Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (104)

Search Parameters:
Keywords = ovarian folliculogenesis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 6745 KiB  
Article
Ovarian Transcriptome Profile from Egg-Laying Period to Incubation Period of Changshun Green-Shell Laying Hens
by Zhi Chen and Di Wen
Genes 2025, 16(4), 394; https://doi.org/10.3390/genes16040394 - 29 Mar 2025
Viewed by 148
Abstract
Background/Objectives: The Changshun green-shell laying hen with a strong broodiness is a Chinese indigenous chicken breed. Little is known about the mechanisms responsible for the ovary development of Changshun green-shell laying hens from the egg-laying period (LP) to the incubation period (BP). Methods: [...] Read more.
Background/Objectives: The Changshun green-shell laying hen with a strong broodiness is a Chinese indigenous chicken breed. Little is known about the mechanisms responsible for the ovary development of Changshun green-shell laying hens from the egg-laying period (LP) to the incubation period (BP). Methods: A total of six hens were selected from LP (n = three) and BP (n = three) at 28 weeks old. The RNA sequencing (RNA-seq) of ovaries from hens in LP and BP groups was performed to identify candidate genes and pathways associated with broodiness. Results: We identified 1650 differently expressed genes (DEGs), including 429 up-regulated and 1221 down-regulated DEGs, in chicken ovaries between LP and BP groups. Gene ontology term (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed that these DEGs were mainly involved in the pathways related to follicle development in chicken ovaries, including focal adhesion, the MAPK signaling pathway, and the FoxO signaling pathway, and vascular smooth muscle contraction, ECM–receptor interaction, and the GnRH signaling pathway were down-regulated in incubating ovaries. Eight candidate genes (EGFR, VEGFRKDRL, FLT1, KDR, PDGFRA, TEK, KIT and FGFR3) related to angiogenesis, folliculogenesis, steroidogenesis and oogenesis in ovaries were suggested to play important roles in the ovarian development of Changshun hens during the transition from LP to BP. Conclusions: This study identified a range of genes and several pathways that may be involved in regulating the broodiness of Changshun green-shell laying hens. These data are helpful to further enrich our understanding of the mechanism of incubation behaviour in chickens. Full article
(This article belongs to the Section Animal Genetics and Genomics)
Show Figures

Figure 1

16 pages, 6378 KiB  
Article
Impact of mRNA and Inactivated COVID-19 Vaccines on Ovarian Reserve
by Enes Karaman, Adem Yavuz, Erol Karakas, Esra Balcioglu, Busra Karaca, Hande Nur Doganay, Koray Gorkem Sacinti and Orhan Yildiz
Vaccines 2025, 13(4), 345; https://doi.org/10.3390/vaccines13040345 - 24 Mar 2025
Viewed by 201
Abstract
Objectives: This study aimed to elucidate the effects of messenger RNA (mRNA) and inactivated coronavirus disease 2019 (COVID-19) vaccines on ovarian histology and reserve in rats. Methods: Thirty female Wistar albino rats, aged 16–24 weeks, were randomly divided into three groups [...] Read more.
Objectives: This study aimed to elucidate the effects of messenger RNA (mRNA) and inactivated coronavirus disease 2019 (COVID-19) vaccines on ovarian histology and reserve in rats. Methods: Thirty female Wistar albino rats, aged 16–24 weeks, were randomly divided into three groups (n = 10): control, mRNA vaccine, and inactivated vaccine groups. Each vaccine group received two doses (on day 0 and day 28) at human-equivalent doses. Four weeks post-second vaccination, ovarian tissues were harvested for analysis. Results: Immunohistochemical analysis was performed to evaluate the expression of transforming growth factor beta-1 (TGF-β1), vascular endothelial growth factor (VEGF), caspase-3, and anti-Müllerian hormone (AMH) in ovarian follicles. Both vaccines induced significant increases in TGF-β1, VEGF, and caspase-3 expression, with more pronounced effects in the mRNA vaccine group. Conversely, AMH expression in the granulosa cells of primary, secondary, and antral follicles showed marked reductions (p < 0.001). The counts of primordial, primary, and secondary follicles decreased significantly in the inactivated vaccine group relative to controls and further in the mRNA vaccine group compared to the inactivated group (p < 0.001). Additionally, the mRNA vaccine group exhibited a decrease in antral and preovulatory follicles and an increase in atretic follicles compared to the other groups (p < 0.05). The serum AMH level was diminished with the mRNA vaccination in comparison with the control and inactivated groups. Conclusions: Our findings suggest that both mRNA and inactivated COVID-19 vaccines may detrimentally impact ovarian reserve in rats, primarily through accelerated follicular loss and alterations in apoptotic pathways during folliculogenesis. Given these observations in a rat model, further investigations into the vaccines’ effects on human ovarian reserve are needed. Full article
Show Figures

Figure 1

13 pages, 826 KiB  
Review
Mechanisms of Endometrioma-Mediated Ovarian Damage: Myths and Facts
by Pınar Özcan, Bulut Varlı, Ertan Sarıdoğan, Engin Oral, Muhammed Mabrouk, Taner Usta and Alin Stefan Constantin
J. Clin. Med. 2025, 14(7), 2147; https://doi.org/10.3390/jcm14072147 - 21 Mar 2025
Viewed by 535
Abstract
Ovarian endometriomas (OEMs), cystic formations within the ovaries, are a significant manifestation of endometriosis and present in 20–40% of affected women. Despite extensive research, the pathogenesis of endometriosis remains unclear, with retrograde menstruation, coelomic metaplasia, and lymphatic dissemination being proposed mechanisms. OEMs negatively [...] Read more.
Ovarian endometriomas (OEMs), cystic formations within the ovaries, are a significant manifestation of endometriosis and present in 20–40% of affected women. Despite extensive research, the pathogenesis of endometriosis remains unclear, with retrograde menstruation, coelomic metaplasia, and lymphatic dissemination being proposed mechanisms. OEMs negatively impact ovarian function by reducing the ovarian reserve, disrupting folliculogenesis, and altering the ovarian microenvironment through oxidative stress, inflammation, and fibrosis. Elevated reactive oxygen species (ROS) accelerate follicular atresia, and extracellular matrix remodeling contributes to ovarian damage, while immune dysregulation and cytokine imbalances further exacerbate the condition. The presence of OEMs does not significantly affect live birth rates in in vitro fertilization (IVF) treatments, despite potential reductions in the quality and quantity of oocytes. However, their surgical excision compromises the ovarian reserve. This review highlights the complex mechanisms by which OEMs impair ovarian function and emphasizes the need for further research to develop strategies that mitigate these effects, ultimately improving reproductive outcomes for women with endometriomas. Full article
Show Figures

Figure 1

23 pages, 3754 KiB  
Article
Depletion of Fkbp5 Protects Against the Rapid Decline in Ovarian Reserve Induced by Prenatal Stress in Female Offspring of Wild-Type Mice
by Monica Moore, Busra Cetinkaya-Un, Papri Sarkar, Umit A. Kayisli, Nihan Semerci-Gunay, Michael Teng, Charles J. Lockwood and Ozlem Guzeloglu-Kayisli
Int. J. Mol. Sci. 2025, 26(6), 2471; https://doi.org/10.3390/ijms26062471 - 10 Mar 2025
Viewed by 248
Abstract
Prenatal stress (PNS) impairs offspring ovarian development by exerting negative long-term effects on postnatal ovarian function and folliculogenesis. FKBP51 is a stress-responsive protein that inhibits glucocorticoid and progesterone receptors. We hypothesize that FKBP51 contributes to impaired ovarian development and folliculogenesis induced by PNS. [...] Read more.
Prenatal stress (PNS) impairs offspring ovarian development by exerting negative long-term effects on postnatal ovarian function and folliculogenesis. FKBP51 is a stress-responsive protein that inhibits glucocorticoid and progesterone receptors. We hypothesize that FKBP51 contributes to impaired ovarian development and folliculogenesis induced by PNS. Timed-pregnant Fkbp5+/+ (wild-type) and Fkbp5−/− (knockout) mice were randomly assigned to either the undisturbed (nonstress) or PNS group, with exposure to maternal restraint stress from embryonic days 8 to 18. Ovaries from the offspring were harvested and stained, and follicles were counted according to their stages. Ovarian expressions of FKBP51 were evaluated by immunohistochemistry and Fkbp5 and steroidogenic enzymes were evaluated by qPCR. Compared to controls, Fkbp5+/+ PNS offspring had increased peripubertal primordial follicle atresia and fewer total follicles in the adult and middle-aged groups. In adult Fkbp5+/+ offspring, PNS elevated FKBP51 levels in granulosa cells of primary to tertiary follicles. Our results suggest that PNS administration increased FKBP51 levels, depleted the ovarian reserve, and dysregulated ovarian steroid synthesis. However, these PNS effects were tolerated in Fkbp5−/− mice, supporting the conclusion that FKBP51 contributes to reduced ovarian reserve induced by PNS. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

21 pages, 1857 KiB  
Review
The Ovary as a Target Organ for New Generation Bisphenols Toxicity
by Paulina Głód, Joanna Smoleniec, Weronika Marynowicz, Justyna Gogola-Mruk and Anna Ptak
Toxics 2025, 13(3), 164; https://doi.org/10.3390/toxics13030164 - 26 Feb 2025
Viewed by 437
Abstract
Bisphenols (BPs) are a group of organic compounds used extensively in plastics, coatings, and epoxy resins; they have been of concern recently due to their endocrine-disrupting effects. Among these, bisphenol A (BPA) is the most studied. Regulatory measures, such as the ban on [...] Read more.
Bisphenols (BPs) are a group of organic compounds used extensively in plastics, coatings, and epoxy resins; they have been of concern recently due to their endocrine-disrupting effects. Among these, bisphenol A (BPA) is the most studied. Regulatory measures, such as the ban on BPA use in baby bottles by the European Union and its restricted use in thermal paper, reflect the growing awareness of the health risks of BPA. To mitigate these risks, analogs such as bisphenol S (BPS), bisphenol F (BPF), and others (BPAF, BPAP, BPB, BPP, BPZ) have been developed as alternatives. Despite their intended safety, these analogs have been detected in environmental media, including indoor dust and thermal receipt paper, as well as in human biological samples. Studies report their presence in urine at levels comparable to BPA, with BPS and BPF found in 78% and 55% of samples, respectively. In addition, BPs have been found in human follicular fluid (FF) at concentrations that could exert some paracrine effects on ovarian function and reproductive health. With the increased global production of BPs, occupational exposure and environmental contamination also increase. This review summarizes what is currently known about the effects of BPs on the ovary and the mechanisms by which PBs exert ovarian toxicity, with a particular focus on oogenesis, folliculogenesis, and steroidogenesis. Further, this review emphasizes their influence on reproductive functions and the need for further biosafety evaluations. Full article
(This article belongs to the Special Issue Endocrine-Disrupting Chemicals and Reproductive Toxicology)
Show Figures

Graphical abstract

16 pages, 16488 KiB  
Article
Peritoneal Endometriosis Impairs Ovarian Reserve and Increases Atresia in a Rat Model
by Analía Ricci, Tatiana Bengochea, Carla Olivares, Sofía del Valle, Julieta Simone, Kristina Gemzell-Danielsson, Rosa Inés Barañao, Gabriela Meresman and Mariela Bilotas
Biomedicines 2025, 13(2), 348; https://doi.org/10.3390/biomedicines13020348 - 3 Feb 2025
Viewed by 648
Abstract
Background/Objectives: Endometriosis has a marked impact on fertility, although the mechanisms behind this relationship remain poorly understood, particularly in cases without significant anatomical distortions or in the context of ovarian endometriomas. This study aimed to investigate the effect of peritoneal endometriosis on ovarian [...] Read more.
Background/Objectives: Endometriosis has a marked impact on fertility, although the mechanisms behind this relationship remain poorly understood, particularly in cases without significant anatomical distortions or in the context of ovarian endometriomas. This study aimed to investigate the effect of peritoneal endometriosis on ovarian function by assessing ovarian reserve and apoptosis. Methods: Peritoneal endometriosis was surgically induced in Sprague Dawley rats through the autotransplantation of uterine fragments onto the bowel mesothelium. One month post-surgery, ovarian structures were counted, follicle and corpora lutea apoptosis was evaluated by TUNEL, and apoptotic-related protein expression in ovaries was assessed by Western blot. Additionally, a co-culture system using 12Z endometriotic and KGN granulosa cell lines was utilized to evaluate gene expression by RT-qPCR. Results: Rats with peritoneal endometriosis exhibited a significant reduction in ovarian structures characterized by a low number of total follicles, particularly primordial, primary, preantral, and late-antral follicles. Consistently, AMH protein expression was decreased in ovaries in the presence of endometriosis. In addition, this disease led to a significant increase in late-antral follicles that were TUNEL-positive and in the number of apoptotic cells in corpora lutea, indicating higher apoptosis in endometriosis ovaries. Concomitantly, the altered expression of apoptosis-related proteins was observed, with increased procaspase 3 and decreased BCL-2 expression. In addition, KGN granulosa cells co-cultured with 12Z endometriotic cells displayed reduced KITLG mRNA expression and increased AMHR2 mRNA expression. Conclusions: Peritoneal endometriosis significantly impairs ovarian health by disrupting folliculogenesis, reducing ovarian reserve, and increasing apoptosis, potentially accelerating ovarian aging and contributing to infertility. These results underscore the need for further research to identify the molecular pathways involved and to develop targeted therapeutic strategies. Full article
(This article belongs to the Special Issue Molecular and Clinical Aspects of Endometriosis Pathophysiology)
Show Figures

Figure 1

16 pages, 592 KiB  
Review
Dual-Double Stem Cell Ovarian Therapy: A Comprehensive Approach in Regenerative Medicine
by Aleksandar Ljubić, Marija Dinić, Dajana Švraka and Svetlana Vujović
Int. J. Mol. Sci. 2025, 26(1), 69; https://doi.org/10.3390/ijms26010069 - 25 Dec 2024
Viewed by 1327
Abstract
Dual-double stem cell therapy, which integrates mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs), represents a cutting-edge approach in regenerative medicine, particularly for conditions such as ovarian decline, premature ovarian insufficiency (POI), and induced ovarian failure. This therapy leverages the unique properties [...] Read more.
Dual-double stem cell therapy, which integrates mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs), represents a cutting-edge approach in regenerative medicine, particularly for conditions such as ovarian decline, premature ovarian insufficiency (POI), and induced ovarian failure. This therapy leverages the unique properties of MSCs and HSCs, enhancing tissue repair, immune modulation, and overall regenerative outcomes. MSCs, known for their ability to differentiate into various cell types, provide a supportive microenvironment and secrete bioactive molecules that promote angiogenesis and reduce inflammation. HSCs, crucial for hematopoiesis and immune function, further enhance this environment by supporting hematopoietic processes and immune regulation. Clinical evidence increasingly supports the effectiveness of stem cell therapy in ovarian regeneration. Studies have demonstrated improved folliculogenesis, normalization of hormone profiles, and successful pregnancies in patients with POI. Furthermore, recent clinical trials in various medical fields underline the superior potential of dual-double therapy compared to monotherapies involving MSCs or HSCs alone, enhancing tissue repair and functional outcomes. However, despite these benefits, the therapy presents risks that require careful consideration. For autologous MSC therapy involving expanded cell populations, risks include tumorigenic potential, with evidence of sarcoma formation in certain cases of cultured MSCs. In contrast, autologous non-expanded MSC and HSC therapies may be limited by low cell yields, potentially compromising therapeutic efficacy. Additionally, non-expanded HSC therapy poses risks of insufficient cell numbers for successful engraftment and delayed immune reconstitution. These considerations underscore the importance of quality control and rigorous screening to optimize safety and efficacy. This article explores the mechanisms of action, clinical applications, and potential complications of dual-double stem cell therapy, underscoring the need for continued research and optimized protocols to enhance safety and outcomes in ovarian insufficiency and related conditions, offering new hope for affected women. Full article
(This article belongs to the Special Issue New Insights into Human Mesenchymal Stem Cells)
Show Figures

Figure 1

29 pages, 673 KiB  
Review
Chemerin and Polycystic Ovary Syndrome: A Comprehensive Review of Its Role as a Biomarker and Therapeutic Target
by Stefano Palomba, Giuseppe Seminara, Flavia Costanzi, Donatella Caserta and Antonio Aversa
Biomedicines 2024, 12(12), 2859; https://doi.org/10.3390/biomedicines12122859 - 16 Dec 2024
Viewed by 1182
Abstract
Background: Chemerin, an adipokine implicated in inflammatory, metabolic, and adipogenic processes, has been detected in high serum concentration in women with polycystic ovary syndrome (PCOS) and seems to play a role in PCOS pathogenesis. Moreover, at present, no comprehensive and critical document is [...] Read more.
Background: Chemerin, an adipokine implicated in inflammatory, metabolic, and adipogenic processes, has been detected in high serum concentration in women with polycystic ovary syndrome (PCOS) and seems to play a role in PCOS pathogenesis. Moreover, at present, no comprehensive and critical document is available in the literature on this topic. The aim of the current study was to comprehensively review the latest available data to confirm the evidence about the association between chemerin and PCOS, highlighting its potential role as an upcoming biomarker and therapeutic target. Methods: A search in the literature of studies published between 2019 and 2024 was conducted using PubMed, Cochrane Library, and Web of Science, focusing on research related to chemerin, PCOS, and PCOS-related features, comorbidities, and complications. A qualitative structured synthesis of key findings was performed according to the specific thematic areas selected, including and discussing clinical data on women with PCOS and experimental studies in humans and animal models of PCOS. Results: Available data confirm increased serum levels of chemerin in women with PCOS compared with controls, independent of obesity and body mass index. Chemerin is associated with insulin resistance, hyperandrogenism, and ovarian dysfunction in PCOS individuals, inhibiting folliculogenesis and steroidogenesis. Experimental animal models underscore chemerin’s regulatory roles through its receptors within the hypothalamic–pituitary–ovarian axis and peripheral tissues. High systemic levels of chemerin in PCOS may also be related to the increased risk of pregnancy complications, especially gestational diabetes mellitus and preeclampsia. Conclusions: The current review study highlights the role of chemerin in PCOS pathophysiology, severity, and associated comorbidities and complications, assessing its value as a future biomarker and foreshadowing its potential as a therapeutic target. Full article
(This article belongs to the Special Issue The Role of Chemerin in Human Disease2nd Edition)
Show Figures

Graphical abstract

19 pages, 836 KiB  
Review
Granulosa Cells: Central Regulators of Female Fertility
by Luis Fernando Schütz and Isadora M. Batalha
Endocrines 2024, 5(4), 547-565; https://doi.org/10.3390/endocrines5040040 - 27 Nov 2024
Cited by 1 | Viewed by 2249
Abstract
Background: Granulosa cells are somatic cells within the ovarian follicle. As the primary site of estradiol production, they are critical regulators of several aspects of female reproduction. This review aims to provide an overview of the physiology of mammalian granulosa cells and their [...] Read more.
Background: Granulosa cells are somatic cells within the ovarian follicle. As the primary site of estradiol production, they are critical regulators of several aspects of female reproduction. This review aims to provide an overview of the physiology of mammalian granulosa cells and their importance for female fertility. Methods: the literature about the function and regulation of granulosa cells was reviewed. Results: a comprehensive summary and discussion of the role of granulosa cells on ovarian steroidogenesis and folliculogenesis, as well as factors that control granulosa cells function, are presented. Conclusion: The functions of granulosa cells are regulated by a plethora of intra- and extra-ovarian factors via autocrine, paracrine, and endocrine pathways, which creates a complex regulatory network. A comprehensive understanding of granulosa cells’ physiology is vital for the development of innovative strategies to enhance reproductive outcomes in several species. Full article
(This article belongs to the Section Female Reproductive System and Pregnancy Endocrinology)
Show Figures

Figure 1

31 pages, 5407 KiB  
Review
Can Mammalian Reproductive Health Withstand Massive Exposure to Polystyrene Micro- and Nanoplastic Derivatives? A Systematic Review
by Chiara Camerano Spelta Rapini, Chiara Di Berardino, Alessia Peserico, Giulia Capacchietti and Barbara Barboni
Int. J. Mol. Sci. 2024, 25(22), 12166; https://doi.org/10.3390/ijms252212166 - 13 Nov 2024
Cited by 4 | Viewed by 2147
Abstract
The widespread use of plastics has increased environmental pollution by micro- and nanoplastics (MNPs), especially polystyrene micro- and nanoplastics (PS-MNPs). These particles are persistent, bioaccumulative, and linked to endocrine-disrupting toxicity, posing risks to reproductive health. This review examines the effects of PS-MNPs on [...] Read more.
The widespread use of plastics has increased environmental pollution by micro- and nanoplastics (MNPs), especially polystyrene micro- and nanoplastics (PS-MNPs). These particles are persistent, bioaccumulative, and linked to endocrine-disrupting toxicity, posing risks to reproductive health. This review examines the effects of PS-MNPs on mammalian reproductive systems, focusing on oxidative stress, inflammation, and hormonal imbalances. A comprehensive search in the Web of Science Core Collection, following PRISMA 2020 guidelines, identified studies on the impact of PS-MNPs on mammalian fertility, including oogenesis, spermatogenesis, and folliculogenesis. An analysis of 194 publications revealed significant reproductive harm, such as reduced ovarian size, depleted follicular reserves, increased apoptosis in somatic cells, and disrupted estrous cycles in females, along with impaired sperm quality and hormonal imbalances in males. These effects were linked to endocrine disruption, oxidative stress, and inflammation, leading to cellular and molecular damage. Further research is urgently needed to understand PS-MNPs toxicity mechanisms, develop interventions, and assess long-term reproductive health impacts across generations, highlighting the need to address these challenges given the growing environmental exposure. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

21 pages, 2693 KiB  
Review
Apigenin as a Promising Agent for Enhancing Female Reproductive Function and Treating Associated Disorders
by Alexander V. Sirotkin and Abdel Halim Harrath
Biomedicines 2024, 12(10), 2405; https://doi.org/10.3390/biomedicines12102405 - 21 Oct 2024
Cited by 1 | Viewed by 2549
Abstract
Apigenin is an organic flavonoid abundant in some plants such as parsley, chamomile, or celery. Recently, it has been investigated for several of its pharmacological characteristics, such as its ability to act as an antioxidant, reduce inflammation, and inhibit the growth of cancer [...] Read more.
Apigenin is an organic flavonoid abundant in some plants such as parsley, chamomile, or celery. Recently, it has been investigated for several of its pharmacological characteristics, such as its ability to act as an antioxidant, reduce inflammation, and inhibit the growth of cancer cells. The purpose of this review is to provide a summary of the existing knowledge regarding the effects of apigenin on female reproductive systems and its dysfunctions. Apigenin can influence reproductive processes by regulating multiple biological events, including oxidative processes, cell proliferation, apoptosis, cell renewal and viability, ovarian blood supply, and the release of reproductive hormones. It could stimulate ovarian folliculogenesis, as well as ovarian and embryonal cell proliferation and viability, which can lead to an increase in fertility and influence the release of reproductive hormones, which may exert its effects on female reproductive health. Furthermore, apigenin could inhibit the activities of ovarian cancer cells and alleviate the pathological changes in the female reproductive system caused by environmental pollutants, harmful medications, cancer, polycystic ovarian syndrome, ischemia, as well as endometriosis. Therefore, apigenin may have potential as a biostimulator for female reproductive processes and as a therapeutic agent for certain reproductive diseases. Full article
Show Figures

Figure 1

14 pages, 2840 KiB  
Article
Regulation of Bone Morphogenetic Protein Receptor Type II Expression by FMR1/Fragile X Mental Retardation Protein in Human Granulosa Cells in the Context of Poor Ovarian Response
by Xuan Phuoc Nguyen, Adriana Vilkaite, Ulrike Bender, Jens E. Dietrich, Katrin Hinderhofer, Thomas Strowitzki and Julia Rehnitz
Int. J. Mol. Sci. 2024, 25(19), 10643; https://doi.org/10.3390/ijms251910643 - 3 Oct 2024
Viewed by 1205
Abstract
Fragile X mental retardation protein (FMRP) is a translational repressor encoded by FMR1. It targets bone morphogenetic protein receptor type II (BMPR2), which regulates granulosa cell (GC) function and follicle development. However, whether this interaction affects folliculogenesis remains unclear. Therefore, this study [...] Read more.
Fragile X mental retardation protein (FMRP) is a translational repressor encoded by FMR1. It targets bone morphogenetic protein receptor type II (BMPR2), which regulates granulosa cell (GC) function and follicle development. However, whether this interaction affects folliculogenesis remains unclear. Therefore, this study investigated the potential effect of FMRP-BMPR2 dysregulation in ovarian reserves and infertility. COV434 cells and patient-derived GCs were used to evaluate FMRP and BMPR2 expression. Similarly, FMR1, BMPR2, LIMK1, and SMAD expression were evaluated in GCs with normal (NOR) and poor (POR) ovarian responses. FMRP and BMPR2 were expressed in both cell types. They were co-localized to the nuclear membrane of COV434 cells and cytoplasm of primary GCs. FMR1 silencing increased the mRNA and protein levels of BMPR2. However, the mRNA levels of FMR1 and BMPR2 were significantly lower in the POR group. FMR1 and BMPR2 levels were strongly positively correlated in the NOR group but weakly correlated in the POR group. Additionally, SMAD9 expression was significantly reduced in the POR group. This study highlights the crucial role of FMR1/FMRP in the regulation of BMPR2 expression and its impact on ovarian function. These findings indicate that the disruption of FMRP-BMPR2 interactions may cause poor ovarian responses and infertility. Full article
(This article belongs to the Special Issue Research on Transcriptional Regulation in Reproductive Biology)
Show Figures

Figure 1

15 pages, 3292 KiB  
Article
Encapsulation of Bovine Primordial Follicles in Rigid Alginate Does Not Affect Growth Dynamics
by Kathryn L. McElhinney, Erin E. Rowell and Monica M. Laronda
Bioengineering 2024, 11(7), 734; https://doi.org/10.3390/bioengineering11070734 - 19 Jul 2024
Cited by 1 | Viewed by 1239
Abstract
The only fertility preservation and subsequent restoration option for many patients facing gonadotoxic treatments is ovarian tissue cryopreservation and transplantation. While this process is successful for some, there is significant room for improvement to extend the life of the transplant and to make [...] Read more.
The only fertility preservation and subsequent restoration option for many patients facing gonadotoxic treatments is ovarian tissue cryopreservation and transplantation. While this process is successful for some, there is significant room for improvement to extend the life of the transplant and to make it safe for patients that may have metastatic disease within their ovarian tissue. We need a deeper understanding of how the physical properties of the ovarian microenvironment may affect folliculogenesis to engineer an environment that supports isolated follicles and maintains primordial follicle quiescence. Bovine ovaries were used here as a monovulatory model of folliculogenesis to examine the effects of primordial follicle activation and growth under different physical conditions. We found that there were no differences in activation, growth or survival when primordial follicles were cultured in isolation or in situ (remaining in the tissue) under two significantly differently rigid alginate gels. To determine if the extra rigid environment did not affect activation in isolated follicles due to an immediate activation event, we used 5-ethynyl-2′-deoxyuridine (EdU) to track follicle activation during the isolation process. We identified EdU incorporation in granulosa cells after primordial follicles were isolated from the surrounding extracellular matrix (ECM). These findings support that isolation of primordial follicles from the ECM is an activating event and that the differentially rigid environments assessed here had no effect on follicle growth. Further work is needed to suppress activation in primordial follicles to maintain the ovarian reserve and extend the life of an ovarian tissue transplant. Full article
(This article belongs to the Special Issue Bioengineering Technologies to Advance Reproductive Health)
Show Figures

Figure 1

14 pages, 1415 KiB  
Review
Gonadotropin Activity during Early Folliculogenesis and Implications for Polycystic Ovarian Syndrome and Premature Ovarian Insufficiency: A Narrative Review
by Salvatore Longobardi, Francesca Gioia Klinger, Wenjing Zheng, Maria Rosaria Campitiello, Thomas D’Hooghe and Antonio La Marca
Int. J. Mol. Sci. 2024, 25(14), 7520; https://doi.org/10.3390/ijms25147520 - 9 Jul 2024
Cited by 1 | Viewed by 2566
Abstract
Female fertility depends on the ovarian reserve of follicles, which is determined at birth. Primordial follicle development and oocyte maturation are regulated by multiple factors and pathways and classified into gonadotropin-independent and gonadotropin-dependent phases, according to the response to gonadotropins. Folliculogenesis has always [...] Read more.
Female fertility depends on the ovarian reserve of follicles, which is determined at birth. Primordial follicle development and oocyte maturation are regulated by multiple factors and pathways and classified into gonadotropin-independent and gonadotropin-dependent phases, according to the response to gonadotropins. Folliculogenesis has always been considered to be gonadotropin-dependent only from the antral stage, but evidence from the literature highlights the role of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) during early folliculogenesis with a potential role in the progression of the pool of primordial follicles. Hormonal and molecular pathway alterations during the very earliest stages of folliculogenesis may be the root cause of anovulation in polycystic ovary syndrome (PCOS) and in PCOS-like phenotypes related to antiepileptic treatment. Excessive induction of primordial follicle activation can also lead to premature ovarian insufficiency (POI), a condition characterized by menopause in women before 40 years of age. Future treatments aiming to suppress initial recruitment or prevent the growth of resting follicles could help in prolonging female fertility, especially in women with PCOS or POI. This review will briefly introduce the impact of gonadotropins on early folliculogenesis. We will discuss the influence of LH on ovarian reserve and its potential role in PCOS and POI infertility. Full article
(This article belongs to the Special Issue Novel Drugs for Infertility Treatment in 2022)
Show Figures

Figure 1

21 pages, 1556 KiB  
Review
An Update on Physiopathological Roles of Akt in the ReprodAKTive Mammalian Ovary
by Carlo Giaccari, Sevastiani Antonouli, George Anifandis, Sandra Cecconi and Valentina Di Nisio
Life 2024, 14(6), 722; https://doi.org/10.3390/life14060722 - 2 Jun 2024
Cited by 2 | Viewed by 1708
Abstract
The phosphoinositide 3-kinase (PI3K)/Akt pathway is a key signaling cascade responsible for the regulation of cell survival, proliferation, and metabolism in the ovarian microenvironment. The optimal finetuning of this pathway is essential for physiological processes concerning oogenesis, folliculogenesis, oocyte maturation, and embryo development. [...] Read more.
The phosphoinositide 3-kinase (PI3K)/Akt pathway is a key signaling cascade responsible for the regulation of cell survival, proliferation, and metabolism in the ovarian microenvironment. The optimal finetuning of this pathway is essential for physiological processes concerning oogenesis, folliculogenesis, oocyte maturation, and embryo development. The dysregulation of PI3K/Akt can impair molecular and structural mechanisms that will lead to follicle atresia, or the inability of embryos to reach later stages of development. Due to its pivotal role in the control of cell proliferation, apoptosis, and survival mechanisms, the dysregulation of this molecular pathway can trigger the onset of pathological conditions. Among these, we will focus on diseases that can harm female fertility, such as polycystic ovary syndrome and premature ovarian failure, or women’s general health, such as ovarian cancer. In this review, we report the functions of the PI3K/Akt pathway in both its physiological and pathological roles, and we address the existing application of inhibitors and activators for the balancing of the molecular cascade in ovarian pathological environments. Full article
(This article belongs to the Special Issue Biological and Clinical Research of Germ Cells)
Show Figures

Figure 1

Back to TopTop