Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Endocrinology ; 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38608138

RESUMEN

Clomiphene citrate is a common treatment for ovulation induction in subfertile women, but its use is associated with elevated risk of adverse perinatal outcomes and birth defects. To investigate the biological plausibility of a causal relationship, this study investigated in mice the consequences for fetal development and pregnancy outcome of peri-conception clomiphene citrate administration at doses approximating human exposures. A dose-dependent adverse effect of clomiphene citrate given twice in the 36 h after mating was seen, with a moderate dose of 0.75 mg/kg sufficient to cause altered reproductive outcomes in three independent cohorts. Viable pregnancy was reduced by 30%, late gestation fetal weight was reduced by 16%, and ∼30% of fetuses exhibited delayed development and/or congenital abnormalities not seen in control dams, including defects of the lung, kidney, liver, eye, skin, limbs, and umbilicus. Clomiphene citrate also caused a 30 h average delay in time of birth, and elevated rate of pup death in the early postnatal phase. In surviving offspring, growth trajectory tracking and body morphometry analysis at 20 weeks of age showed post-weaning growth and development comparable to controls. A dysregulated inflammatory response in the endometrium was observed and may contribute to the underlying pathophysiological mechanism. These results demonstrate that in utero exposure to clomiphene citrate during early pregnancy can inhibit implantation and impact fetal growth and development, causing adverse perinatal outcomes. The findings raise the prospect of similar iatrogenic effects in women where clomiphene citrate may be present in the peri-conception phase unless its use is well-supervised.

2.
Genes (Basel) ; 15(3)2024 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-38540426

RESUMEN

Mitochondria undergo a myriad of changes during pre-implantation embryo development, including shifts in activity levels and mitochondrial DNA (mtDNA) replication. However, how these distinct aspects of mitochondrial function are linked and their responsiveness to diverse stressors is not well understood. Here, we show that mtDNA content increased between 8-cell embryos and the blastocyst stage, with similar copy numbers per cell in the inner cell mass (ICM) and trophectoderm (TE). In contrast, mitochondrial membrane potential (MMP) was higher in TE than ICM. Culture in ambient oxygen (20% O2) altered both aspects of mitochondrial function: the mtDNA copy number was upregulated in ICM, while MMP was diminished in TE. Embryos cultured in 20% O2 also exhibited delayed development kinetics, impaired implantation, and reduced mtDNA levels in E18 fetal liver. A model of oocyte mitochondrial stress using rotenone showed only a modest effect on on-time development and did not alter the mtDNA copy number in ICM; however, following embryo transfer, mtDNA was higher in the fetal heart. Lastly, endogenous mitochondrial dysfunction, induced by maternal age and obesity, altered the blastocyst mtDNA copy number, but not within the ICM. These results demonstrate that mitochondrial activity and mtDNA content exhibit cell-specific changes and are differentially responsive to diverse types of oxidative stress during pre-implantation embryogenesis.


Asunto(s)
Variaciones en el Número de Copia de ADN , ADN Mitocondrial , Animales , Ratones , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Variaciones en el Número de Copia de ADN/genética , Potenciales de la Membrana , Mitocondrias/metabolismo , Estrés Oxidativo/genética , Desarrollo Embrionario/genética , Oxígeno/metabolismo
3.
Front Cell Dev Biol ; 11: 1261038, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37941899

RESUMEN

The ovarian follicle is a complex structure that protects and helps in the maturation of the oocyte, and then releases it through the controlled molecular and structural remodeling process of ovulation. The progesterone receptor (PGR) has been shown to be essential in regulating ovulation-related gene expression changes. In this study, we found disrupted expression of the cellular adhesion receptor gene Sema7A in the granulosa cells of PGR-/- mice during ovulation. We subsequently found that expression of Sema7A in preovulatory follicles is promoted by gonadotropins and hypoxia, establishing an asymmetrical pattern with the SEMA7A protein enriched at the apex of large antral follicles. Sema7A expression was downregulated through a PGR-dependent mechanism in the periovulatory period, the abundance of SEMA7A protein was reduced, and the asymmetric pattern became more homogeneous after an ovulatory stimulus. Receptors for Sema7A can either repel or promote intercellular adhesion. During ovulation, striking inverse regulation of repulsive Plxnc1 and adhesive Itga5/Itgb1 receptors likely contributes to dramatic tissue remodeling. The adhesive receptor Itga5 was significantly increased in periovulatory granulosa cells and cumulus-oocyte complexes (COCs), and functional assays showed that periovulatory granulosa cells and COCs acquire increased adhesive phenotypes, while Sema7A repels granulosa cell contact. These findings suggest that the regulation of Sema7A and its associated receptors, along with the modulation of integrin α5, may be critical in establishing the multilaminar ovarian follicle structure and facilitating the remodeling and apical release of the cumulus-oocyte complex during ovulation.

4.
Biomolecules ; 13(10)2023 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-37892227

RESUMEN

The Hypoxia Inducible Factor (HIF) transcription factors are imperative for cell adaption to low oxygen conditions and development; however, they also contribute to ischaemic disease and cancer. To identify novel genetic regulators which target the HIF pathway or small molecules for therapeutic use, cell-based reporter systems are commonly used. Here, we present a new, highly sensitive and versatile reporter system, NanoFIRE: a NanoLuciferase and Fluorescent Integrated Reporter Element. Under the control of a Hypoxic Response Element (HRE-NanoFIRE), this system is a robust sensor of HIF activity within cells and potently responds to both hypoxia and chemical inducers of the HIF pathway in a highly reproducible and sensitive manner, consistently achieving 20 to 150-fold induction across different cell types and a Z' score > 0.5. We demonstrate that the NanoFIRE system is adaptable via substitution of the response element controlling NanoLuciferase and show that it can report on the activity of the transcriptional regulator Factor Inhibiting HIF, and an unrelated transcription factor, the Progesterone Receptor. Furthermore, the lentivirus-mediated stable integration of NanoFIRE highlights the versatility of this system across a wide range of cell types, including primary cells. Together, these findings demonstrate that NanoFIRE is a robust reporter system for the investigation of HIF and other transcription factor-mediated signalling pathways in cells, with applications in high throughput screening for the identification of novel small molecule and genetic regulators.


Asunto(s)
Regulación de la Expresión Génica , Factores de Transcripción , Humanos , Factores de Transcripción/genética , Elementos de Respuesta , Proteínas Nucleares/genética , Hipoxia/genética , Hipoxia de la Célula/genética
5.
Nucleic Acids Res ; 51(12): 5981-5996, 2023 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-37099375

RESUMEN

Progesterone receptor (PGR) plays diverse roles in reproductive tissues and thus coordinates mammalian fertility. In the ovary, rapid acute induction of PGR is the key determinant of ovulation through transcriptional control of a unique set of genes that culminates in follicle rupture. However, the molecular mechanisms for this specialized PGR function in ovulation is poorly understood. We have assembled a detailed genomic profile of PGR action through combined ATAC-seq, RNA-seq and ChIP-seq analysis in wildtype and isoform-specific PGR null mice. We demonstrate that stimulating ovulation rapidly reprograms chromatin accessibility in two-thirds of sites, correlating with altered gene expression. An ovary-specific PGR action involving interaction with RUNX transcription factors was observed with 70% of PGR-bound regions also bound by RUNX1. These transcriptional complexes direct PGR binding to proximal promoter regions. Additionally, direct PGR binding to the canonical NR3C motif enable chromatin accessibility. Together these PGR actions mediate induction of essential ovulatory genes. Our findings highlight a novel PGR transcriptional mechanism specific to ovulation, providing new targets for infertility treatments or new contraceptives that block ovulation.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal , Regulación de la Expresión Génica , Receptores de Progesterona , Transcripción Genética , Animales , Femenino , Ratones , Cromatina/genética , Ensamble y Desensamble de Cromatina/genética , Mamíferos/genética , Ratones Noqueados , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo
6.
Sci Adv ; 8(24): eabn4564, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35714185

RESUMEN

The female ovary contains a finite number of oocytes, and their release at ovulation becomes sporadic and disordered with aging and with obesity, leading to loss of fertility. Understanding the molecular defects underpinning this pathology is essential as age of childbearing and obesity rates increase globally. We identify that fibrosis within the ovarian stromal compartment is an underlying mechanism responsible for impaired oocyte release, which is initiated by mitochondrial dysfunction leading to diminished bioenergetics, oxidative damage, inflammation, and collagen deposition. Furthermore, antifibrosis drugs (pirfenidone and BGP-15) eliminate fibrotic collagen and restore ovulation in reproductively old and obese mice, in association with dampened M2 macrophage polarization and up-regulated MMP13 protease. This is the first evidence that ovarian fibrosis is reversible and indicates that drugs targeting mitochondrial metabolism may be a viable therapeutic strategy for women with metabolic disorders or advancing age to maintain ovarian function and extend fertility.


Asunto(s)
Longevidad , Ovario , Animales , Colágeno/metabolismo , Femenino , Fibrosis , Humanos , Ratones , Obesidad/metabolismo , Oocitos/metabolismo , Ovario/metabolismo , Ovario/patología
7.
Cells ; 11(9)2022 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-35563869

RESUMEN

Progesterone receptor (PGR) activity is obligatory for mammalian ovulation; however, there is no established direct functional pathway explaining how progesterone receptor completely and specifically regulates oocyte release. This study examined the overarching cell- and isoform-specific effects of the PGR within each cellular compartment of the ovary, using mice null for the PGR (PRKO), as well as isoform-specific null mice. The PGR was expressed in ovarian granulosa and stromal cells and although PRKO ovaries showed no visible histological changes in preovulatory ovarian morphology, follicle rupture did not occur. Reciprocal ovarian transplant experiments established the necessity of ovarian PGR expression for ovulation. Cumulus-oocyte complexes of PRKO mice exhibited normal morphology but showed some altered gene expression. The examination of mitochondrial activity showed subtle differences in PRKO oocytes but no differences in granulosa cell respiration, glycolysis or ß-oxidation. Concurrently, RNA-seq identified novel functional pathways through which the PGR may regulate ovulation. PGR-A was the predominant transcriptionally active isoform in granulosa cells and 154 key PGR-dependent genes were identified, including a secondary network of transcription factors. In addition, the PGR regulated unique gene networks in the ovarian stroma. Collectively, we establish the effector pathways activated by the PGR across the ovarian cell types and conclude that PGR coordinates gene expression in the cumulus, granulosa and stromal cells at ovulation. Identifying these networks linking the PGR to ovulation provides novel targets for fertility therapeutics and nonhormonal contraceptive development.


Asunto(s)
Ovulación , Receptores de Progesterona , Animales , Femenino , Células de la Granulosa/metabolismo , Mamíferos/metabolismo , Ratones , Ratones Noqueados , Progesterona/farmacología , Isoformas de Proteínas/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
8.
Front Endocrinol (Lausanne) ; 12: 607539, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33912131

RESUMEN

Endocrine disrupting compounds (EDCs) are prevalent and ubiquitous in our environment and have substantial potential to compromise human and animal health. Amongst the chronic health conditions associated with EDC exposure, dysregulation of reproductive function in both females and males is prominent. Human epidemiological studies demonstrate links between EDC exposure and infertility, as well as gestational disorders including miscarriage, fetal growth restriction, preeclampsia, and preterm birth. Animal experiments show EDCs administered during gestation, or to either parent prior to conception, can interfere with gamete quality, embryo implantation, and placental and fetal development, with consequences for offspring viability and health. It has been presumed that EDCs operate principally through disrupting hormone-regulated events in reproduction and fetal development, but EDC effects on maternal immune receptivity to pregnancy are also implicated. EDCs can modulate both the innate and adaptive arms of the immune system, to alter inflammatory responses, and interfere with generation of regulatory T (Treg) cells that are critical for pregnancy tolerance. Effects of EDCs on immune cells are complex and likely exerted by both steroid hormone-dependent and hormone-independent pathways. Thus, to better understand how EDCs impact reproduction and pregnancy, it is imperative to consider how immune-mediated mechanisms are affected by EDCs. This review will describe evidence that several EDCs modify elements of the immune response relevant to pregnancy, and will discuss the potential for EDCs to disrupt immune tolerance required for robust placentation and optimal fetal development.


Asunto(s)
Disruptores Endocrinos/toxicidad , Tolerancia Inmunológica/efectos de los fármacos , Inflamación/inducido químicamente , Complicaciones del Embarazo/inducido químicamente , Animales , Femenino , Desarrollo Fetal/efectos de los fármacos , Humanos , Recién Nacido , Inflamación/complicaciones , Inflamación/inmunología , Masculino , Embarazo , Complicaciones del Embarazo/inmunología
9.
FASEB Bioadv ; 2(6): 339-353, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32617520

RESUMEN

N-cadherin is a homophilic cell-cell adhesion molecule that plays a critical role in maintaining vascular stability and modulating endothelial barrier permeability. Pre-clinical studies have shown that the N-cadherin antagonist peptide, ADH-1, increases the permeability of tumor-associated vasculature thereby increasing anti-cancer drug delivery to tumors and enhancing tumor response. Small molecule library screens have identified a novel compound, LCRF-0006, that is a mimetic of the classical cadherin His-Ala-Val sequence-containing region of ADH-1. Here, we evaluated the vascular permeability-enhancing and anti-cancer properties of LCRF-0006 using in vitro vascular disruption and cell apoptosis assays, and a well-established pre-clinical model (C57BL/KaLwRij/5TGM1) of the hematological cancer multiple myeloma (MM). We found that LCRF-0006 disrupted endothelial cell junctions in a rapid, transient and reversible manner, and increased vascular permeability in vitro and at sites of MM tumor in vivo. Notably, LCRF-0006 synergistically increased the in vivo anti-MM tumor response to low-dose bortezomib, a frontline anti-MM agent, leading to regression of disease in 100% of mice. Moreover, LCRF-0006 and bortezomib synergistically induced 5TGM1 MM tumor cell apoptosis in vitro. Our findings demonstrate the potential clinical utility of LCRF-0006 to significantly increase bortezomib effectiveness and enhance the depth of tumor response in patients with MM.

10.
Cell Physiol Biochem ; 52(6): 1553-1568, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31135123

RESUMEN

BACKGROUND/AIMS: Despite, several studies demonstrating pro-metastatic effects of the metalloproteinase ADAMTS1 in breast cancer, its role in facilitating the metastatic cascade remains unclear. To date there have been limited studies that have examined the expression of ADAMTS1 in primary and metastatic breast cancer tissues. METHODS: We assessed ADAMTS1 mRNA levels in publically available breast cancer sets and analysed ADAMTS1 protein levels by immunohistochemistry in breast tissue microarrays containing normal breast tissue (n=9), primary invasive ductal breast carcinomas (n=79) and metastatic lesions (n=58). To understand the underlying events influenced by ADAMTS1 and provide a mechanism by which tumors expressing this protease promote metastasis, we assessed the ability of PyMT/Adamts1+/+, PyMT/Adamts1+/- and PyMT/Adamts1-/- primary mammary cancer cells to adhere to matrigel and migrate or invade towards a chemoattractive environment. RESULTS: High ADAMTS1 expression was associated with reduced disease-free survival, distant metastasis free-survival and overall survival in breast cancer patients with node negative disease. Although ADAMTS1 expression was reduced in primary breast cancers compared to normal tissue and not elevated in metastatic lesions, high ADAMTS1 immunostaining was associated with a higher number of positive lymph nodes (p=0.006) and the presence of distant metastasis (p=0.023). Depletion of Adamts1 in mammary cancer cells impeded their adhesion to a biological matrix substratum and diminished cell migration but not invasion. CONCLUSION: The effects observed on cell adhesion and migration demonstrates a potential mechanism whereby ADAMTS1 promotes breast cancer metastasis.


Asunto(s)
Proteína ADAMTS1/metabolismo , Neoplasias de la Mama/patología , Proteínas de la Matriz Extracelular/metabolismo , Proteína ADAMTS1/genética , Animales , Neoplasias de la Mama/mortalidad , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Supervivencia sin Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Metástasis Linfática , Ratones , Ratones Transgénicos , Estadificación de Neoplasias , Pronóstico
11.
Endocrinology ; 159(9): 3209-3218, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30010832

RESUMEN

Ovulation is the appropriately timed release of a mature, developmentally competent oocyte from the ovary into the oviduct, where fertilization occurs. Importantly, ovulation is tightly linked with oocyte maturation, demonstrating the interdependency of these two parallel processes, both essential for female fertility. Initiated by pituitary gonadotropins, the ovulatory process is mediated by intrafollicular paracrine factors from the theca, mural, and cumulus granulosa cells, as well as the oocyte itself. The result is the induction of cumulus expansion, proteolysis, angiogenesis, inflammation, and smooth muscle contraction, which are each required for follicular rupture. These complex intercellular communication networks and the essential ovulatory genes have been well defined in mouse models and are highly conserved in primates, including humans. Importantly, recent discoveries in regulation of ovulation highlight new areas of investigation.


Asunto(s)
Hormona Luteinizante/metabolismo , Oocitos/crecimiento & desarrollo , Ovulación/metabolismo , Animales , Células del Cúmulo/fisiología , Femenino , Hormona Folículo Estimulante/metabolismo , Hormona Folículo Estimulante/fisiología , Humanos , Hormona Luteinizante/fisiología , Ratones , Contracción Muscular/fisiología , Músculo Liso , Neovascularización Fisiológica/fisiología , Oocitos/metabolismo , Folículo Ovárico/fisiología , Ovulación/fisiología , Primates , Proteolisis , Células Tecales/fisiología , Factores de Tiempo
12.
Am J Reprod Immunol ; 79(6): e12835, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29484756

RESUMEN

PROBLEM: The nuclear progesterone receptor (PGR) transcription factor is essential for ovulation; however, the exact mechanisms by which PGR controls ovulation are not known. The aim of this study was to determine whether PGR regulates inflammatory mediators in the ovary. METHOD OF STUDY: Ovaries from mice lacking PGR (PRKO) and heterozygous PR+/- littermates were subjected to microarray analysis of a large panel of inflammatory genes. Immune cell subsets were detected by gene expression; and neutrophils by immunohistochemistry and chemotaxis assay. RESULTS: PRKO ovaries exhibited dysregulated expression of vasodilator (Edn1), cytokine (Il-6, Tgfb1), adhesion receptor (Cd34), apoptotic factor (Bax) and transcription factors (Nfkb2, Socs1, Stat3). Ptgs2 was also reduced in PRKO ovaries, but mRNA and protein were not different in granulosa cells. There were reduced neutrophils in ovaries of PRKO mice at ovulation; however, chemotaxis assays showed PRKO neutrophils migrate normally and that PRKO ovarian extracts exhibit chemotactic properties in vitro. CONCLUSION: Specific inflammatory mediators are altered in the ovaries of PRKO mice indicating that progesterone regulates features of inflammation at ovulation.


Asunto(s)
Núcleo Celular/metabolismo , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Ovario/metabolismo , Ovulación/metabolismo , Receptores de Progesterona/metabolismo , Animales , Quimiotaxis/fisiología , Femenino , Expresión Génica/fisiología , Células de la Granulosa/metabolismo , Ratones , Neutrófilos/metabolismo , ARN Mensajero/metabolismo
13.
Front Physiol ; 8: 422, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28674503

RESUMEN

The hormone relaxin is important in female reproduction for embryo implantation, cardiovascular function, and during labor and lactation. Relaxin is also synthesized in males by organs of the male tract. We hypothesized that relaxin might be one component of seminal plasma responsible for eliciting the female cytokine response induced in the uterus at mating. When recombinant relaxin was injected into the uterus of wild-type (Rln+/+) mice at estrus, it evoked the production of Cxcl1 mRNA and its secreted protein product CXCL1 in four of eight animals. Mating experiments were then conducted using mice with a null mutation in the relaxin gene (Rln-/- mice). qRT-PCR analysis of mRNA expression in wild-type females showed diminished uterine expression of several cytokine and chemokine genes in the absence of male relaxin. Similar differences were also noted comparing Rln-/- and Rln+/+ females mated to wild-type males. Quantification of uterine luminal fluid cytokine content confirmed that male relaxin provokes the production of CXCL10 and CSF3 in Rln+/+ females. Differences were also seen comparing Rln-/- and Rln+/+ females mated with Rln-/- males for CXCL1, CSF3, and CCL5, implying that endogenous relaxin in females might prime the uterus to respond appropriately to seminal fluid at coitus. Finally, pan-leukocyte CD45 mRNA was increased in wild-type matings compared to other combinations, implying that male and female relaxin may trigger leukocyte expansion in the uterus. We conclude that male and/or female relaxin may be important in activating the uterine cytokine/chemokine network required to initiate maternal immune adaptation to pregnancy.

14.
Reproduction ; 153(3): R109-R120, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27879344

RESUMEN

In vitro maturation (IVM) offers significant benefits for human infertility treatment and animal breeding, but this potential is yet to be fully realised due to reduced oocyte developmental competence in comparison with in vivo matured oocytes. Cumulus cells occupy an essential position in determining oocyte developmental competence. Here we have examined the areas of deficient gene expression, as determined within microarrays primarily from cumulus cells of mouse COCs, but also other species, between in vivo matured and in vitro matured oocytes. By retrospectively analysing the literature, directed by focussing on downregulated genes, we provide an insight as to why the in vitro cumulus cells fail to support full oocyte potential and dissect molecular pathways that have important roles in oocyte competence. We conclude that the roles of epidermal growth factor signalling, the expanded extracellular matrix, cumulus cell metabolism and the immune system are critical deficiencies in cumulus cells of IVM COCs.


Asunto(s)
Células del Cúmulo/citología , Regulación de la Expresión Génica , Técnicas de Maduración In Vitro de los Oocitos , Oocitos/citología , Oogénesis/fisiología , Animales , Células del Cúmulo/metabolismo , Femenino , Humanos , Oocitos/metabolismo
15.
Endocrinology ; 157(11): 4209-4211, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27799008
16.
Theriogenology ; 86(1): 62-8, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-27160446

RESUMEN

Cumulus cell-oocyte communication is an essential feature of mammalian reproduction. Established mechanisms involve the bidirectional transfer of ions and small molecules through gap junctions that fundamentally regulate the process of oocyte maturation. Also, well established is the paracrine signaling from the oocyte to the cumulus, which regulates much of the flow of ions and molecules to the oocyte and orchestrates many of the associated local signaling events around ovulation, which is the key to establishing oocyte competence to sustain early embryo development. Less well-characterized and new potential players include exosomal transfer of noncoding RNAs from cumulus to oocytes and the recent observations of the presence of hemoglobin in oocytes and cumulus cells. The impact of these new communication pathways is either poorly defined or even unknown. Finally, signaling between the two cell types most likely continues after ovulation and even fertilization; however, this too is largely undefined but may play roles in substrate transport, sperm chemotaxis and "trapping", and potential signaling to the rest of the reproductive tract.


Asunto(s)
Células del Cúmulo/fisiología , Oocitos/fisiología , Animales , Comunicación Celular , Femenino , Regulación de la Expresión Génica
17.
Nutrients ; 8(1)2016 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-26771633

RESUMEN

The Robinson Research Institute of the University of Adelaide convened a multidisciplinary group of n = 33 clinicians, researchers and representatives of government organisations on the 2 October 2014 for a workshop entitled "Promoting fertility and healthy conception. How do we generate greater reproductive health awareness?" The key aim of the workshop was to assess the body of knowledge that informs clinical practice and government policy, and to identify questions and additional information needed by health practitioners and government representatives working in the field of reproductive health and to frame future research and policy. The workshop identified topics that fell mostly into three categories: lifestyle-related, societal and biological factors. The lifestyle topics included nutrition and diet, exercise, obesity, shift work and other factors deemed to be modifiable at the level of the individual. The societal topics included discussions of matters that are structural, and resistant to change by individuals, including specific ethical issues, social disadvantage, government and educational policies. The biological factors are intrinsic physical states of the individual, and included many factors where there is a dense body of scientific knowledge which may not be readily accessible in less academic language. This workshop thus provided an opportunity to identify further actions that could be undertaken to meet the needs of diverse organisations and groups of professionals with an interest in human fertility. Since so many factors in our social and biological environment can impact fertility and preconception health, it is imperative to involve many disciplines or levels of government or societal organisations that have not traditionally been involved in this area.


Asunto(s)
Investigación Biomédica , Personal de Salud , Medicina Reproductiva , Investigadores , Investigación , Australia , Conferencias de Consenso como Asunto , Fertilidad , Fertilización , Humanos , Comunicación Interdisciplinaria
19.
Biol Reprod ; 92(6): 153, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25972011

RESUMEN

Oocytes within antral follicles are thought to have restricted access to O2, as follicle vascularity is not adjacent and both granulosa and cumulus cells are metabolically active. Indeed, measured follicular antrum partial pressure (pO2) is regarded as low, but accurate and direct measurement represents a technical challenge that has yet to be overcome. The oocyte itself is highly dependent on oxidative phosphorylation for survival and competence for further development following fertilization, and it has been suggested that follicular pO2 levels are correlated with this capacity for further development. It is clear that gonadotropins are involved in regulating antrum formation, follicle vascularization, cellular differentiation, and the hypoxia-inducible factors (HIF), which are mainly regulated by dissolved O2 concentration. A newly discovered player in this story is the intracellular production of hemoglobin by both granulosa and cumulus cells, as well as the oocyte. Furthermore, cellular hemoglobin levels are dynamic, responding to the ovulatory luteinizing hormone (LH) surge. We hypothesize that this gas transport and antioxidant molecule is involved in the prevention of hypoxic response signaling by HIFs within the preovulatory antral follicle; and the transition of granulosa cells to luteal tissue by facilitating the stabilization of HIFs, enabling induction of luteinization signaling. Another possible role is by sequestering nitric oxide (NO) during the ovulatory period, which may facilitate the resumption of meiosis in the oocyte. Testing these hypotheses will be challenging but important if the regulation of ovarian function is to be fully understood.


Asunto(s)
Hipoxia/metabolismo , Oocitos/metabolismo , Folículo Ovárico/metabolismo , Animales , Femenino , Células de la Granulosa/citología , Células de la Granulosa/metabolismo , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Oocitos/citología , Folículo Ovárico/citología
20.
Matrix Biol ; 44-46: 54-63, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25818315

RESUMEN

The reproductive organs are unique among adult organs in that they must undergo continual tissue remodelling as a key aspect of their normal function. The processes for persistent maturation and release of new gametes, as well as fertilisation, implantation, placentation, gestation and parturition involve cyclic development and regression of tissues that must continually regenerate to support fertility. The ADAMTS family of proteases has been shown to contribute to many aspects of the tissue morphogenesis required for development and function of each of the reproductive organs. Dysregulation or functional changes in ADAMTS family proteases have been associated with reproductive disorders such as polycystic ovarian syndrome (PCOS) and premature ovarian failure (POF). Likewise, proteolytic substrates of ADAMTS enzymes have also been linked to reproductive function. New insight into the roles of ADAMTS proteases has yielded a deeper understanding of the molecular mechanisms behind fertility with clinical potential to generate therapeutic targets to resolve infertility, develop biomarkers that predict dysfunction of the reproductive organs and potentially offer targets for development of non-hormonal male and female contraceptives.


Asunto(s)
Proteínas ADAM/metabolismo , Fertilidad , Gametogénesis , Proteínas ADAM/genética , Anticoncepción , Femenino , Humanos , Masculino , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/metabolismo , Insuficiencia Ovárica Primaria/genética , Insuficiencia Ovárica Primaria/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...