Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Clin Endocrinol (Oxf) ; 91(2): 237-244, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31004515

RESUMEN

BACKGROUND: Discordance between gonadal type and gender identity has often led to an assumption of infertility in patients with differences in sex development (DSD). However, there is now greater recognition of fertility being an important issue for this group of patients. Currently, gonadal tissue that may have fertility potential is not being stored for individuals with DSD and, where gonadectomy forms part of management, is often discarded. The area of fertility preservation has been predominantly driven by oncofertility which is a field dedicated to preserving the fertility of patients undergoing gonadotoxic cancer treatment. The use of fertility preservation techniques could be expanded to include individuals with DSD where functioning gonads are present. METHODS: This is a systematic literature review evaluating original research articles and relevant reviews between 1974 and 2018 addressing DSD and fertility, in vitro maturation of sperm, and histological/ultrastructural assessment of gonadal tissue in complete and partial androgen insensitivity syndrome, 17ß-hydroxysteroid dehydrogenase type 3 and 5α-reductase deficiency. CONCLUSION: Successful clinical outcomes of ovarian tissue cryopreservation are paving the way for similar research being conducted using testicular tissue and sperm. There have been promising results from both animal and human studies leading to cryopreservation of testicular tissue now being offered to boys prior to cancer treatment. Although data are limited, there is evidence to suggest the presence of reproductive potential in the gonads of some individuals with DSD. Larger, more detailed studies are required, but if these continue to be encouraging, individuals with DSD should be given the same information, opportunities and access to fertility preservation as other patient groups.


Asunto(s)
3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/deficiencia , Criopreservación/métodos , Trastorno del Desarrollo Sexual 46,XY/fisiopatología , Trastornos del Desarrollo Sexual/fisiopatología , Preservación de la Fertilidad/métodos , Hipospadias/fisiopatología , Errores Congénitos del Metabolismo Esteroideo/fisiopatología , Trastorno del Desarrollo Sexual 46,XY/diagnóstico , Trastornos del Desarrollo Sexual/diagnóstico , Femenino , Humanos , Hipospadias/diagnóstico , Masculino , Ovario/fisiología , Reproducción/fisiología , Espermatozoides/fisiología , Errores Congénitos del Metabolismo Esteroideo/diagnóstico
2.
Reproduction ; 157(2): 135-148, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30601757

RESUMEN

Follicle development requires complex and coordinated interactions between both the oocyte and its associated somatic cells. In ovarian dysfunction, follicle development may be abnormal due to defective somatic cell function; for example, premature ovarian insufficiency or malignancies. Replacing defective somatic cells, using the reaggregated ovary (RO) technique, may 'rescue' follicle development. ROs containing mature follicles have been generated when transplanted to a host mouse to develop. We have developed a RO culture technique and the aims were to determine how follicle development differed between transplanted and cultured ROs, and the influence of ovarian age (P2 vs P6). Mouse ROs were cultured for 14 days; P2 and P6 ovaries cultured as Controls. Follicle development was compared to ROs transplanted for 14 days and ovaries from P16 and P20 mice. ROs generated from either P2 or P6 exhibited similar follicle development in culture whereas in vivo follicle development was more advanced in P6 ROs. Follicles were more developed in cultured ROs than transplanted ROs. However, follicles in cultured ROs and ovaries had smaller oocytes with fewer theca and granulosa cells than in vivo counterparts. Our results demonstrate the fluidity of follicle development despite ovary dissociation and that environment is more important to basal lamina formation and theca cell development. Furthermore, follicle development within cultured ROs appears to be independent of oocyte nest breakdown and primordial follicle formation in source ovaries. Our results highlight the need for understanding follicle development in vitro, particularly in the development of the RO technique as a potential fertility treatment.


Asunto(s)
Folículo Ovárico/fisiología , Ovario/citología , Animales , Femenino , Células de la Granulosa/citología , Ratones , Oocitos/citología , Técnicas de Cultivo de Órganos
3.
Reprod Fertil Dev ; 31(3): 529-537, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30373703

RESUMEN

Cumulus-oocyte complex (COC) expansion is essential for ovulation and fertilisation and is linked to oocyte quality. Hyaluronan (HA), the major matrix constituent, is cross-linked via inter-α-inhibitor heavy chains (HCs), pentraxin 3 (PTX3) and tumour necrosis factor-stimulated gene 6 (TSG-6). All except HCs are secreted by cumulus cells in response to oocyte-secreted factors, which signal via SMAD pathways. The double mutant (DM) mouse generates oocytes lacking complex N- and O-glycans due to oocyte-specific deletion of core 1 ß1,3-galactosyltransferase (C1galt1) and N-acetylglucosaminyltransferase I (Mgat1) and has modified cumulus expansion. We compared COCs before expansion (48 h-post-pregnant mare serum gonadotrophin (PMSG)) and at late-stage expansion (9 h-post-human chorionic gonadotrophin (hCG); control n=3 mice, DM n=3 per group). Using histochemistry the levels of HA, HCs, PTX3, TSG-6 and phosphorylated-SMAD1/5/8 and -SMAD2 (12-25 COCs per group) were assessed. DM COCs did not differ from Controls in cumulus size or cell density at 9 h-post-hCG; however, HA and HC levels and phosphorylated-SMAD1/5/8 were reduced. Furthermore, no correlations were found between the levels of matrix molecules and cumulus area in DM or Control samples. These data suggest that HA and HCs can support cumulus expansion provided that they are present above minimum threshold levels. We propose that oocyte-specific ablation of C1galt1 and Mgat1 may affect bone morphogenetic protein 15 synthesis or bioactivity, thereby reducing SMAD1/5/8 phosphorylation and HA production.


Asunto(s)
Células del Cúmulo/metabolismo , Matriz Extracelular/metabolismo , Oocitos/metabolismo , Polisacáridos/metabolismo , Transducción de Señal/fisiología , Animales , Femenino , Ratones , Folículo Ovárico/metabolismo , Ovulación/metabolismo , Fosforilación , Polisacáridos/genética
5.
Reproduction ; 154(1): R23-R35, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28389520

RESUMEN

For many decades, the dogma prevailed that female mammals had a finite pool of oocytes at birth and this was gradually exhausted during a lifetime of reproductive function. However, in 2004, a new era began in the field of female oogenesis. A study was published that appeared to detect oocyte-stem cells capable of generating new eggs within mouse ovaries. This study was highly controversial and the years since this initial finding have produced extensive research and even more extensive debate into their possibility. Unequivocal evidence testifying to the existence of oocyte-stem cells (OSCs) has yet to be produced, meanwhile the spectrum of views from both sides of the debate are wide-ranging and surprisingly passionate. Although recent studies have presented some convincing results that germ cells exist and are capable of creating new oocytes, many questions remain. Are these cells present in humans? Do they exist in physiological conditions in a dormant state? This comprehensive review first examines where and how the dogma of a finite pool was established, how this has been challenged over the years and addresses the most pertinent questions as to the current status of their existence, their role in female fertility, and perhaps most importantly, if they do exist, how can we harness these cells to improve a woman's oocyte reserve and treat conditions such as premature ovarian insufficiency (POI: also known as premature ovarian failure, POF).


Asunto(s)
Oocitos , Células Madre Oogoniales , Células Madre , Adulto , Animales , Femenino , Fertilidad , Humanos , Menopausia , Ratones , Persona de Mediana Edad , Oocitos/fisiología , Oogénesis/fisiología , Células Madre Oogoniales/fisiología , Reserva Ovárica/fisiología , Insuficiencia Ovárica Primaria/terapia , Reproducción/fisiología , Especificidad de la Especie , Células Madre/fisiología
6.
Reprod Fertil Dev ; 29(8): 1538-1544, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27652508

RESUMEN

Premature ovarian insufficiency (POI) occurs in 1% of women under 40 years of age and is predominantly idiopathic. In a transgenic mouse model of follicular POI, the Double Mutant (DM), female mice are fertile at 6 weeks of age, become infertile by 9 weeks and exhibit POI by 3 months. DM female mice generate oocytes lacking mucin O-glycans and complex N-glycans due to deletion of core 1 synthase, glycoprotein-N-acetylgalactosamine 3-ß-galactosyltransferase 1 (C1galt1) and mannoside acetylglucosaminyltransferase 1 (Mgat1) respectively (DM, C1galt1F/FMgat1F/F:ZP3Cre; Control, C1galt1F/FMgat1F/F). To determine whether DM follicle development could be improved in a controlled environment, follicles from DM and Control mice were cultured individually and follicle growth, morphology, survival and antrum formation were evaluated. DM ovaries were more rigid than Control ovaries at 3, 6 and 9 weeks, which was exacerbated with age, resulting in a failure to isolate follicles from 9 week-old DM females. DM follicles had decreased survival compared with Control follicles from females at 3 and 6 weeks of age. Furthermore, survival rate of DM follicles decreased with age between 3 and 6 weeks. DM follicles at both 3 and 6 weeks had accelerated follicle growth and altered antrum formation during the first few days of culture but, after 6 days, follicles were equivalent in size to the Controls. In conclusion, a population of DM follicles retain the potential to develop in vitro, and therefore follicle culture offers a reliable method to generate antral follicles from preantral follicles after the onset of POI in these female mice.


Asunto(s)
Folículo Ovárico/crecimiento & desarrollo , Ovario/crecimiento & desarrollo , Insuficiencia Ovárica Primaria/patología , Animales , Modelos Animales de Enfermedad , Femenino , Fertilidad , Ratones , Folículo Ovárico/patología , Ovario/patología
7.
Dev Biol ; 401(2): 287-98, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25557622

RESUMEN

Maintaining follicle integrity during development, whereby each follicle is a functional unit containing a single oocyte, is essential for the generation of healthy oocytes. However, the mechanisms that regulate this critical function have not been determined. In this paper we investigate the role of the oocyte in maintaining follicle development. To investigate this role, we use a mouse model with oocyte-specific deletion of C1galt1 which is required for the generation of core 1-derived O-glycans. The loss of oocyte-generated O-glycans results in the joining of follicles and the generation of Multiple-Oocyte Follicles (MOFs). The aim was to determine how Mutant follicle development is modified thus enabling follicles to join. Extracellular matrix and follicle permeability were studied using histology, immunohistochemistry and electron microscopy (EM). In ovaries containing Mutant Oocytes, the Follicle basal lamina (FBL) is altered both functionally and structurally from the primary stage onwards with Mutant follicles possessing unexpectedly thicker FBL. In Mutant ovaries, the theca cell layer is also modified with intermingling of theca between adjacent follicles. MOF function was analysed but despite increased numbers of preantral MOFs in Mutants, these do not reach the preovulatory stage after gonadotrophin stimulation. We propose a model describing how oocyte initiated changes in FBL and theca cells result in follicles joining. These data reveal new and important roles for the oocyte in follicle development and follicle integrity.


Asunto(s)
Membrana Basal/embriología , Galactosiltransferasas/genética , Oocitos/metabolismo , Folículo Ovárico/embriología , Células Tecales/citología , Animales , Membrana Basal/citología , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Femenino , Glicoproteínas/metabolismo , Gonadotropinas/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Folículo Ovárico/citología , Permeabilidad , Polisacáridos/genética
8.
Biochem Soc Trans ; 44(4): 1091-100, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27528757

RESUMEN

One in 400 people has a maternally inherited mutation in mtDNA potentially causing incurable disease. In so-called heteroplasmic disease, mutant and normal mtDNA co-exist in the cells of carrier women. Disease severity depends on the proportion of inherited abnormal mtDNA molecules. Families who have had a child die of severe, maternally inherited mtDNA disease need reliable information on the risk of recurrence in future pregnancies. However, prenatal diagnosis and even estimates of risk are fraught with uncertainty because of the complex and stochastic dynamics of heteroplasmy. These complications include an mtDNA bottleneck, whereby hard-to-predict fluctuations in the proportions of mutant and normal mtDNA may arise between generations. In 'mitochondrial replacement therapy' (MRT), damaged mitochondria are replaced with healthy ones in early human development, using nuclear transfer. We are developing non-invasive alternatives, notably activating autophagy, a cellular quality control mechanism, in which damaged cellular components are engulfed by autophagosomes. This approach could be used in combination with MRT or with the regular management, pre-implantation genetic diagnosis (PGD). Mathematical theory, supported by recent experiments, suggests that this strategy may be fruitful in controlling heteroplasmy. Using mice that are transgenic for fluorescent LC3 (the hallmark of autophagy) we quantified autophagosomes in cleavage stage embryos. We confirmed that the autophagosome count peaks in four-cell embryos and this correlates with a drop in the mtDNA content of the whole embryo. This suggests removal by mitophagy (mitochondria-specific autophagy). We suggest that modulating heteroplasmy by activating mitophagy may be a useful complement to mitochondrial replacement therapy.


Asunto(s)
ADN Mitocondrial/genética , Mitocondrias/genética , Enfermedades Mitocondriales/genética , Mutación , Animales , Autofagia/genética , Niño , ADN Mitocondrial/metabolismo , Desarrollo Embrionario/genética , Femenino , Heterocigoto , Humanos , Masculino , Herencia Materna/genética , Ratones Endogámicos C57BL , Enfermedades Mitocondriales/prevención & control , Enfermedades Mitocondriales/terapia , Terapia de Reemplazo Mitocondrial/métodos , Mitofagia/genética , Modelos Genéticos
9.
FASEB J ; 29(2): 525-39, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25416550

RESUMEN

The number of eggs ovulated varies within and between species and is influenced by many variables. However, the regulatory mechanisms remain poorly understood. We previously demonstrated a key role for the oocyte because mice generating oocytes deficient in core 1-derived O-glycans ovulate ∼40-50% more eggs than Controls. Here we analyze the basis of this phenotype using Mutant [core 1 ß1,3-galactosyltransferase 1 (C1galt1)(FF):zona pellucida glycoprotein 3 Cre (ZP3Cre)] and Control (C1galt1(FF)) female mice. In culture, Mutant follicles exhibited delayed antrum formation [indicative of follicle stimulant hormone (FSH) dependence] and increased sensitivity to FSH. Although the Mutant estrous cycle was extended, comprehensive endocrine changes were not observed; rather FSH, LH, inhibin B, and anti-Mullerian hormone were temporally altered, revealing estrous cycle stage-specific modifications to the hypothalamic-pituitary-gonadal axis. At proestrus, when FSH levels were decreased in Mutants, ovaries contained more, smaller, preantral follicles. Mutant follicles exhibited reduced levels of apoptosis, and both B-cell lymphoma 2 (Bcl-2) and BCL-2-associated X protein (Bax) were altered compared with Controls. Mutant ovaries also had an increase in the expression ratio of growth differentiation factor 9 (GDF9):bone morphogenetic protein 15 (BMP15) at diestrus. On the basis of these data, we propose that modified oocyte glycoproteins alter GDF9:BMP15 expression modifying follicle development resulting in the generation of more follicles. Thus, the oocyte is a key regulator of follicle development and has a crucial role in determining ovulation rate.


Asunto(s)
Apoptosis , Proteína Morfogenética Ósea 15/metabolismo , Hormona Folículo Estimulante/metabolismo , Factor 9 de Diferenciación de Crecimiento/metabolismo , Oocitos/citología , Polisacáridos/química , Alelos , Animales , Ciclo Estral , Femenino , Fertilidad , Regulación de la Expresión Génica , Genotipo , Ratones , Mutación , Folículo Ovárico/metabolismo , Ovario/metabolismo , Fenotipo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo
10.
Reproduction ; 149(5): 533-43, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25855670

RESUMEN

During follicle development, oocytes secrete factors that influence the development of granulosa and cumulus cells (CCs). In response to oocyte and somatic cell signals, CCs produce extracellular matrix (ECM) molecules resulting in cumulus expansion, which is essential for ovulation, fertilisation, and is predictive of oocyte quality. The cumulus ECM is largely made up of hyaluronan (HA), TNF-stimulated gene-6 (TSG-6, also known as TNFAIP6), pentraxin-3 (PTX3), and the heavy chains (HCs) of serum-derived inter-α-inhibitor proteins. In contrast to other in vivo models where modified expansion impairs fertility, the cumulus mass of C1galt1 Mutants, which have oocyte-specific deletion of core 1-derived O-glycans, is modified without impairing fertility. In this report, we used C1galt1 Mutant (C1galt1(FF):ZP3Cre) and Control (C1galt1(FF)) mice to investigate how cumulus expansion is affected by oocyte-specific deletion of core 1-derived O-glycans without adversely affecting oocyte quality. Mutant cumulus-oocyte complexes (COCs) are smaller than Controls, with fewer CCs. Interestingly, the CCs in Mutant mice are functionally normal as each cell produced normal levels of the ECM molecules HA, TSG-6, and PTX3. However, HC levels were elevated in Mutant COCs. These data reveal that oocyte glycoproteins carrying core 1-derived O-glycans have a regulatory role in COC development. In addition, our study of Controls indicates that a functional COC can form provided all essential components are present above a minimum threshold level, and thus some variation in ECM composition does not adversely affect oocyte development, ovulation or fertilisation. These data have important implications for IVF and the use of cumulus expansion as a criterion for oocyte assessment.


Asunto(s)
Células del Cúmulo/metabolismo , Matriz Extracelular/metabolismo , Galactosiltransferasas/fisiología , Oocitos/metabolismo , Folículo Ovárico/metabolismo , Polisacáridos/deficiencia , Animales , Células Cultivadas , Células del Cúmulo/citología , Femenino , Fertilización , Técnicas para Inmunoenzimas , Ratones , Ratones Noqueados , Oocitos/citología , Folículo Ovárico/citología , Ovulación/fisiología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
11.
Front Cell Dev Biol ; 12: 1449156, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39258229

RESUMEN

Introduction: Survival rates of the childhood cancer patients are improving, however cancer treatments such as chemotherapy may lead to infertility due to loss of the primordial follicle (PMF) reserve. Doxorubicin (DXR) is a gonadotoxic chemotherapy agent commonly used in childhood cancers. Anti-Müllerian Hormone (AMH) has been reported to have a protective effect on the mouse ovarian reserve against DXR in vivo. However, whether AMH can prevent PMF loss in conjunction with DXR in human ovarian tissue in vivo has not been determined. Methods: In order to investigate this, we first established an optimum dose of DXR that induced PMF loss in cultured mouse ovaries and investigated the efficacy of AMH on reducing DXR-induced PMF loss in mice in vitro. Second, we investigated the effects of DXR on pre-pubertal human ovarian tissue and the ability of AMH to prevent DXR-induced damage comparing using a mouse xenograft model with different transplantation sites. Results: Mouse ovaries treated with DXR in vitro and in vivo had reduced PMF populations and damaged follicle health. We did not observe effect of DXR-induced PMF loss or damage to follicle/stromal health in human ovarian cortex, this might have been due to an insufficient dose or duration of DXR. Although AMH does not prevent DXR-induced PMF loss in pre-pubertal and adult mouse ovaries, in mouse ovaries treated with higher concentration of AMH in vitro, DXR did not cause a significant loss in PMFs. This is the first study to illustrate an effect of AMH on DXR-induced PMF loss on pre-pubertal mouse ovaries. However, more experiments with higher doses of AMH and larger sample size are needed to confirm this finding. Discussion: We did not observe that AMH could prevent DXR-induced PMF loss in mouse ovaries in vivo. Further studies are warranted to investigate whether AMH has a protective effect against DXR in xenotransplanted human ovarian tissue. Thus, to obtain robust evidence about the potential of AMH in fertility preservation during chemotherapy treatment, alternative AMH administration strategies need to be explored alongside DXR administration to fully interrogate the effect of DXR and AMH on human xenografted tissues.

12.
Annu Rev Anim Biosci ; 12: 91-112, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-37988633

RESUMEN

Cloning as it relates to the animal kingdom generally refers to the production of genetically identical individuals. Because cloning is increasingly the subject of renewed attention as a tool for rescuing endangered or extinct species, it seems timely to dissect the role of the numerous reproductive techniques encompassed by this term in animal species conservation. Although cloning is typically associated with somatic cell nuclear transfer, the recent advent of additional techniques that allow genome replication without genetic recombination demands that the use of induced pluripotent stem cells to generate gametes or embryos, as well as older methods such as embryo splitting, all be included in this discussion. Additionally, the phenomenon of natural cloning (e.g., a subset of fish, birds, invertebrates, and reptilian species that reproduce via parthenogenesis) must also be pointed out. Beyond the biology of these techniques are practical considerations and the ethics of using cloning and associated procedures in endangered or extinct species. All of these must be examined in concert to determine whether cloning has a place in species conservation. Therefore, we synthesize progress in cloning and associated techniques and dissect the practical and ethical aspects of these methods as they pertain to endangered species conservation.


Asunto(s)
Clonación de Organismos , Especies en Peligro de Extinción , Animales , Clonación de Organismos/veterinaria , Clonación de Organismos/métodos , Técnicas de Transferencia Nuclear/veterinaria , Peces/genética , Clonación Molecular
13.
Biol Rev Camb Philos Soc ; 98(4): 1225-1249, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37016502

RESUMEN

The term 'cloning' refers to the production of genetically identical individuals but has meant different things throughout the history of science: a natural means of reproduction in bacteria, a routine procedure in horticulture, and an ever-evolving gamut of molecular technologies in vertebrates. Mammalian cloning can be achieved through embryo splitting, somatic cell nuclear transfer, and most recently, by the use of induced pluripotent stem cells. Several emerging biotechnologies also facilitate the propagation of genomes from one generation to the next whilst bypassing the conventional reproductive processes. In this review, we examine the state of the art of available cloning technologies and their progress in species other than humans and rodent models, in order to provide a critical overview of their readiness and relevance for application in endangered animal conservation.


Asunto(s)
Especies en Peligro de Extinción , Técnicas de Transferencia Nuclear , Animales , Humanos , Clonación de Organismos/métodos , Vertebrados , Mamíferos , Embrión de Mamíferos
14.
Front Cell Dev Biol ; 11: 1202411, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37614224

RESUMEN

The mechanisms and aetiology underlying the development of premature ovarian insufficiency (POI) are poorly understood. However, the oocyte clearly has a role as demonstrated by the Double Mutant (DM) mouse model where ovarian dysfunction (6 weeks) is followed by POI (3 months) due to oocyte-specific deletion of complex and hybrid N- and O-glycans. The ovaries of DM mice contain more primary follicles (3a stage) accompanied by fewer developing follicles, indicating a block in follicle development. To investigate this block, we first analysed early follicle development in postnatal (8-day), pre-pubertal (3-week) and post-pubertal (6-week and 3-month) DM (C1galt1 F/F Mgat1 F/F:ZP3Cre) and Control (C1galt1 F/F Mgat1 F/F) mice. Second, we investigated if transplantation of DM ovaries into a "normal" endocrine environment would restore follicle development. Third, we determined if replacing DM ovarian somatic cells would rescue development of DM oocytes. At 3-week, DM primary 3a follicles contain large oocytes accompanied by early development of a second GC layer and increased GC proliferation. At 6-week, DM primary 3a follicles contain abnormally large oocytes, accompanied with decreased GC proliferation. Transplantation of DM ovaries into a 'normal' endocrine environment did not restore normal follicle development. However, replacing somatic cells by generating reaggregated ovaries (ROs) did enable follicle development to progress and thus highlighted intra-ovarian factors were responsible for the onset of POI in DM females. Thus, these studies demonstrate oocyte-initiated altered communication between GCs and oocytes results in abnormal primary follicles which fail to progress and leads to POI.

15.
Commun Biol ; 6(1): 1049, 2023 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-37848538

RESUMEN

The northern white rhinoceros is functionally extinct with only two females left. Establishing methods to culture ovarian tissues, follicles, and oocytes to generate eggs will support conservation efforts using in vitro embryo production. To the best of our knowledge, this is the first description of the structure and molecular signature of any rhinoceros, more specifically, we describe the neonatal and adult southern white rhinoceros (Ceratotherium simum simum) ovary; the closest relation of the northern white rhinoceros. Interestingly, all ovaries contain follicles despite advanced age. Analysis of the neonate reveals a population of cells molecularly characterised as mitotically active, pluripotent with germ cell properties. These results indicate that unusually, the neonatal ovary still contains oogonia in germ cell nests at birth, providing an opportunity for fertility preservation. Therefore, utilising ovaries from stillborn and adult rhinoceros can provide cells for advanced assisted reproductive technologies and investigating the neonatal ovaries of other endangered species is crucial for conservation.


Asunto(s)
Oogonios , Ovario , Animales , Femenino , Oocitos , Especies en Peligro de Extinción , Perisodáctilos
16.
Biol Reprod ; 86(6): 179, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22492969

RESUMEN

To identify roles in spermatogenesis for major subclasses of N- and O-glycans and Notch signaling, male mice carrying floxed C1galt1, Pofut1, Notch1 or Mgat1 alleles and a testis-specific Cre recombinase transgene were generated. T-synthase (C1GALT1) transfers Gal to generate core 1 and core 2 mucin O-glycans; POFUT1 transfers O-fucose to particular epidermal growth factor-like repeats and is essential for canonical Notch signaling; and MGAT1 (GlcNAcT-I) transfers GlcNAc to initiate hybrid and complex N-glycan synthesis. Cre recombinase transgenes driven by various promoters were investigated, including Stra8-iCre expressed in spermatogonia, Sycp1-Cre expressed in spermatocytes, Prm1-Cre expressed in spermatids, and AMH-Cre expressed in Sertoli cells. All Cre transgenes deleted floxed alleles, but efficiencies varied widely. Stra8-iCre was the most effective, deleting floxed Notch1 and Mgat1 alleles with 100% efficiency and floxed C1galt1 and Pofut1 alleles with ~80% efficiency, based on transmission of deleted alleles. Removal of C1galt1, Pofut1, or Notch1 in spermatogonia had no effect on testicular weight, histology, or fertility. However, males in which the synthesis of complex N-glycans was blocked by deletion of Mgat1 in spermatogonia did not produce sperm. Spermatogonia, spermatocytes, and spermatids were generated, but most spermatids formed giant multinucleated cells or symplasts, and apoptosis was increased. Therefore, although core 1 and 2 mucin O-glycans, NOTCH1, POFUT1, O-fucose glycans, and Notch signaling are dispensable, MGAT1 and complex N-glycans are essential for spermatogenesis.


Asunto(s)
Aciltransferasas/metabolismo , Polisacáridos/biosíntesis , Receptor Notch1/metabolismo , Espermatogénesis , Espermatozoides/metabolismo , Aciltransferasas/genética , Animales , Masculino , Ratones , Ratones Transgénicos , N-Acetilglucosaminiltransferasas
17.
Biol Reprod ; 87(5): 114, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23018181

RESUMEN

Assisted reproductive technology (ART) has resulted in more than 5 million births worldwide. However, mainstream ART techniques are not always successful for an estimated 30% of infertile patients in whom gametes are nonviable. Most patients would clearly prefer genetic parenthood, currently possible only via the use of donated gametes or, in future, via the clinical use of artificial gametes (AGs) incorporating parental DNA. Despite much recent progress in the derivation of AGs, significant obstacles remain. Although it is possible to create artificial cells exhibiting some of the molecular and physiological traits of human gametes, they do not yet exhibit the same level of functionality as their in vivo counterparts. Most current effort pays scant attention to confirmation of molecular integrity and clinical applicability of AGs. Here we discuss the various clinical parameters used to assess gamete and embryo viability and discuss markers of gamete function that may be used within future studies attempting to derive AGs. The use of AGs may prove controversial to some members of the general public, and, as such, there is significant need for an appropriate ethical and legal framework governing the clinical use of such cells. However, provided these issues can be successfully overcome, it is highly likely that AGs will represent powerful biological tools for reproductive science, a valuable training resource for embryologists and for potential use in the clinical treatment of human infertility.


Asunto(s)
Células Germinativas/fisiología , Técnicas Reproductivas Asistidas , Animales , Biomarcadores , Supervivencia Celular , ADN , Epigénesis Genética/genética , Femenino , Humanos , Infertilidad/terapia , Masculino , Donación de Oocito , Oocitos/fisiología , Embarazo , Técnicas Reproductivas Asistidas/ética , Técnicas Reproductivas Asistidas/tendencias , Espermatozoides/fisiología , Donantes de Tejidos
18.
Reproduction ; 144(4): 455-65, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22919046

RESUMEN

Female mice generating oocytes lacking complex N- and O-glycans (double mutants (DM)) produce only one small litter before undergoing premature ovarian failure (POF) by 3 months. Here we investigate the basis of the small litter by evaluating ovulation rate and embryo development in DM (Mgat1(F/F)C1galt1(F/F):ZP3Cre) and Control (Mgat1(F/F)C1galt1(F/F)) females. Surprisingly, DM ovulation rate was normal at 6 weeks, but declined dramatically by 9 weeks. In vitro development of zygotes to blastocysts was equivalent to Controls although all embryos from DM females lacked a normal zona pellucida (ZP) and ∼30% lacked a ZP entirely. In contrast, in vivo preimplantation development resulted in less embryos recovered from DM females compared with Controls at 3.5 days post coitum (dpc) (3.2±1.3 vs 7.0±0.6). Furthermore, only 45% of mated DM females contained embryos at 3.5 dpc. Of the preimplantation embryos collected from DM females, approximately half were morulae unlike Controls where the majority were blastocysts, indicating delayed embryo development in DM females. Post-implantation development in DM females was analysed to determine whether delayed preimplantation development affected subsequent development. In DM females at 5.5 dpc, only ∼40% of embryos found at 3.5 dpc had implanted. However, at 6.5 dpc, implantation sites in DM females corresponded to embryo numbers at 3.5 dpc indicating delayed implantation. At 9.5 dpc, the number of decidua corresponded to embryo numbers 6 days earlier indicating that all implanted embryos progress to midgestation. Therefore, a lack of complex N- and O-glycans in oocytes during development impairs early embryo development and viability in vivo leading to delayed implantation and a small litter.


Asunto(s)
Aciltransferasas/metabolismo , Implantación Tardía del Embrión , Pérdida del Embrión/metabolismo , Embrión de Mamíferos/metabolismo , Galactosiltransferasas/metabolismo , Polisacáridos/metabolismo , Aciltransferasas/genética , Animales , Blastocisto/enzimología , Blastocisto/metabolismo , Blastocisto/patología , Decidua/enzimología , Decidua/metabolismo , Ectogénesis , Proteínas del Huevo/genética , Proteínas del Huevo/metabolismo , Pérdida del Embrión/enzimología , Pérdida del Embrión/patología , Embrión de Mamíferos/enzimología , Embrión de Mamíferos/patología , Femenino , Galactosiltransferasas/genética , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Mutantes , Ratones Transgénicos , Mórula/enzimología , Mórula/metabolismo , Mórula/patología , N-Acetilglucosaminiltransferasas , Ovulación , Embarazo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Zona Pelúcida/enzimología , Zona Pelúcida/metabolismo , Glicoproteínas de la Zona Pelúcida , Cigoto/enzimología , Cigoto/metabolismo , Cigoto/patología
19.
Reprod Fertil ; 3(1): C1-C3, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-35350654

RESUMEN

Fertility preservation is a rapidly advancing field with numerous broad applications ranging from retaining the prospect of fertility in a child with cancer to protecting an entire species from extinction. In recent years, huge strides have been made in understanding the biology of male and female reproduction in animals and humans and using this knowledge to develop strategies for fertility preservation across a range of clinical and ecological applications. This Reproduction and Fertility preservation series is composed of articles from experts on this topic and these will highlight key developments in fertility preservation and also identify the challenges that still face this exciting and relatively new field.


Asunto(s)
Preservación de la Fertilidad , Neoplasias , Animales , Niño , Femenino , Fertilidad , Humanos , Masculino , Reproducción
20.
Reprod Fertil ; 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-35972313

RESUMEN

The manuscript has been submitted without altering abstract in line with Reproduction's Flexible Submission Process. The abstract is extended and thus does not fit this space.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA