Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.712
Filter
Add more filters

Publication year range
1.
Cell ; 187(4): 981-998.e25, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38325365

ABSTRACT

The female reproductive tract (FRT) undergoes extensive remodeling during reproductive cycling. This recurrent remodeling and how it shapes organ-specific aging remains poorly explored. Using single-cell and spatial transcriptomics, we systematically characterized morphological and gene expression changes occurring in ovary, oviduct, uterus, cervix, and vagina at each phase of the mouse estrous cycle, during decidualization, and into aging. These analyses reveal that fibroblasts play central-and highly organ-specific-roles in FRT remodeling by orchestrating extracellular matrix (ECM) reorganization and inflammation. Our results suggest a model wherein recurrent FRT remodeling over reproductive lifespan drives the gradual, age-related development of fibrosis and chronic inflammation. This hypothesis was directly tested using chemical ablation of cycling, which reduced fibrotic accumulation during aging. Our atlas provides extensive detail into how estrus, pregnancy, and aging shape the organs of the female reproductive tract and reveals the unexpected cost of the recurrent remodeling required for reproduction.


Subject(s)
Aging , Genitalia, Female , Animals , Female , Mice , Pregnancy , Genitalia, Female/cytology , Genitalia, Female/metabolism , Inflammation/metabolism , Uterus/cytology , Vagina/cytology , Single-Cell Analysis
2.
Cell ; 180(3): 585-600.e19, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32004457

ABSTRACT

Molecular mechanisms of ovarian aging and female age-related fertility decline remain unclear. We surveyed the single-cell transcriptomic landscape of ovaries from young and aged non-human primates (NHPs) and identified seven ovarian cell types with distinct gene-expression signatures, including oocyte and six types of ovarian somatic cells. In-depth dissection of gene-expression dynamics of oocytes revealed four subtypes at sequential and stepwise developmental stages. Further analysis of cell-type-specific aging-associated transcriptional changes uncovered the disturbance of antioxidant signaling specific to early-stage oocytes and granulosa cells, indicative of oxidative damage as a crucial factor in ovarian functional decline with age. Additionally, inactivated antioxidative pathways, increased reactive oxygen species, and apoptosis were observed in granulosa cells from aged women. This study provides a comprehensive understanding of the cell-type-specific mechanisms underlying primate ovarian aging at single-cell resolution, revealing new diagnostic biomarkers and potential therapeutic targets for age-related human ovarian disorders.


Subject(s)
Aging/genetics , Ovary/physiology , Single-Cell Analysis/methods , Transcriptome , Aged , Animals , Antioxidants/metabolism , Apoptosis/physiology , Atlases as Topic , Biomarkers , Cell Line, Tumor , Female , Granulosa Cells/metabolism , Humans , Macaca fascicularis , Oocytes/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/physiology
3.
Physiol Rev ; 103(4): 2623-2677, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37171807

ABSTRACT

Mammalian eggs (oocytes) are formed during fetal life and establish associations with somatic cells to form primordial follicles that create a store of germ cells (the primordial pool). The size of this pool is influenced by key events during the formation of germ cells and by factors that influence the subsequent activation of follicle growth. These regulatory pathways must ensure that the reserve of oocytes within primordial follicles in humans lasts for up to 50 years, yet only approximately 0.1% will ever be ovulated with the rest undergoing degeneration. This review outlines the mechanisms and regulatory pathways that govern the processes of oocyte and follicle formation and later growth, within the ovarian stroma, through to ovulation with particular reference to human oocytes/follicles. In addition, the effects of aging on female reproductive capacity through changes in oocyte number and quality are emphasized, with both the cellular mechanisms and clinical implications discussed. Finally, the details of current developments in culture systems that support all stages of follicle growth to generate mature oocytes in vitro and emerging prospects for making new oocytes from stem cells are outlined.


Subject(s)
Oocytes , Ovarian Follicle , Animals , Humans , Female , Oocytes/physiology , Ovarian Follicle/metabolism , Ovary/metabolism , Oogenesis/physiology , Mammals/physiology , Aging
4.
Genes Dev ; 34(3-4): 239-249, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31919193

ABSTRACT

Addressing the complexity of organogenesis at a system-wide level requires a complete understanding of adult cell types, their origin, and precursor relationships. The Drosophila ovary has been a model to study how coordinated stem cell units, germline, and somatic follicle stem cells maintain and renew an organ. However, lack of cell type-specific tools have limited our ability to study the origin of individual cell types and stem cell units. Here, we used a single-cell RNA sequencing approach to uncover all known cell types of the developing ovary, reveal transcriptional signatures, and identify cell type-specific markers for lineage tracing. Our study identifies a novel cell type corresponding to the elusive follicle stem cell precursors and predicts subtypes of known cell types. Altogether, we reveal a previously unanticipated complexity of the developing ovary and provide a comprehensive resource for the systematic analysis of ovary morphogenesis.


Subject(s)
Drosophila/cytology , Ovarian Follicle/cytology , Stem Cells/cytology , Animals , Drosophila/genetics , Drosophila/metabolism , Female , Models, Animal , Ovary/cytology , Sequence Analysis, RNA , Single-Cell Analysis , Transcription, Genetic
5.
Development ; 151(20)2024 Oct 15.
Article in English | MEDLINE | ID: mdl-38646855

ABSTRACT

Ecdysone-induced protein 93 (E93), known as the 'adult-specifier' transcription factor in insects, triggers metamorphosis in both hemimetabolous and holometabolous insects. Although E93 is conserved in ametabolous insects, its spatiotemporal expression and physiological function remain poorly understood. In this study, we first discover that, in the ametabolous firebrat Thermobia domestica, the previtellogenic ovary exhibits cyclically high E93 expression, and E93 mRNA is broadly distributed in previtellogenic ovarioles. E93 homozygous mutant females of T. domestica exhibit severe fecundity deficiency due to impaired previtellogenic development of the ovarian follicles, likely because E93 induces the expression of genes involved in ECM (extracellular matrix)-receptor interactions during previtellogenesis. Moreover, we reveal that in the hemimetabolous cockroach Blattella germanica, E93 similarly promotes previtellogenic ovarian development. In addition, E93 is also essential for vitellogenesis that is necessary to guarantee ovarian maturation and promotes the vitellogenesis-previtellogenesis switch in the fat body of adult female cockroaches. Our findings deepen the understanding of the roles of E93 in controlling reproduction in insects, and of E93 expression and functional evolution, which are proposed to have made crucial contributions to the origin of insect metamorphosis.


Subject(s)
Metamorphosis, Biological , Ovary , Reproduction , Animals , Female , Reproduction/genetics , Metamorphosis, Biological/genetics , Ovary/metabolism , Gene Expression Regulation, Developmental , Vitellogenesis/genetics , Insect Proteins/metabolism , Insect Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/genetics
6.
Genes Dev ; 33(13-14): 844-856, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31123065

ABSTRACT

The Piwi-interacting RNA (piRNA) pathway is a small RNA-based immune system that silences mobile genetic elements in animal germlines. piRNA biogenesis requires a specialized machinery that converts long single-stranded precursors into small RNAs of ∼25-nucleotides in length. This process involves factors that operate in two different subcellular compartments: the nuage/Yb body and mitochondria. How these two sites communicate to achieve accurate substrate selection and efficient processing remains unclear. Here, we investigate a previously uncharacterized piRNA biogenesis factor, Daedalus (Daed), that is located on the outer mitochondrial membrane. Daed is essential for Zucchini-mediated piRNA production and the correct localization of the indispensable piRNA biogenesis factor Armitage (Armi). We found that Gasz and Daed interact with each other and likely provide a mitochondrial "anchoring platform" to ensure that Armi is held in place, proximal to Zucchini, during piRNA processing. Our data suggest that Armi initially identifies piRNA precursors in nuage/Yb bodies in a manner that depends on Piwi and then moves to mitochondria to present precursors to the mitochondrial biogenesis machinery. These results represent a significant step in understanding a critical aspect of transposon silencing; namely, how RNAs are chosen to instruct the piRNA machinery in the nature of its silencing targets.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/metabolism , RNA Helicases/metabolism , RNA, Small Interfering/biosynthesis , Animals , Cell Line , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Knockdown Techniques , Protein Binding , Protein Transport , RNA, Small Interfering/metabolism
7.
Development ; 150(17)2023 09 01.
Article in English | MEDLINE | ID: mdl-37694610

ABSTRACT

Heparan sulfate (HS) and chondroitin sulfate (CS) are evolutionarily conserved glycosaminoglycans that are found in most animal species, including the genetically tractable model organism Drosophila. In contrast to extensive in vivo studies elucidating co-receptor functions of Drosophila HS proteoglycans (PGs), only a limited number of studies have been conducted for those of CSPGs. To investigate the global function of CS in development, we generated mutants for Chondroitin sulfate synthase (Chsy), which encodes the Drosophila homolog of mammalian chondroitin synthase 1, a crucial CS biosynthetic enzyme. Our characterizations of the Chsy mutants indicated that a fraction survive to adult stage, which allowed us to analyze the morphology of the adult organs. In the ovary, Chsy mutants exhibited altered stiffness of the basement membrane and muscle dysfunction, leading to a gradual degradation of the gross organ structure as mutant animals aged. Our observations show that normal CS function is required for the maintenance of the structural integrity of the ECM and gross organ architecture.


Subject(s)
Chondroitin Sulfates , Drosophila , Animals , Female , Drosophila/genetics , Ovarian Follicle , Ovary , Glycosaminoglycans , Mammals
8.
Development ; 150(7)2023 04 01.
Article in English | MEDLINE | ID: mdl-36971372

ABSTRACT

Computational analysis of bio-images by deep learning (DL) algorithms has made exceptional progress in recent years and has become much more accessible to non-specialists with the development of ready-to-use tools. The study of oogenesis mechanisms and female reproductive success has also recently benefited from the development of efficient protocols for three-dimensional (3D) imaging of ovaries. Such datasets have a great potential for generating new quantitative data but are, however, complex to analyze due to the lack of efficient workflows for 3D image analysis. Here, we have integrated two existing open-source DL tools, Noise2Void and Cellpose, into an analysis pipeline dedicated to 3D follicular content analysis, which is available on Fiji. Our pipeline was developed on larvae and adult medaka ovaries but was also successfully applied to different types of ovaries (trout, zebrafish and mouse). Image enhancement, Cellpose segmentation and post-processing of labels enabled automatic and accurate quantification of these 3D images, which exhibited irregular fluorescent staining, low autofluorescence signal or heterogeneous follicles sizes. In the future, this pipeline will be useful for extensive cellular phenotyping in fish or mammals for developmental or toxicology studies.


Subject(s)
Deep Learning , Female , Animals , Mice , Ovary/diagnostic imaging , Zebrafish , Imaging, Three-Dimensional/methods , Image Processing, Computer-Assisted/methods , Mammals
9.
Development ; 150(13)2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37272421

ABSTRACT

Oocytes develop in the germline cyst, a cellular organization in which germ cells are tightly interconnected and surrounded by somatic cells. The cyst produces oocytes for follicle formation and is a hub for essential processes in meiosis and oocyte differentiation. However, the formation and organization of the cyst, and their contribution to oocyte production in vertebrates remain unclear. Here, we provide tools for three-dimensional and functional in vivo analyses of the germline cyst in the zebrafish ovary. We describe the use of serial block-face scanning electron microscopy (SBF-SEM) to resolve the three-dimensional architecture of cells and organelles in the cyst at ultrastructural resolution. We present a deep learning-based pipeline for high-throughput quantitative analysis of three-dimensional confocal datasets of cysts in vivo. We provide a method for laser ablation of cellular components for manipulating cyst cells in ovaries. These methods will facilitate the investigation of the cyst cellular organization, expand the toolkit for the study of the zebrafish ovary, and advance our understanding of female developmental reproduction. They could also be further applied to the investigation of other developmental systems.


Subject(s)
Oogenesis , Zebrafish , Animals , Female , Oocytes , Ovary , Germ Cells/ultrastructure
10.
Bioessays ; 46(9): e2400056, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39072829

ABSTRACT

X chromosome centromeric drive may explain the prevalence of polycystic ovary syndrome and contribute to oocyte aneuploidy, menopause, and other conditions. The mammalian X chromosome may be vulnerable to meiotic drive because of X inactivation in the female germline. The human X pericentromeric region contains genes potentially involved in meiotic mechanisms, including multiple SPIN1 and ZXDC paralogs. This is consistent with a multigenic drive system comprising differential modification of the active and inactive X chromosome centromeres in female primordial germ cells and preferential segregation of the previously inactivated X chromosome centromere to the polar body at meiosis I. The drive mechanism may explain differences in X chromosome regulation in the female germlines of the human and mouse and, based on the functions encoded by the genes in the region, the transmission of X pericentromeric genetic or epigenetic variants to progeny could contribute to preeclampsia, autism, and differences in sexual differentiation.


Subject(s)
Centromere , Chromosomes, Human, X , Meiosis , Polycystic Ovary Syndrome , Humans , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/pathology , Female , Meiosis/genetics , Chromosomes, Human, X/genetics , Animals , Centromere/genetics , Mice , X Chromosome Inactivation/genetics , Prevalence
11.
J Biol Chem ; 300(3): 105663, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38246353

ABSTRACT

The maternal nutritional environment can impact progeny development, stress tolerance, and longevity. Such phenotypic variation of offspring resulting from the maternal environment is often referred to as the 'maternal effect' and is observed across taxa, including in humans. While some mechanisms behind maternal effects have been revealed, such as histone modification, many studies rely on drastic genetic or nutritional manipulation in describing these mechanisms. Here we aimed to reveal how the maternal environment is regulated under physiological conditions to affect the progeny. Specifically, we detailed metabolic regulation in oocytes in response to mating using Drosophila melanogaster fruit flies. Using liquid chromatography-mass spectrometry, we found that upon mating, the ovary metabolites shifted, predominantly toward increasing amino acids and the tryptophan/kynurenine (Kyn) pathway. This mating-induced increase in ovary Kyn was driven by increased Kyn production in the fat body, a functional counterpart of the mammalian liver and white adipose tissue and the source of Kyn storage for the ovary after mating. Furthermore, we show that maternal Kyn repression decreased the starvation resistance of progeny and that administering exogenous Kyn to the maternal generation enhanced the starvation resistance of female progeny. Taken together, these findings point to a previously unidentified role of fat body Kyn distribution during reproduction on progeny survival.


Subject(s)
Kynurenine , Starvation , Animals , Female , Drosophila melanogaster/metabolism , Kynurenine/metabolism , Ovary/metabolism , Reproduction , Male , Signal Transduction , Oocytes/metabolism , Biological Transport
12.
Trends Genet ; 38(5): 468-482, 2022 05.
Article in English | MEDLINE | ID: mdl-35094873

ABSTRACT

Identifying etiological risk factors is significant for preventing and treating patients with polycystic ovary syndrome (PCOS). Through genetic variation, Mendelian randomization (MR) assesses causal associations between PCOS risk and related exposure factors. This emerging technology has provided evidence of causal associations of anti-Müllerian hormone (AMH) levels, sex hormone-binding globulin (SHBG) levels, menopause age, adiposity, insulin resistance (IR), depression, breast cancer, ovarian cancer, obsessive-compulsive disorder (OCD), and forced vital capacity (FVC) with PCOS, while lacking associations of type 2 diabetes mellitus (T2DM), coronary heart disease (CHD), stroke, anxiety disorder (AD), schizophrenia (SCZ), bipolar disorder (BIP), and offspring birth weight with PCOS. In this review, we briefly introduce the concept and methodology of MR in terms of the opportunities and challenges in this field based on recent results obtained from MR analyses involving PCOS.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Polycystic Ovary Syndrome , Anti-Mullerian Hormone/genetics , Diabetes Mellitus, Type 2/genetics , Female , Humans , Insulin Resistance/genetics , Mendelian Randomization Analysis , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/genetics
13.
Development ; 149(18)2022 09 15.
Article in English | MEDLINE | ID: mdl-35993314

ABSTRACT

In the absence of pollination, female reproductive organs senesce, leading to an irrevocable loss in the reproductive potential of the flower, which directly affects seed set. In self-pollinating crops like wheat (Triticum aestivum), the post-anthesis viability of unpollinated carpels has been overlooked, despite its importance for hybrid seed production systems. To advance our knowledge of carpel development in the absence of pollination, we created a high-throughput phenotyping approach to quantify stigma and ovary morphology. We demonstrate the suitability of the approach, which uses light-microscopy imaging and machine learning, for the analysis of floral organ traits in field-grown plants using fresh and fixed samples. We show that the unpollinated carpel undergoes a well-defined initial growth phase, followed by a peak phase in which stigma area reaches its maximum and the radial expansion of the ovary slows, and a final deterioration phase. These developmental dynamics were consistent across years and could be used to classify male-sterile cultivars. This phenotyping approach provides a new tool for examining carpel development, which we hope will advance research into female fertility of wheat.


Subject(s)
Pollination , Triticum , Crops, Agricultural , Flowers/anatomy & histology , Flowers/genetics , Seeds/genetics , Triticum/genetics
14.
Annu Rev Genet ; 51: 265-285, 2017 11 27.
Article in English | MEDLINE | ID: mdl-28853925

ABSTRACT

Sexual reproduction crucially depends on the production of sperm in males and oocytes in females. Both types of gamete arise from the same precursor, the germ cells. We review the events that characterize the development of germ cells during fetal life as they commit to, and prepare for, oogenesis or spermatogenesis. In females, fetal germ cells enter meiosis, whereas in males they delay meiosis and instead lose pluripotency, activate an irreversible program of prospermatogonial differentiation, and temporarily cease dividing. Both pathways involve sex-specific molecular signals from the somatic cells of the developing gonads and a suite of intrinsic receptors, signal transducers, transcription factors, RNA stability factors, and epigenetic modulators that act in complex, interconnected positive and negative regulatory networks. Understanding these networks is important in the contexts of the etiology, diagnosis, and treatment of infertility and gonadal cancers, and in efforts to augment human and animal fertility using stem cell approaches.


Subject(s)
Infertility, Female/genetics , Infertility, Male/genetics , Oogenesis/genetics , Sex Determination Processes , Sex Differentiation/genetics , Spermatogenesis/genetics , Animals , Female , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Humans , Infertility, Female/metabolism , Infertility, Female/pathology , Infertility, Male/metabolism , Infertility, Male/pathology , Male , Meiosis , Oocytes/cytology , Oocytes/growth & development , Oocytes/metabolism , Ovum/cytology , Ovum/growth & development , Ovum/metabolism , Signal Transduction , Spermatozoa/cytology , Spermatozoa/growth & development , Spermatozoa/metabolism
15.
FASEB J ; 38(14): e23804, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39037422

ABSTRACT

Natural Nicotinamide Adenine Dinucleotide (NAD+) precursors have attracted much attention due to their positive effects in promoting ovarian health. However, their target tissue, synthesis efficiency, advantages, and disadvantages are still unclear. This review summarizes the distribution of NAD+ at the tissue, cellular and subcellular levels, discusses its biosynthetic pathways and the latest findings in ovary, include: (1) NAD+ plays distinct roles both intracellularly and extracellularly, adapting its distribution in response to requirements. (2) Different precursors differs in target tissues, synthetic efficiency, biological utilization, and adverse effects. Importantly: tryptophan is primarily utilized in the liver and kidneys, posing metabolic risks in excess; nicotinamide (NAM) is indispensable for maintaining NAD+ levels; nicotinic acid (NA) constructs a crucial bridge between intestinal microbiota and the host with diverse functions; nicotinamide riboside (NR) and nicotinamide mononucleotide (NMN) increase NAD+ systemically and can be influenced by delivery route, tissue specificity, and transport efficiency. (3) The biosynthetic pathways of NAD+ are intricately intertwined. They provide multiple sources and techniques for NAD+ synthesis, thereby reducing the dependence on a single molecule to maintain cellular NAD+ levels. However, an excess of a specific precursor potentially influencing other pathways. In addition, Protein expression analysis suggest that ovarian tissues may preferentially utilize NAM and NMN. These findings summarize the specific roles and potential of NAD+ precursors in enhancing ovarian health. Future research should delve into the molecular mechanisms and intervention strategies of different precursors, aiming to achieve personalized prevention or treatment of ovarian diseases, and reveal their clinical application value.


Subject(s)
NAD , Niacinamide , Ovary , Humans , NAD/metabolism , NAD/biosynthesis , Ovary/metabolism , Female , Animals , Niacinamide/metabolism , Niacinamide/biosynthesis , Biosynthetic Pathways , Nicotinamide Mononucleotide/metabolism
16.
FASEB J ; 38(7): e23602, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38581236

ABSTRACT

Neurotensin (NTS) is a 13-amino acid peptide which is highly expressed in the mammalian ovary in response to the luteinizing hormone surge. Antibody neutralization of NTS in the ovulatory follicle of the cynomolgus macaque impairs ovulation and induces follicular vascular dysregulation, with excessive pooling of red blood cells in the follicle antrum. We hypothesize that NTS is an essential intrafollicular regulator of vascular permeability. In the present study, follicle injection of the NTS receptor antagonist SR142948 also resulted in vascular dysregulation. To measure vascular permeability changes in vitro, primary macaque ovarian microvascular endothelial cells (mOMECs) were enriched from follicle aspirates and studied in vitro. When treated with NTS, permeability of mOMECs decreased. RNA sequencing (RNA-Seq) of mOMECs revealed high mRNA expression of the permeability-regulating adherens junction proteins N-cadherin (CDH2) and K-cadherin (CDH6). Immunofluorescent detection of CDH2 and CDH6 confirmed expression and localized these cadherins to the cell-cell boundaries, consistent with function as components of adherens junctions. mOMECs did not express detectable levels of the typical vascular endothelial cadherin, VE-cadherin (CDH5) as determined by RNA-Seq, qPCR, western blot, and immunofluorescence. Knockdown of CDH2 or CDH6 via siRNA abrogated the NTS effect on mOMEC permeability. Collectively, these data suggest that NTS plays an ovulation-critical role in vascular permeability maintenance, and that CDH2 and CDH6 are involved in the permeability modulating effect of NTS on the ovarian microvasculature. NTS can be added to a growing number of angiogenic regulators which are critical for successful ovulation.


Subject(s)
Endothelial Cells , Ovary , Female , Animals , Ovary/metabolism , Endothelial Cells/metabolism , Neurotensin/metabolism , Adherens Junctions/metabolism , Capillary Permeability , Cadherins/genetics , Cadherins/metabolism , Macaca/metabolism , Permeability , Endothelium, Vascular/metabolism , Mammals/metabolism
17.
FASEB J ; 38(1): e23377, 2024 01.
Article in English | MEDLINE | ID: mdl-38133902

ABSTRACT

The roles of anti-Müllerian hormone (AMH) continue to expand, from its discovery as a critical factor in sex determination, through its identification as a regulator of ovarian folliculogenesis, its use in fertility clinics as a measure of ovarian reserve, and its emerging role in hypothalamic-pituitary function. In light of these actions, AMH is considered an attractive therapeutic target to address diverse reproductive needs, including fertility preservation. Here, we set out to characterize the molecular mechanisms that govern AMH synthesis and activity. First, we enhanced the processing of the AMH precursor to >90% by introducing more efficient proprotein convertase cleavage sites (RKKR or ISSRKKRSVSS [SCUT]). Importantly, enhanced processing corresponded with a dramatic increase in secreted AMH activity. Next, based on species differences across the AMH type II receptor-binding interface, we generated a series of human AMH variants and assessed bioactivity. AMHSCUT potency (EC50 4 ng/mL) was increased 5- or 10-fold by incorporating Gln484 Met/Leu535 Thr (EC50 0.8 ng/mL) or Gln484 Met/Gly533 Ser (EC50 0.4 ng/mL) mutations, respectively. Furthermore, the Gln484 Met/Leu535 Thr double mutant displayed enhanced efficacy, relative to AMHSCUT . Finally, we identified residues within the wrist pre-helix of AMH (Trp494 , Gln496 , Ser497 , and Asp498 ) that likely mediate type I receptor binding. Mutagenesis of these residues generated gain- (Trp494 Phe or Gln496 Leu) or loss- (Ser497 Ala) of function AMH variants. Surprisingly, combining activating type I and type II receptor mutations only led to modest additive increases in AMH potency/efficacy. Our study is the first to characterize AMH residues involved in type I receptor binding and suggests a step-wise receptor-complex assembly mechanism, in which enhancement in the affinity of the ligand for either receptor can increase AMH activity beyond the natural level.


Subject(s)
Anti-Mullerian Hormone , Peptide Hormones , Female , Humans , Anti-Mullerian Hormone/genetics , Ovary , Amino Acid Sequence , Peptide Fragments
18.
Biol Cell ; : e00010, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38895958

ABSTRACT

BACKGROUND: The Endosomal Sorting Complex Required for Transport (ESCRT) is a highly conserved cellular machinery essential for many cellular functions, including transmembrane protein sorting, endosomal trafficking, and membrane scission. CHMP4B is a key component of ESCRT-III subcomplex and has been thoroughly studied in the meroistic ovaries of Drosophila melanogaster showing its relevance in maintaining this reproductive organ during the life of the fly. However, the role of the CHMP4B in the most basal panoistic ovaries remains elusive. RESULTS: Using RNAi, we examined the function of CHMP4B in the ovary of Blattella germanica in two different physiological stages: in last instar nymphs, with proliferative follicular cells, and in vitellogenic adults when follicular cells enter in polyploidy and endoreplication. In Chmp4b-depleted specimens, the actin fibers change their distribution, appearing accumulated in the basal pole of the follicular cells, resulting in an excess of actin bundles that surround the basal ovarian follicle and modifying their shape. Depletion of Chmp4b also determines an actin accumulation in follicular cell membranes, resulting in different cell morphologies and sizes. In the end, these changes disrupt the opening of intercellular spaces between the follicular cells (patency) impeding the incorporation of yolk proteins to the growing oocyte and resulting in female sterility. In addition, the nuclei of follicular cells appeared unusually elongated, suggesting an incomplete karyokinesis. CONCLUSIONS: These results proved CHMP4B essential in preserving the proper expression of cytoskeleton proteins vital for basal ovarian follicle growth and maturation and for yolk protein incorporation. Moreover, the correct distribution of actin fibers in the basal ovarian follicle emerged as a critical factor for the successful completion of ovulation and oviposition. SIGNIFICANCE: The overall results, obtained in two different proliferative stages, suggest that the requirement of CHMP4B in B. germanica follicular epithelium is not related to the proliferative stage of the tissue.

19.
Exp Cell Res ; 435(2): 113950, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38309674

ABSTRACT

The existing knowledge of the involvement of vinculin (VCL) in the control of ovarian cell functions is insufficient. To understand the role of VCL in the control of basic porcine ovarian granulosa cell functions, we decreased VCL activity by small interfering RNA (VCL siRNA). The expression of VCL, accumulation of VCL protein, cell viability, proliferation (accumulation of PCNA and cyclin B1), proportion of proliferative active cells, apoptosis (accumulation of bax, caspase 3, p53, antiapoptotic marker bcl2, and bax/bcl-2 ratio), DNA fragmentation, and release of steroid hormones and IGF-I were analyzed by RT‒qPCR, Trypan blue exclusion test, quantitative immunocytochemistry, XTT assay, TUNEL assay, and ELISA. The suppression of VCL activity inhibited cell viability, the accumulation of the proliferation-related proteins PCNA and cyclin B1, the antiapoptotic protein bcl2, and the proportion of proliferative active cells. Moreover, VCL siRNA inhibited the release of progesterone, estradiol, and IGF-1. VCL siRNA increased the proportion of the proapoptotic proteins bax, caspase 3, p53, the proportion of DNA fragmented cells, and stimulated testosterone release. Taken together, the present study is the first evidence that inhibition of VCL suppresses porcine granulosa cell functions. Moreover, the results suggest that VCL can be a potent physiological stimulator of ovarian functions.


Subject(s)
Progesterone , Tumor Suppressor Protein p53 , Female , Swine , Animals , Cyclin B1/metabolism , Cyclin B1/pharmacology , Caspase 3/genetics , Caspase 3/metabolism , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , Vinculin/genetics , Vinculin/metabolism , Progesterone/pharmacology , Apoptosis , Proto-Oncogene Proteins c-bcl-2/metabolism , Cell Proliferation , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Cells, Cultured , Insulin-Like Growth Factor I/metabolism
20.
Cell Mol Life Sci ; 81(1): 324, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080028

ABSTRACT

Polycystic ovary syndrome (PCOS) is a complex common endocrine disorder affecting women of reproductive age. Ovulatory dysfunction is recognized as a primary infertile factor, however, even when ovulation is medically induced and restored, PCOS patients continue to experience reduced cumulative pregnancy rates and a higher spontaneous miscarriage rate. Hyperandrogenism, a hallmark feature of PCOS, affects ovarian folliculogenesis, endometrial receptivity, and the establishment and maintenance of pregnancy. Decidualization denotes the transformation that the stromal compart of the endometrium must undergo to accommodate pregnancy, driven by the rising progesterone levels and local cAMP production. However, studies on the impact of hyperandrogenism on decidualization are limited. In this study, we observed that primary endometrial stromal cells from women with PCOS exhibit abnormal responses to progesterone during in vitro decidualization. A high concentration of testosterone inhibits human endometrial stromal cells (HESCs) decidualization. RNA-Seq analysis demonstrated that pyruvate dehydrogenase kinase 4 (PDK4) expression was significantly lower in the endometrium of PCOS patients with hyperandrogenism compared to those without hyperandrogenism. We also characterized that the expression of PDK4 is elevated in the endometrium stroma at the mid-secretory phase. Artificial decidualization could enhance PDK4 expression, while downregulation of PDK4 leads to abnormal decidualization both in vivo and in vitro. Mechanistically, testosterone excess inhibits IGFBP1 and PRL expression, followed by phosphorylating of AMPK that stimulates PDK4 expression. Based on co-immunoprecipitation analysis, we observed an interaction between SIRT1 and PDK4, promoting glycolysis to facilitate decidualization. Restrain of AR activation resumes the AMPK/SIRT1/PDK4 pathway suppressed by testosterone excess, indicating that testosterone primarily acts on decidualization through AR stimulation. Androgen excess in the endometrium inhibits decidualization by disrupting the AMPK/SIRT1/PDK4 signaling pathway. These data demonstrate the critical roles of endometrial PDK4 in regulating decidualization and provide valuable information for understanding the underlying mechanism during decidualization.


Subject(s)
AMP-Activated Protein Kinases , Endometrium , Polycystic Ovary Syndrome , Sirtuin 1 , Stromal Cells , Humans , Female , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Stromal Cells/metabolism , Stromal Cells/pathology , Stromal Cells/drug effects , Sirtuin 1/metabolism , Sirtuin 1/genetics , Endometrium/metabolism , Endometrium/pathology , Endometrium/drug effects , AMP-Activated Protein Kinases/metabolism , Adult , Hyperandrogenism/metabolism , Hyperandrogenism/pathology , Decidua/metabolism , Decidua/pathology , Testosterone/metabolism , Testosterone/pharmacology , Androgens/pharmacology , Androgens/metabolism , Progesterone/metabolism , Progesterone/pharmacology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL