Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Hum Reprod Update ; 30(1): 3-25, 2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-37639630

RESUMO

BACKGROUND: While oocyte IVM is practiced sporadically it has not achieved widespread clinical practice globally. However, recently there have been some seminal advances in our understanding of basic aspects of oocyte biology and ovulation from animal studies that have led to novel approaches to IVM. A significant recent advance in IVM technology is the use of biphasic IVM approaches. These involve the collection of immature oocytes from small antral follicles from minimally stimulated patients/animals (without hCG-priming) and an ∼24 h pre-culture of oocytes in an advanced culture system ('pre-IVM') prior to IVM, followed by routine IVF procedures. If safe and efficacious, this novel procedure may stand to make a significant impact on human ART practices. OBJECTIVE AND RATIONALE: The objectives of this review are to examine the major scientific advances in ovarian biology with a unique focus on the development of pre-IVM methodologies, to provide an insight into biphasic IVM procedures, and to report on outcomes from animal and clinical human data, including safety data. The potential future impact of biphasic IVM on ART practice is discussed. SEARCH METHODS: Peer review original and review articles were selected from PubMed and Web of Science searches for this narrative review. Searches were performed using the following keywords: oocyte IVM, pre-IVM, biphasic IVM, CAPA-IVM, hCG-triggered/primed IVM, natural cycle IVF/M, ex-vivo IVM, OTO-IVM, oocyte maturation, meiotic competence, oocyte developmental competence, oocyte capacitation, follicle size, cumulus cell (CC), granulosa cell, COC, gap-junction communication, trans-zonal process, cAMP and IVM, cGMP and IVM, CNP and IVM, EGF-like peptide and IVM, minimal stimulation ART, PCOS. OUTCOMES: Minimizing gonadotrophin use means IVM oocytes will be collected from small antral (pre-dominant) follicles containing oocytes that are still developing. Standard IVM yields suboptimal clinical outcomes using such oocytes, whereas pre-IVM aims to continue the oocyte's development ex vivo, prior to IVM. Pre-IVM achieves this by eliciting profound cellular changes in the oocyte's CCs, which continue to meet the oocyte's developmental needs during the pre-IVM phase. The literature contains 25 years of animal research on various pre-IVM and biphasic IVM procedures, which serves as a large knowledge base for new approaches to human IVM. A pre-IVM procedure based on c-type natriuretic peptide (named 'capacitation-IVM' (CAPA-IVM)) has undergone pre-clinical human safety and efficacy trials and its adoption into clinical practice resulted in healthy live birth rates not different from conventional IVF. WIDER IMPLICATIONS: Over many decades, improvements in clinical IVM have been gradual and incremental but there has likely been a turning of the tide in the past few years, with landmark discoveries in animal oocyte biology finally making their way into clinical practice leading to improved outcomes for patients. Demonstration of favorable clinical results with CAPA-IVM, as the first clinically tested biphasic IVM system, has led to renewed interest in IVM as an alternative, low-intervention, low-cost, safe, patient-friendly ART approach, and especially for patients with PCOS. The same new approach is being used as part of fertility preservation in patients with cancer and holds promise for social oocyte freezing.


Assuntos
Técnicas de Maturação in Vitro de Oócitos , Síndrome do Ovário Policístico , Animais , Feminino , Humanos , Técnicas de Maturação in Vitro de Oócitos/métodos , Oócitos/fisiologia , Oogênese/fisiologia , Folículo Ovariano
2.
Clin Endocrinol (Oxf) ; 98(4): 567-577, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36372988

RESUMO

OBJECTIVE: Growth differentiation factor-9 (GDF9) and bone morphogenetic protein-15 (BMP15) are critical paracrine regulators of female fertility and are predominantly expressed by oocytes. However, it is unknown if serum concentrations reflect changes in ovarian function and/or reproductive endocrine disorders. This study aimed to determine if serum GDF9/BMP15 are associated with ovarian, pituitary, oestrogenic, androgenic and metabolic characteristics and the ovarian pathologies, polycystic ovarian morphology (PCOM) and polycystic ovary syndrome (PCOS). DESIGN: Women aged 21-45 years (n = 381) were included from a cross-sectional study at the National University Hospital, Singapore. PATIENTS: Participants were volunteers and patients with possible PCOS. MEASUREMENTS: Anthropometric measurements, transvaginal ultrasound scans and serum sampling were performed and a questionnairecompleted. Serum GDF9 and BMP15 concentrations were matched with menstrual cycle length, ovarian protein and steroid hormone production, pituitary hormone production and metabolic assessments in women with PCOM or PCOS and those with neither (control). RESULTS: Serum GDF9 and BMP15 were detectable in 40% and 41% of women, respectively and were positively correlated with each other (r = 0.08, p = 0.003). GDF9, but not BMP15, was positively correlated with ovarian volume (p = 0.02) and antral follicle count (AFC) (p = 0.004), but not with anti-Müllerian hormone (p = 0.05). However, serum GDF9 and BMP15 concentrations were not significantly different between control, PCOM and PCOS women, nor associated with androgenic or metabolic PCOS features. However, the relationship between GDF9 and AFC differed between control, PCOM and PCOS women (p = 0.02). CONCLUSIONS: Serum GDF9 and BMP15 concentrations somewhat reflect ovarian but not androgenic or metabolic characteristics of PCOS, with increased GDF9 reflecting high AFC as seen in PCOM/PCOS.


Assuntos
Síndrome do Ovário Policístico , Feminino , Humanos , Folículo Ovariano/patologia , Estudos Transversais , Oócitos , Hormônio Antimülleriano , Proteína Morfogenética Óssea 15/metabolismo , Fator 9 de Diferenciação de Crescimento/metabolismo
3.
Mol Metab ; 65: 101583, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36096453

RESUMO

Polycystic ovary syndrome (PCOS) is a common endocrine disorder, defined by reproductive and endocrine abnormalities, with metabolic dysregulation including obesity, insulin resistance and hepatic steatosis. Recently, it was found that skeletal muscle insulin sensitivity could be improved in obese, post-menopausal, pre-diabetic women through treatment with nicotinamide mononucleotide (NMN), a precursor to the prominent redox cofactor nicotinamide adenine dinucleotide (NAD+). Given that PCOS patients have a similar endocrine profile to these patients, we hypothesised that declining NAD levels in muscle might play a role in the pathogenesis of the metabolic syndrome associated with PCOS, and that this could be normalized through NMN treatment. Here, we tested the impact of NMN treatment on the metabolic syndrome of the dihydrotestosterone (DHT) induced mouse model of PCOS. We observed lower NAD levels in the muscle of PCOS mice, which was normalized by NMN treatment. PCOS mice were hyperinsulinaemic, resulting in increased adiposity and hepatic lipid deposition. Strikingly, NMN treatment completely normalized these aspects of metabolic dysfunction. We propose that addressing the decline in skeletal muscle NAD levels associated with PCOS can normalize insulin sensitivity, preventing compensatory hyperinsulinaemia, which drives obesity and hepatic lipid deposition, though we cannot discount an impact of NMN on other tissues to mediate these effects. These findings support further investigation into NMN treatment as a new therapy for normalizing the aberrant metabolic features of PCOS.


Assuntos
Hiperandrogenismo , Resistência à Insulina , Síndrome Metabólica , Síndrome do Ovário Policístico , Animais , Di-Hidrotestosterona/metabolismo , Feminino , Humanos , Hiperandrogenismo/metabolismo , Resistência à Insulina/fisiologia , Lipídeos , Síndrome Metabólica/metabolismo , Camundongos , Músculo Esquelético/metabolismo , NAD/metabolismo , Mononucleotídeo de Nicotinamida/metabolismo , Obesidade/metabolismo , Síndrome do Ovário Policístico/metabolismo
4.
Am J Physiol Endocrinol Metab ; 323(2): E145-E158, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35658542

RESUMO

Polycystic ovary syndrome (PCOS) is a common, multifactorial disorder characterized by endocrine, reproductive, and metabolic dysfunction. As the etiology of PCOS is unknown, there is no cure and symptom-oriented treatments are suboptimal. Hyperandrogenism is a key diagnostic trait, and evidence suggests that androgen receptor (AR)-mediated actions are critical to PCOS pathogenesis. However, the key AR target sites involved remain to be fully defined. Adipocyte and muscle dysfunction are proposed as important sites involved in the manifestation of PCOS traits. We investigated the role of AR signaling in white adipose tissue (WAT), brown adipose tissue (BAT), and skeletal muscle in the development of PCOS in a hyperandrogenic PCOS mouse model. As expected, dihydrotestosterone (DHT) exposure induced key reproductive and metabolic PCOS traits in wild-type (WT) females. Transplantation of AR-insensitive (AR-/-) WAT or BAT from AR knockout females (ARKO) into DHT-treated WT mice ameliorated some metabolic PCOS features, including increased body weight, adiposity, and adipocyte hypertrophy, but not reproductive PCOS traits. In contrast, DHT-treated ARKO female mice transplanted with AR-responsive (AR+/+) WAT or BAT continued to resist developing PCOS traits. DHT-treated skeletal muscle-specific AR knockout females (SkMARKO) displayed a comparable phenotype with that of DHT-treated WT females, with full development of PCOS traits. Taken together, these findings infer that both WAT and BAT, but less likely skeletal muscle, are key sites of AR-mediated actions involved in the experimental pathogenesis of metabolic PCOS traits. These data further support targeting adipocyte AR-driven pathways in future research aimed at developing novel therapeutic interventions for PCOS.NEW & NOTEWORTHY Hyperandrogenism is a key feature in the pathogenesis of polycystic ovary syndrome (PCOS); however, the tissue sites of androgen receptor (AR) signaling are unclear. In this study, AR signaling in white and brown adipose tissue, but less likely in skeletal muscle, was found to be involved in the development of metabolic PCOS traits, highlighting the importance of androgen actions in adipose tissue and obesity in the manifestation of metabolic disturbances.


Assuntos
Tecido Adiposo Marrom , Tecido Adiposo , Androgênios , Hiperandrogenismo , Síndrome do Ovário Policístico , Tecido Adiposo/metabolismo , Tecido Adiposo Marrom/metabolismo , Androgênios/farmacologia , Animais , Di-Hidrotestosterona/farmacologia , Modelos Animais de Doenças , Feminino , Hiperandrogenismo/genética , Hiperandrogenismo/metabolismo , Camundongos , Músculo Esquelético/metabolismo , Fenótipo , Síndrome do Ovário Policístico/metabolismo , Receptores Androgênicos/genética
5.
Reprod Biomed Online ; 44(3): 573-581, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35151576

RESUMO

RESEARCH QUESTION: Do women receiving corifollitropin alfa with a gonadotrophin-releasing hormone (GnRH) antagonist experience less emotional and/or physical exhaustion than women receiving standard of care gonadotrophin with daily administration of GnRH agonist or antagonist? DESIGN: The CoRifollitropin EvAluation in PracTicE (CREATE) study was a prospective observational study of fertility clinics in 17 countries in Europe and the Asia-Pacific region. Women undergoing IVF were categorized by treatment. Group A received single-dose corifollitropin alfa plus a GnRH antagonist; group B received usual care daily gonadotrophin regimens with a GnRH agonist or antagonist; and group B1i received daily GnRH agonist injections. For the primary analysis, two items from the Controlled Ovarian Stimulation Impact questionnaire were used to assess the level of emotional and physical exhaustion associated with ovarian stimulation. Secondary end-points included the impact of ovarian stimulation-related healthcare resource use. RESULTS: No statistical difference was found between the percentage of participants reporting emotional exhaustion in group A (11.6%) and B (13.1%) or the percentage reporting being 'often' or 'always' physically exhausted. More participants in group B1i (16.4%) reported being emotionally exhausted 'often' or 'always' during ovarian stimulation compared with group A (11.6%; P = 0.026). Patient questionnaire scores for psychological impact were higher in group A compared with group B, indicating less negative impact (72.7 versus 70.9; P = 0.004). Group A had fewer clinic visits, physician consultations, nurse contacts and transvaginal ultrasound scans (all P < 0.001) than group B1. CONCLUSIONS: Treatment with corifollitropin alfa resulted in similar or numerically small differences in psychological impact and lower clinic service use compared with daily gonadotrophin regimens.


Assuntos
Fertilização in vitro , Infertilidade Feminina , Atenção à Saúde , Feminino , Fertilização in vitro/métodos , Hormônio Foliculoestimulante Humano/uso terapêutico , Hormônio Liberador de Gonadotropina , Antagonistas de Hormônios , Humanos , Infertilidade Feminina/tratamento farmacológico , Indução da Ovulação/métodos , Gravidez , Taxa de Gravidez
6.
Nat Commun ; 11(1): 5262, 2020 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-33067453

RESUMO

Lifestyle, mainly dietary, interventions are first-line treatment for women with polycystic ovary syndrome (PCOS), but the optimal diet remains undefined. We combined a hyperandrogenized PCOS mouse model with a systematic macronutrient approach, to elucidate the impact of dietary macronutrients on the development of PCOS. We identify that an optimum dietary macronutrient balance of a low protein, medium carbohydrate and fat diet can ameliorate key PCOS reproductive traits. However, PCOS mice display a hindered ability for their metabolic system to respond to diet variations, and varying macronutrient balance did not have a beneficial effect on the development of metabolic PCOS traits. We reveal that PCOS traits in a hyperandrogenic PCOS mouse model are ameliorated selectively by diet, with reproductive traits displaying greater sensitivity than metabolic traits to dietary macronutrient balance. Hence, providing evidence to support the development of evidence-based dietary interventions as a promising strategy for the treatment of PCOS, especially reproductive traits.


Assuntos
Nutrientes/metabolismo , Síndrome do Ovário Policístico/metabolismo , Animais , Dieta , Dieta com Restrição de Proteínas , Feminino , Humanos , Estilo de Vida , Camundongos , Camundongos Endogâmicos C57BL , Nutrientes/análise , Síndrome do Ovário Policístico/dietoterapia
7.
Endocrinology ; 161(7)2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32301482

RESUMO

Polycystic ovary syndrome (PCOS) is a complex disorder characterized by endocrine, reproductive, and metabolic abnormalities. Despite PCOS being the most common endocrinopathy affecting women of reproductive age, the etiology of PCOS is poorly understood, so there is no cure and symptomatic treatment is suboptimal. Hyperandrogenism is the most consistent feature observed in PCOS patients, and recently aberrant neuroendocrine signaling and adipose tissue function have been proposed as playing a role in the development of PCOS. To investigate the role of adipose tissue and the brain as key sites for androgen receptor (AR)-mediated development of PCOS, we combined a white and brown adipose and brain-specific AR knockout (AdBARKO) mouse model with a dihydrotestosterone (DHT)-induced mouse model of PCOS. As expected, in wildtype (WT) control females, DHT exposure induced the reproductive PCOS traits of cycle irregularity, ovulatory dysfunction, and reduced follicle health, whereas in AdBARKO females, DHT did not produce the reproductive features of PCOS. The metabolic PCOS characteristics of increased adiposity, adipocyte hypertrophy, and hepatic steatosis induced by DHT in WT females were not evident in DHT-treated AdBARKO females, which displayed normal white adipose tissue weight and no adipocyte hypertrophy or liver steatosis. Dihydrotestosterone treatment induced increased fasting glucose levels in both WT and AdBARKO females. These findings demonstrate that adipose tissue and the brain are key loci of androgen-mediated actions involved in the developmental origins of PCOS. These data support targeting adipocyte and neuroendocrine AR-driven pathways in the future development of novel therapeutic strategies for PCOS.


Assuntos
Tecido Adiposo/metabolismo , Encéfalo/metabolismo , Modelos Animais de Doenças , Síndrome do Ovário Policístico/metabolismo , Receptores Androgênicos/fisiologia , Animais , Di-Hidrotestosterona , Feminino , Masculino , Camundongos Knockout , Síndrome do Ovário Policístico/etiologia
8.
J Endocrinol ; 245(3): 381-395, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32229702

RESUMO

As the mechanistic basis of polycystic ovary syndrome (PCOS) remains unknown, current management relies on symptomatic treatment. Hyperandrogenism is a major PCOS characteristic and evidence supports it playing a key role in PCOS pathogenesis. Classically, androgens can act directly through the androgen receptor (AR) or, indirectly, following aromatization, via the estrogen receptor (ER). We investigated the mechanism of androgenic actions driving PCOS by comparing the capacity of non-aromatizable dihydrotestosterone (DHT) and aromatizable testosterone to induce PCOS traits in WT and Ar-knockout (ARKO) mice. DHT and testosterone induced the reproductive PCOS-like features of acyclicity and anovulation in WT females. In ARKO mice, DHT did not cause reproductive dysfunction; however, testosterone treatment induced irregular cycles and ovulatory disruption. These findings indicate that direct AR actions and indirect, likely ER, actions of androgens are important mediators of PCOS reproductive traits. DHT, but not testosterone, induced an increase in body weight, body fat, serum cholesterol and adipocyte hypertrophy in WT mice, but neither androgen induced these metabolic features in ARKO mice. These data infer that direct AR-driven mechanisms are key in driving the development of PCOS metabolic traits. Overall, these findings demonstrate that differing PCOS traits can be mediated via different steroid signaling pathways and indicate that a phenotype-based treatment approach would ensure effective targeting of the underlying mechanisms.


Assuntos
Androgênios/metabolismo , Síndrome do Ovário Policístico/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Hiperandrogenismo/metabolismo , Camundongos , Camundongos Knockout , Receptores Androgênicos/metabolismo , Reprodução/fisiologia , Transdução de Sinais/fisiologia , Testosterona/metabolismo
9.
JCO Glob Oncol ; 6: 317-330, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-35275745

RESUMO

Fertility preservation in the cancer setting, known as oncofertility, is a field that requires cross-disciplinary interaction between physicians, basic scientists, clinical researchers, ethicists, lawyers, educators, and religious leaders. Funded by the National Institutes of Health, the Oncofertility Consortium (OC) was formed to be a scientifically grounded, transparent, and altruistic resource, both intellectual and monetary, for building this new field of practice capable of addressing the unique needs of young patients with cancer. The OC has expanded its attention to include other nonmalignant conditions that can threaten fertility, and the work of the OC now extends around the globe, involving partners who together have created a community of shared effort, resources, and practices. The OC creates materials that are translated, disseminated, and amended by all participants in the field, and local programs of excellence have developed worldwide to accelerate the pace and improve the quality of oncofertility research and practice. Here we review the global oncofertility programs and the capacity building activities that strengthen these research and clinical programs, ultimately improving patient care.

10.
Endocrinology ; 160(10): 2298-2313, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31211369

RESUMO

Oocyte-secreted factors bone morphogenetic protein 15 (BMP15) and growth differentiation factor 9 (GDF9) are critical for folliculogenesis and fertility. This study developed ELISAs for the measurement of BMP15 and GDF9 in serum and investigated their usefulness as biomarkers of female reproductive function. Serum samples were obtained from women undergoing infertility treatments (n = 154) and from perimenopausal and postmenopausal women (n = 28). Serum concentrations of BMP15 and GDF9 were analyzed in women relative to age, anti-Müllerian hormone, number of oocytes retrieved, and polycystic ovary syndrome (PCOS) after superovulation for in vitro fertilization. BMP15 and GDF9 immunoassays were validated for specificity, sensitivity (24 and 26 pg/mL, respectively), and reproducibility. BMP15 and GDF9 were detectable in 61% and 29% of women, respectively. BMP15 and GDF9 varied 64-fold and 15-fold, respectively, between women, but they did not change within subjects following ovarian stimulation with gonadotropins. Serum GDF9 concentration, but not BMP15 concentration, was associated with oocyte number retrieved in patients without PCOS (P = 0.018). GDF9 and BMP15 associations with oocyte number differed significantly (P < 0.05) with PCOS status. GDF9 concentrations were lower in poor responders (women with fewer than four oocytes retrieved or with cancelled cycles; P = 0.020). Serum BMP15, but not GDF9, was lower in women >55 years of age, compared with women of reproductive age (P < 0.01). This study develops and validates immunoassays to quantitate BMP15 and GDF9 in human serum and to correlate concentrations with female reproductive potential. Although assay sensitivities require improvement, this study demonstrates the diagnostic potential of oocyte-secreted BMP15 and GDF9 as serum biomarkers in reproductive medicine.


Assuntos
Proteína Morfogenética Óssea 15/metabolismo , Fertilização in vitro , Gonadotropinas/farmacologia , Fator 9 de Diferenciação de Crescimento/metabolismo , Infertilidade Feminina/metabolismo , Síndrome do Ovário Policístico/metabolismo , Adulto , Biomarcadores/sangue , Biomarcadores/química , Proteína Morfogenética Óssea 15/química , Proteína Morfogenética Óssea 15/genética , Ensaio de Imunoadsorção Enzimática , Feminino , Líquido Folicular/química , Regulação da Expressão Gênica/efeitos dos fármacos , Fator 9 de Diferenciação de Crescimento/química , Fator 9 de Diferenciação de Crescimento/genética , Humanos , Oócitos/metabolismo , Folículo Ovariano , Ovário/patologia , Síndrome do Ovário Policístico/sangue , Reprodutibilidade dos Testes , Superovulação
11.
Med Sci (Basel) ; 7(7)2019 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-31247909

RESUMO

In clomiphene-citrate-resistant anovulatory women with polycystic ovary syndrome (PCOS) and no other infertility factors, either metformin combined with clomiphene citrate or gonadotrophins could be used as a second-line pharmacological therapy, although gonadotrophins are more effective. Gonadotrophins could also be used as a second-line pharmacological therapy in anovulatory women with PCOS and clomiphene-citrate-failure. Laparoscopic ovarian surgery can also be used as a second-line therapy for ovulation induction in anovulatory women with clomiphene-citrate-resistant PCOS and no other infertility factors. The usefulness of letrozole as a second-line pharmacological treatment for ovulation induction in clomiphene-citrate-resistant women with PCOS requires further research. In terms of improving fertility, both pharmacological anti-obesity agents and bariatric surgery should be considered an experimental therapy in anovulatory women with PCOS and no other infertility factors. Where first- or second-line ovulation induction therapies have failed, in vitro fertilization (IVF)/ intracytoplasmic sperm injection (ICSI) could be offered as a third-line therapy in women with PCOS in the absence of an absolute indication for IVF/ICSI. For women with PCOS undergoing IVF/ICSI treatment, the gonadotropin-releasing hormone (GnRH) antagonist protocol is preferred and an elective frozen embryo transfer strategy could be considered. In assisted conception units with sufficient expertise, in-vitro maturation (IVM) of oocytes could be offered to women with PCOS.

12.
Endocrinology ; 160(4): 853-862, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30753406

RESUMO

The oocyte-secreted factors bone morphogenetic protein 15 (BMP15) and growth differentiation factor 9 (GDF9) interact functionally, and it is hypothesized that this interaction may be mediated by formation of a GDF9:BMP15 heterodimer termed cumulin. GDF9 and BMP15 regulate folliculogenesis and ovulation rate and have been shown to regulate inhibin and activin, local regulators of folliculogenesis. The objective of this study was to determine whether cumulin regulates granulosa cell inhibin and activin production and whether this requires cooperation with FSH. Human granulosa-lutein (hGL) cells collected from patients undergoing in vitro fertilization were cultured with or without FSH with various forms of recombinant cumulin (native and cysteine mutants, with or without the prodomains), and cysteine mutant GDF9 or BMP15. Messenger RNA expression of the subunits of inhibins/activins (INHA, INHBA, INHBB) and secretion of inhibin A, inhibin B, and activin B were measured. Mature forms and proforms of cumulin stimulated comparable INHBB mRNA expression and secretion of inhibin B and activin B, whereas GDF9 or BMP15 exhibited no effect. Cumulin, but not GDF9 or BMP15, interacted synergistically with FSH to increase INHBB mRNA and inhibin B expression. FSH markedly stimulated INHA, which encodes the α subunit of inhibin A/B, and suppressed activin B. Cumulin with or without FSH did not significantly alter inhibin A. Together these data demonstrate that cumulin, but not GDF9 or BMP15, exerts paracrine control of FSH-induced regulation of inhibin B and activin B. The prodomains of cumulin may have a minimal role in its actions on granulosa cells.


Assuntos
Ativinas/metabolismo , Proteína Morfogenética Óssea 15/farmacologia , Hormônio Foliculoestimulante/farmacologia , Fator 9 de Diferenciação de Crescimento/farmacologia , Inibinas/metabolismo , Células Lúteas/metabolismo , Oócitos/metabolismo , Gonadotropina Coriônica/farmacologia , Feminino , Humanos , Células Lúteas/efeitos dos fármacos , Recuperação de Oócitos , Oócitos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
13.
Endocrinology ; 160(3): 699-715, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30657917

RESUMO

Polycystic ovary syndrome (PCOS) is a common cause of female infertility. Hyperandrogenism is both a major symptom and key diagnostic trait of PCOS; however, the direct impact of this androgen excess on ovarian dynamics is unclear. By combining a DHT-induced PCOS mouse model with an ex vivo follicle culture system, we investigated the impact of hyperandrogenism on ovarian function. Ovaries from PCOS mice exhibited the characteristic polycystic ovary morphology with numerous large cystic follicles and no corpora lutea present. Isolation and individual culture of preantral and antral follicles from PCOS mice resulted in slower growth rates during 5 days compared with the follicles isolated from control mice (P < 0.01). In contrast, preovulatory follicles from PCOS mice exhibited a significant increase in growth rate compared with controls (P < 0.01). Preantral follicles from PCOS ovaries maintained comparable follicular health as control follicles, but antral and preovulatory PCOS follicles exhibited reduced follicle health (P < 0.01) and survival rates (P < 0.01). Compared with controls, PCOS females also exhibited a poorer response to hyperstimulation (P < 0.01), impaired oocyte function evident by increased levels of reactive oxygen species (P < 0.01), and a reduction in on-time embryo development (P < 0.01). These results demonstrate that prolonged exposure to androgen excess leads to aberrant follicle development, which persists even after removal from the hyperandrogenic environment, causing perturbed follicular developmental trajectories. These findings indicate that an in vivo hyperandrogenic environment in patients with PCOS may intrinsically induce detrimental effects on follicles and oocytes.


Assuntos
Hiperandrogenismo/fisiopatologia , Folículo Ovariano/fisiopatologia , Síndrome do Ovário Policístico/fisiopatologia , Animais , Modelos Animais de Doenças , Desenvolvimento Embrionário , Feminino , Camundongos Endogâmicos C57BL , Oócitos/metabolismo , Folículo Ovariano/enzimologia , Folículo Ovariano/crescimento & desenvolvimento , Indução da Ovulação , Estresse Oxidativo , Progesterona/metabolismo
14.
Trends Endocrinol Metab ; 29(12): 841-852, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30195991

RESUMO

Polycystic ovary syndrome (PCOS) is the most common endocrine condition in reproductive-aged women. It is characterized by reproductive, endocrine, metabolic, and psychological features. The cause of PCOS is unknown, thus there is no cure and its management remains suboptimal because it relies on the ad hoc empirical management of symptoms only. We review here the strong support for PCOS having a neuroendocrine origin. In particular, we focus on the role of aberrant hypothalamic-pituitary function and associated hyperandrogenism, and their role as major drivers of the mechanisms underpinning the development of PCOS. This important information now provides a target site and a potential mechanism for the future development of novel, targeted, and mechanism-based effective therapies for the treatment of PCOS.


Assuntos
Encéfalo/metabolismo , Hiperandrogenismo , Sistemas Neurossecretores , Síndrome do Ovário Policístico , Animais , Feminino , Humanos , Hiperandrogenismo/complicações , Hiperandrogenismo/metabolismo , Sistemas Neurossecretores/metabolismo , Sistemas Neurossecretores/fisiopatologia , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/etiologia , Síndrome do Ovário Policístico/metabolismo , Síndrome do Ovário Policístico/fisiopatologia
15.
Reprod Biomed Online ; 36(5): 491-499, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29503209

RESUMO

Ovarian tissue is increasingly being collected from cancer patients and cryopreserved for fertility preservation. While the only available option to restore fertility is autologous transplantation, this treatment is not appropriate for all patients due to the risk of reintroducing cancer cells and causing disease recurrence. Harnessing the full reproductive potential of this tissue to restore fertility requires the development of culture systems that support oocyte development from the primordial follicle stage. While this has been achieved in the mouse, the goal of obtaining oocytes of sufficient quality to support embryo development has not been reached in higher mammals despite decades of effort. In vivo, primordial follicles gradually exit the resting pool, whereas when primordial follicles are placed into culture, global activation of these follicles occurs. Therefore, the addition of a factor(s) that can regulate primordial follicle activation in vitro may be beneficial to the development of culture systems for ovarian tissue from cancer patients. Several factors have been observed to inhibit follicle activation, including anti-Müllerian hormone, stromal-derived factor 1 and members of the c-Jun-N-terminal kinase pathway. This review summarizes the findings from studies of these factors and discusses their potential integration into ovarian tissue culture strategies for fertility preservation.


Assuntos
Preservação da Fertilidade/métodos , Folículo Ovariano/crescimento & desenvolvimento , Técnicas de Cultura de Tecidos , Hormônio Antimülleriano/farmacologia , Quimiocina CXCL12/farmacologia , Feminino , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Folículo Ovariano/efeitos dos fármacos , Transdução de Sinais
16.
Hum Reprod ; 32(5): 1033-1045, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28333356

RESUMO

STUDY QUESTION: Can controlled ovarian hyperstimulation (COH) for fertility preservation be effectively conducted in women with breast cancer without worsening their prognosis? SUMMARY ANSWER: COH with co-administration of letrozole suppresses oestradiol levels without significantly impacting oocyte yield or decreasing disease-free survival rates. WHAT IS KNOWN ALREADY: Oestradiol has the capacity to stimulate the proliferation of breast cancer cells. COH can cause oestradiol levels to rise by an order of magnitude above physiological levels. Concern exists regarding the effect of supra-physiological oestradiol levels in women with a recent diagnosis of breast cancer. STUDY DESIGN, SIZE, DURATION: A systematic review of the literature was performed using MEDLINE (PubMed database), EMBASE and the Cochrane Library. The search was restricted to articles written in English. No restrictions regarding the date of publication were applied. Safety was assessed in terms of relapse rates and cancer-related mortality rates. Peak oestradiol concentrations were recorded for different stimulation protocols. Efficacy was measured in terms of the total number of oocytes collected and proportion of mature oocytes. The primary outcome was mortality/recurrence in premenopausal women with Stage I-IIIB breast cancer who underwent COH in the immediate post-operative period, prior to chemotherapy. PARTICIPANTS/MATERIALS, SETTING, METHODS: This is a systematic review of randomized control trials (RCTs), case control and cohort studies reporting on the primary outcome of mortality/recurrence after COH in women with early breast cancer, or secondary outcomes of oocyte yield and peak oestrogen concentration. Owing to the small number of RCTs available, other study types were included. The last electronic search was run in April 2016. Two prospective non-randomized studies reported relapse and breast cancer-related mortality rates in 397 women with breast cancer, of whom 149 underwent COH. Twelve studies reported the peak oestradiol concentrations of 882 women undergoing COH with letrozole co-administration. Four studies compared the oocyte yield of 248 women who underwent COH plus letrozole with 254 women who underwent standard COH. Two studies compared peak oestradiol concentrations and oocyte yield in 61 women who underwent COH with tamoxifen co-administration and 49 women who underwent COH without tamoxifen. One study compared letrozole and tamoxifen co-administration, and another study compared the co-administration of letrozole and anastrozole. MAIN RESULTS AND THE ROLE OF CHANCE: The search identified 1002 records of which 15 were included in the final analysis. There was no evidence of a decline in relapse-free survival rates in the two studies of women with breast cancer who received COH with letrozole co-administration compared with women who did not undergo fertility preservation procedures. The largest of these studies reported recurrences in 6/120 (5.0%) women who received COH plus letrozole compared with 12/217 (5.5%) women who did not undergo COH (mean follow-up 5.0 versus 6.9 years; hazard ratio for recurrence 0.77, 95%CI 0.28-2.13). Conclusions regarding women with breast cancer who received tamoxifen during COH could not be made due to insufficient data. Peak oestradiol concentrations (338-829 pg/ml) were suppressed by letrozole when commenced on Days 2-3, with no decrease in oocyte yield. Tamoxifen does not suppress oestradiol concentrations, but may convey protection via its inhibitory action on the oestrogen receptor. LIMITATIONS, REASONS FOR CAUTION: Any statements regarding the safety of COH in women with breast cancer are based on a limited number of observational studies. High quality evidence is unlikely to become available for ethical and practical reasons. Whilst available data do not indicate a decline in disease-free survival, a small effect cannot be excluded. Breast cancers are heterogeneous in their genetic profile and receptor status, making the results of studies difficult to generalize to individual cases. The implication of alterations in other hormone levels such as androgens, progestins or vascular endothelial growth factor secondary to COH in women with breast cancer has not been quantified. WIDER IMPLICATIONS OF THE FINDINGS: The co-administration of 5 mg of letrozole daily commencing on Day 2 and continuing throughout COH is recommended as it reduces peak oestradiol concentrations without significantly decreasing oocyte yield. The use of a GnRH agonist trigger is beneficial as oestradiol concentrations rapidly decrease post-administration and rates of ovarian hyperstimulation are lower than with an hCG trigger, without a corresponding reduction in clinical pregnancy or live birth rates in cryopreservation cycles. The protective effect of tamoxifen has not been evaluated although theoretically may be of benefit due to its action on the oestrogen receptor. STUDY FUNDING/COMPETING INTEREST(S): None. REGISTRATION NUMBER: None.


Assuntos
Neoplasias da Mama/complicações , Preservação da Fertilidade/métodos , Infertilidade Feminina/etiologia , Indução da Ovulação/métodos , Feminino , Preservação da Fertilidade/efeitos adversos , Humanos , Indução da Ovulação/efeitos adversos
17.
J Glob Oncol ; 2(2): 83-96, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27284576

RESUMO

Fertility preservation in the cancer setting, known as oncofertility, is a field that requires cross-disciplinary interaction between physicians, basic scientists, clinical researchers, ethicists, lawyers, educators, and religious leaders. Funded by the National Institutes of Health, the Oncofertility Consortium (OC) was formed to be a scientifically grounded, transparent, and altruistic resource, both intellectual and monetary, for building this new field of practice capable of addressing the unique needs of young patients with cancer. The OC has expanded its attention to include other nonmalignant conditions that can threaten fertility, and the work of the OC now extends around the globe, involving partners who together have created a community of shared effort, resources, and practices. The OC creates materials that are translated, disseminated, and amended by all participants in the field, and local programs of excellence have developed worldwide to accelerate the pace and improve the quality of oncofertility research and practice. Here we review the global oncofertility programs and the capacity building activities that strengthen these research and clinical programs, ultimately improving patient care.

19.
Fertil Steril ; 105(2): 417-22, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26607023

RESUMO

OBJECTIVE: To describe the findings of a national inquiry into cases of misdiagnosis of miscarriage in the Republic of Ireland and to report the results of implementation of the findings of the inquiry, including investment in new equipment and training, new national guidelines, and rigorous annual audit of early pregnancy units. DESIGN: Narrative description of the inquiry and its findings and results of a subsequent audit. SETTING: Not applicable. PATIENT(S): Women with problems of bleeding and/or pain in early pregnancy who were erroneously diagnosed as having a nonviable intrauterine pregnancy. INTERVENTION(S): After two cases of misdiagnosis of miscarriage that were widely reported in the Republic of Ireland in June 2010, a Miscarriage Misdiagnosis Review Team was commissioned by the Irish Health Service Executive (HSE) to undertake a national review of other possible cases of misdiagnosis of miscarriage. The Review Team made a series of recommendations that were subsequently implemented in full. The results of the implementation of the findings of the Review Team have been the subject of three annual audits across the country. MAIN OUTCOME MEASURE(S): The main outcome measure was the occurrence of misdiagnosis of miscarriage in the Republic of Ireland before and after implementation of the findings of the Review Team. RESULT(S): Twenty-four confirmed cases of misdiagnosis of miscarriage were identified, mostly occurring between 2005 and 2010. Analysis led to a series of recommendations by the Review Team, which were implemented in full by the HSE. Over € 3 million was provided to fund implementation; 26 high-quality gynecological ultrasound machines were purchased to reequip 19 units involved in provision of care to women with suspected miscarriage. There was further allocation of resources for new equipment and improvement in the management and staffing of early pregnancy units across Ireland, with each center now having a dedicated and properly staffed Early Pregnancy Assessment Unit. A national training program in the management of early pregnancy problems has been implemented, along with regular national meetings to discuss early pregnancy problems. National clinical guidelines on the diagnosis and management of miscarriage for implementation have been distributed to all hospitals. CONCLUSION(S): No cases of miscarriage misdiagnosis were identified in any of the three annual audits, suggesting that implementation of the findings of the review has been successful. We believe that this is the first report of national change in practice leading to improvement in clinical outcomes in the management of suspected miscarriage.


Assuntos
Aborto Espontâneo/diagnóstico , Erros de Diagnóstico , Programas Nacionais de Saúde/normas , Cuidado Pré-Natal/normas , Melhoria de Qualidade/normas , Indicadores de Qualidade em Assistência à Saúde/normas , Aborto Espontâneo/economia , Orçamentos , Erros de Diagnóstico/economia , Feminino , Fidelidade a Diretrizes , Custos de Cuidados de Saúde , Alocação de Recursos para a Atenção à Saúde , Necessidades e Demandas de Serviços de Saúde , Humanos , Auditoria Médica , Programas Nacionais de Saúde/economia , Avaliação das Necessidades , Irlanda do Norte , Guias de Prática Clínica como Assunto , Valor Preditivo dos Testes , Gravidez , Cuidado Pré-Natal/economia , Desenvolvimento de Programas , Avaliação de Programas e Projetos de Saúde , Melhoria de Qualidade/economia , Indicadores de Qualidade em Assistência à Saúde/economia , Fatores de Tempo , Ultrassonografia Pré-Natal
20.
Aust N Z J Obstet Gynaecol ; 54(6): 581-5, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25476811

RESUMO

AIMS: To analyse the data from all controlled ovarian hyperstimulation antagonist cycles that used an agonist trigger and a freeze-all strategy to quantify the risk of ovarian hyperstimulation syndrome (OHSS) and subsequent pregnancy rates. MATERIALS AND METHODS: A retrospective study of all women attending fertility clinics at IVF Australia, Sydney, undergoing controlled ovarian hyperstimulation (COH) using an antagonist protocol that had a subsequent gonadotropin-releasing hormone (GnRH) agonist trigger and freezing of all oocytes or embryos. The primary outcome measure was to determine the rate of OHSS. The secondary outcome measure was the clinical pregnancy rate. RESULTS: We collected data for 123 women. 25.2% were undergoing oocyte freezing and 74.8% underwent embryo freezing. There were no cases of OHSS, either early or late onset. The pregnancy rate was 31.7% after the first frozen cycle transfer with a cumulative pregnancy rate of 50% after two frozen embryo transfers. CONCLUSION: Our results support the hypothesis that a GnRH agonist trigger and a freeze-all approach prevents OHSS with a good pregnancy rate.


Assuntos
Criopreservação , Embrião de Mamíferos , Fármacos para a Fertilidade Feminina/uso terapêutico , Hormônio Liberador de Gonadotropina/agonistas , Oócitos , Síndrome de Hiperestimulação Ovariana/prevenção & controle , Indução da Ovulação/efeitos adversos , Adulto , Feminino , Fertilização in vitro , Hormônio Foliculoestimulante/administração & dosagem , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Humanos , Leuprolida/uso terapêutico , Nafarelina/uso terapêutico , Recuperação de Oócitos , Síndrome de Hiperestimulação Ovariana/induzido quimicamente , Indução da Ovulação/métodos , Gravidez , Taxa de Gravidez , Estudos Retrospectivos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA