ovarian Insufficiency; Rats; Rats, Sprague-Dawley; Transforming Growth Factor beta; Uterine Diseases

2014
A large-scale candidate gene association study of age at menarche and age at natural menopause.
    Human genetics, 2010, Volume: 128, Issue:5

    Recent genome-wide association (GWA) studies have identified several novel genetic loci associated with age at menarche and age at natural menopause. However, the stringent significance threshold used in GWA studies potentially led to false negatives and true associations may have been overlooked. Incorporating biologically relevant information, we examined whether common genetic polymorphisms in candidate genes of nine groups of biologically plausible pathways and related phenotypes are associated with age at menarche and age at natural menopause. A total of 18,862 genotyped and imputed single nucleotide polymorphisms (SNPs) in 278 genes were assessed for their associations with these two traits among a total of 24,341 women from the Nurses' Health Study (NHS, N = 2,287) and the Women's Genome Health Study (WGHS, N = 22,054). Linear regression was used to assess the marginal association of each SNP with each phenotype. We adjusted for multiple testing within each gene to identify statistically significant SNP associations at the gene level. To evaluate the overall evidence for an excess of statistically significant gene associations over the proportion expected by chance, we applied a one-sample test of proportion to each group of candidate genes. The steroid-hormone metabolism and biosynthesis pathway was found significantly associated with both age at menarche and age at natural menopause (P = 0.040 and 0.011, respectively). In addition, the group of genes associated with precocious or delayed puberty was found significantly associated with age at menarche (P = 0.013), and the group of genes involved in premature ovarian failure with age at menopause (P = 0.025).

    Topics: Adolescent; Age Factors; Age of Onset; Child; Female; Genetic Association Studies; Genetic Predisposition to Disease; Genotype; Gonadal Steroid Hormones; Humans; Linear Models; Menarche; Menopause; Middle Aged; Nurses; Obesity; Phenotype; Polycystic Ovary Syndrome; Polymorphism, Single Nucleotide; Primary Ovarian Insufficiency; Puberty, Delayed; Puberty, Precocious; Signal Transduction; Smoking; Somatomedins; Thrombophilia; Tobacco Use Disorder; Transforming Growth Factor beta; Women's Health

2010
[Innovative genetic, clinical and endocrinologic aspects in young females affected by premature ovarian insufficiency].
    Minerva ginecologica, 2010, Volume: 62, Issue:6

    Heterogeneity premature ovarian insufficiency (POI) is one of the reasons why there are different causes that contribute in determining this type of hormonal disorder. Although the causes have already been established for many types of premature ovarian failure, are still uncertain causes in most cases of idiopathic forms, despite the description of several candidate genes, including BMP-15 gene. The gene under study is precisely the BMP-15, which is part of the superfamily of Transforming Growth Factors-beta or the TGF-β, which also belong to the growth differentiation factors (GDFs).. This study examined a sample of Sicilian women suffering from POI, carefully selected according to their age, since in these cases, the genetic factor probably has a greater impact.. Identify a mutant gene that causes ovarian failure may be important to make a diagnosis that can predict the possible future development of the disease. The outcome of the studies, however, has not found the gene in question, but it is hypothesized that this may be a direct consequence of the limited amount of women that was done the study, a case which may be rebutted by increasing the number of patients.

    Topics: Adolescent; Adult; Bone Morphogenetic Protein 15; Case-Control Studies; Female; Genetic Linkage; Genetic Markers; Genetic Predisposition to Disease; Genome, Human; Humans; Mutation; Polymorphism, Genetic; Primary Ovarian Insufficiency; Transforming Growth Factor beta

2010
Bone morphogenetic protein 15 (BMP15) acts as a BMP and Wnt inhibitor during early embryogenesis.
    The Journal of biological chemistry, 2009, Sep-18, Volume: 284, Issue:38

    Bone morphogenetic protein 15 (BMP15) belongs to an unusual subgroup of the transforming growth factor beta (TGFbeta) superfamily of signaling ligands as it lacks a key cysteine residue in the mature region required for proper intermolecular dimerization. Naturally occurring BMP15 mutation leads to early ovarian failure in humans, and BMP15 has been shown to activate the Smad1/5/8 pathway in that context. Despite its important role in germ cell specification, the embryological function of BMP15 remains unknown. Surprisingly, we find that during early Xenopus embryogenesis BMP15 acts solely as an inhibitor of the Smad1/5/8 pathway and the Wnt pathway. BMP15 gain-of-function leads to embryos with secondary ectopic heads and to direct neural induction in intact explants. BMP15 inhibits BMP4-mediated epidermal induction in dissociated explants. BMP15 strongly inhibits BRE response induced by BMP4 and blocks phosphorylation and activation of Smad1/5/8 MH2-domain. Mechanistically, BMP15 protein specifically interacts with BMP4 protein, suggesting inhibition upstream of receptor binding. Loss-of-function experiments using morpholinos or a naturally occurring human BMP15 dominant-negative mutant (BMP15-Y235C) leads to embryos lacking head. BMP15-Y235C also eliminates the inhibitory activity of BMP15 on BRE (BMP-responsive element). Finally, we show that BMP15 inhibits the canonical branch of the Wnt pathway, upstream of beta-catenin. We, thus, demonstrate that BMP15 is necessary and sufficient for the specification of dorso-anterior structures and highlight novel mechanisms of BMP15 function that strongly suggest a reinterpretation of its function in ovaries specially for ovarian failure.

    Topics: Amino Acid Substitution; Animals; Bone Morphogenetic Protein 15; Bone Morphogenetic Protein 4; Embryo, Nonmammalian; Embryonic Development; Female; Humans; Mutation, Missense; Phosphorylation; Primary Ovarian Insufficiency; Protein Structure, Tertiary; Signal Transduction; Smad Proteins; Transforming Growth Factor beta; Wnt Proteins; Xenopus laevis; Xenopus Proteins

2009
Actions of anti-Mullerian hormone on the ovarian transcriptome to inhibit primordial to primary follicle transition.
    Reproduction (Cambridge, England), 2007, Volume: 134, Issue:2

    The oocytes found within the primordial follicles of mammalian ovaries remain quiescent for months to years until they receive the appropriate signals to undergo the primordial to primary follicle transition and initiate folliculogenesis. The molecular mechanisms and extracellular signaling factors that regulate this process remain to be fully elucidated. The current study investigates the mechanisms utilized by anti-Müllerian hormone (AMH; i.e. Müllerian inhibitory substance) to inhibit the primordial to primary follicle transition. Ovaries from 4-day-old rats were placed into organ culture and incubated in the absence or presence of AMH, either alone or in combination with known stimulators of follicle transition, including basic fibroblast growth factor (bFGF), kit ligand (KITL), or keratinocyte growth factor (KGF). Following 10 days of culture, the ovaries were sectioned, stained, and morphologically evaluated to determine the percentage of primordial versus developing follicles. As previously demonstrated, AMH treatment decreased primordial to primary follicle transition. Interestingly, AMH inhibited the stimulatory actions of KITL, bFGF, and KGF. Therefore, AMH can inhibit the basal and stimulated development of primordial follicles. To investigate the mechanism of AMH actions, the influence AMH has on the ovarian transcriptome was analyzed. AMH treatment when compared with controls was found to alter the expression of 707 genes. The overall effect of AMH exposure is to decrease the expression of stimulatory factors, increase the expression of inhibitory factors, and regulate cellular pathways (e.g. transforming growth factor beta signaling pathway) that result in the inhibition of primordial follicle development. Analysis of the regulatory factors and cellular pathways altered by AMH provides a better understanding of the molecular control of primordial follicle development.

    Topics: Animals; Animals, Newborn; Anti-Mullerian Hormone; Female; Fibroblast Growth Factor 2; Fibroblast Growth Factor 7; Gene Expression; Gene Expression Profiling; Gene Expression Regulation, Developmental; Humans; MAP Kinase Signaling System; Oligonucleotide Array Sequence Analysis; Oocytes; Organ Culture Techniques; Ovarian Follicle; Ovary; Primary Ovarian Insufficiency; Rats; Rats, Sprague-Dawley; Recombinant Proteins; Signal Transduction; Stem Cell Factor; Transcription, Genetic; Transforming Growth Factor beta

2007
[Current status and prospects of the study on autocrine and paracrine regulation of ovarian function].
    Zhonghua fu chan ke za zhi, 1997, Volume: 32, Issue:10

    Topics: Epidermal Growth Factor; Female; Humans; Hypothalamo-Hypophyseal System; Insulin-Like Growth Factor I; Ovary; Primary Ovarian Insufficiency; Transforming Growth Factor beta

1997