Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Biol Reprod ; 110(1): 130-139, 2024 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-37801701

RESUMO

The endocrinology regulating ovulation of the desired number of oocytes in the ovarian cycle is well described, particularly in mono-ovulatory species. Less is known about the characteristics that make one follicle suitable for ovulation while most other follicles die by atresia. Bromodeoxyuridine (BrdU) injection was used to characterize granulosa cell proliferation rates in developing ovarian follicles in the estrous cycle of mice. This methodology allowed identification of follicle diameters of secondary (80-130 µm), follicle-stimulating hormone (FSH)-sensitive (130-170 µm), FSH-dependent (170-350 µm), and preovulatory (>350 µm) follicles. Few preovulatory-sized follicles were present in the ovaries of mice at estrus, the beginning of the cycle. Progressive increases were seen at metestrus and diestrus, when full accumulation of the preovulatory cohort (~10 follicles) occurred. BrdU pulse-chase studies determined granulosa cell proliferation rates in the 24-48 h before the follicle reached the preovulatory stage. This showed that slow-growing follicles were not able to survive to the preovulatory stage. Mathematical modeling of follicle growth rates determined that the largest follicles at the beginning of the cycle had the greatest chance of becoming preovulatory. However, smaller follicles could enter the preovulatory follicle pool if low numbers of large antral follicles were present at the beginning of the cycle. In this instance, rapidly growing follicles had a clear selection advantage. The developing follicle pool displays heterogeneity in granulosa cell proliferation rates, even among follicles at the same stage of development. This parameter appears to influence whether a follicle can ovulate or become atretic.


Assuntos
Folículo Ovariano , Ovulação , Humanos , Feminino , Camundongos , Animais , Bromodesoxiuridina/metabolismo , Folículo Ovariano/metabolismo , Ovulação/fisiologia , Ovário , Hormônio Foliculoestimulante/farmacologia , Hormônio Foliculoestimulante/metabolismo
2.
Biol Reprod ; 109(5): 684-692, 2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-37552056

RESUMO

There are conflicting estimates of the duration of mouse primary follicle development. An accurate determination is needed for studies examining preantral follicle survival and mathematical modeling of folliculogenesis. Primary follicle granulosa cell proliferation rates are low and variable, which may explain the variation in duration estimates. In the present study, female C57Bl6/J mice were exposed to bromodeoxyuridine for 48 hours, to label the proliferating granulosa cells in a large proportion of primary follicles. The bromodeoxyuridine-containing water was then withdrawn and replaced with drug-free water and the mice were euthanized at 0, 1, 3, 6, 10, or 13 days post-bromodeoxyuridine withdrawal. Granulosa cells were bromodeoxyuridine labeled in 48% of primary follicles at day 0, but this decreased to 5% over the 13-day period, as the labeled primary follicles progressed to the secondary follicle stage. Curve-fitting estimated that the last of the bromodeoxyuridine-labeled primary follicles would progress to the secondary stage by 13.7 days. Mathematical models that assumed constant rates of primary follicle proliferation were fitted to the data, but the observed pattern of bromodeoxyuridine-labeled primary follicle disappearance could not be replicated. The level of immunoreactivity for bromodeoxyuridine and proliferating-cell nuclear antigen in primary follicles revealed follicles with no granulosa cell proliferation during the 48-h bromodeoxyuridine-exposure period had resumed proliferation 1 or 3 days later. Therefore, primary follicle granulosa cells proliferate after follicle activation, but proliferation rates gradually increase as the follicle develops. Prior estimates of primary follicle duration are inaccurate due to the assumption that follicles develop at a constant rate.


Assuntos
Células da Granulosa , Folículo Ovariano , Feminino , Camundongos , Animais , Bromodesoxiuridina , Folículo Ovariano/fisiologia , Células da Granulosa/fisiologia , Proliferação de Células , Água
3.
Biol Reprod ; 102(4): 915-922, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-31837140

RESUMO

Anti-Müllerian hormone (AMH) inhibits the activation of primordial follicles in the ovary. This causes an increased rate of ovarian reserve depletion in Amh-/- mice. The depletion of the ovarian reserve is responsible for the onset of menopause but age-related infertility occurs in advance of ovarian reserve depletion. To determine whether accelerated loss of primordial follicles leads to earlier onset infertility, Amh-/- and Amh+/+ females were paired with Amh+/+ stud males and birth rates were recorded across the females' reproductive lifespan. The number of primordial follicles remaining in the ovaries of Amh-/- and Amh+/+ females were quantified in two cohorts at 11-12 and 12-13 months of age. As expected, the ovarian reserve in the Amh-/- females became depleted approximately 1 month earlier than Amh+/+ females. However, no difference was observed in the cumulative number of births over the lifespan, nor were there any differences in mean littersize at any age. It is possible that the reproductive lifespan of mice is too short for sufficient divergence of primordial follicles numbers to cause differences in Amh-/- and Amh+/+ female fertility. An alternative explanation contradicts current thinking; the function of AMH may be unrelated to the longevity of the reproductive lifespan in female mice.


Assuntos
Hormônio Antimülleriano/genética , Fertilidade/fisiologia , Reserva Ovariana/fisiologia , Ovário/metabolismo , Animais , Hormônio Antimülleriano/metabolismo , Feminino , Infertilidade Feminina/genética , Infertilidade Feminina/metabolismo , Camundongos , Camundongos Knockout
4.
Reproduction ; 160(6): R145-R153, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33112785

RESUMO

Serum anti-Müllerian hormone (AMH) levels decrease after surgical treatment of ovarian endometrioma. This is the main reason that surgery for ovarian endometrioma endometriosis is not recommended before in vitro fertilization, unless the patient has severe pain or suspected malignant cysts. Furthermore, it has been suggested that ovarian endometrioma itself damages ovarian reserve. This raises two important challenges: (1) determining how to prevent surgical damage to the ovarian reserve in women with ovarian endometrioma and severe pain requiring surgical treatment and (2) deciding the best treatment for women with ovarian endometrioma without pain, who do not wish to conceive immediately. The mechanisms underlying the decline in ovarian reserve are potentially induced by both ovarian endometrioma and surgical injury but the relative contribution of each process has not been determined. Data obtained from various animal models and human studies suggest that hyperactivation of dormant primordial follicles caused by the local microenvironment of ovarian endometrioma (mechanical and/or chemical cues) is the main factor responsible for the decreased primordial follicle numbers in women with ovarian endometrioma. However, surgical injury also induces hyperactivation of dormant primordial follicles, which may further reduce ovarian reserve after removal of the endometriosis. Although further studies are required to elucidate the mechanisms underlying diminished ovarian reserve in women with ovarian endometrioma, the available data strongly suggests the need to prevent/minimize hyperactivation of dormant primordial follicles, regardless of whether surgery is performed, for better clinical management of ovarian endometrioma.


Assuntos
Endometriose/patologia , Infertilidade Feminina/patologia , Folículo Ovariano/patologia , Reserva Ovariana , Ovário/patologia , Endometriose/complicações , Feminino , Humanos , Infertilidade Feminina/etiologia
5.
Reprod Biomed Online ; 41(6): 1112-1121, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33046375

RESUMO

RESEARCH QUESTION: Does the relative distribution of anti-Müllerian hormone (AMH) isoforms differ between patients depending on their body mass index (BMI) and polycystic ovary syndrome (PCOS) status in serum and follicular fluid? DESIGN: Obese and normal weight patients (PCOS [n = 70]; non-PCOS [n = 37]) were selected for this case-control study in the serum. Between 2018 and 2019, obese (n = 19) and normal weight (n = 20) women with or without PCOS who were receiving IVF treatment were included in the follicular fluid study. The bio-banked serums and follicular fluid were tested for total AMH (proAMH and AMHN,C combined) and proAMH using an automatic analyzer. The AMH prohormone index (API = [proAMH]/[total AMH]x 100) was calculated as an inverse marker of conversion of proAMH to AMHN,C, with only the latter isoform that could bind to the AMH receptor complex. RESULTS: The API was not significantly different between controls and women with PCOS, whereas obese women had a lower API compared with their normal weight counterparts. Grouping PCOS and controls, a lower API was found in obese versus normal weight women, suggesting a greater conversion of proAMH to AMHN,C. The API in the serum was significantly correlated with metabolic parameters. In the follicular fluid, API is not different between obese and normal weight women independently of PCOS and is higher than in the concomitant serum. CONCLUSIONS: The proportion of inactive form of AMH in the serum is higher in normal weight versus obese women but not in the follicular fluid, independently of PCOS. The conversion of proAMH into the cleaved isoform is likely to occur in extra-ovarian tissues and to exacerbate in obese individuals.


Assuntos
Hormônio Antimülleriano/metabolismo , Líquido Folicular/metabolismo , Obesidade/metabolismo , Síndrome do Ovário Policístico/metabolismo , Adolescente , Adulto , Hormônio Antimülleriano/sangue , Hormônio Antimülleriano/química , Biomarcadores/sangue , Biomarcadores/metabolismo , Índice de Massa Corporal , Estudos de Casos e Controles , Feminino , Líquido Folicular/química , França/epidemiologia , Humanos , Obesidade/complicações , Obesidade/epidemiologia , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/epidemiologia , Isoformas de Proteínas/análise , Isoformas de Proteínas/sangue , Isoformas de Proteínas/metabolismo , Precursores de Proteínas/sangue , Precursores de Proteínas/metabolismo , Adulto Jovem
6.
Reprod Fertil Dev ; 31(5): 829-836, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30636191

RESUMO

The 2018 edition of the Society for Reproductive Biology's (SRB) Annual Meeting was a celebration of 50 years of Australian research into reproductive biology. The past 50 years has seen many important contributions to this field, and these advances have led to changes in practice and policy, improvements in the efficiency of animal reproduction and improved health outcomes. This conference review delivers a dedicated summary of the symposia, discussing emerging concepts, raising new questions and proposing directions forward. Notably, the symposia discussed in this review emphasised the impact that reproductive research can have on quality of life and the health trajectories of individuals. The breadth of the research discussed encompasses the central regulation of fertility and cyclicity, life course health and how the environment of gametes and embryos can affect subsequent generations, significant advances in our understanding of placental biology and pregnancy disorders and the implications of assisted reproductive technologies on population health. The importance of a reliable food supply and protection of endangered species is also discussed. The research covered at SRB's 2018 meeting not only recognised the important contributions of its members over the past 50 years, but also highlighted key findings and avenues for innovation moving forward that will enable the SRB to continue making significant contributions for the next 50 years.


Assuntos
Reprodução , Técnicas de Reprodução Assistida , Animais , Austrália , Fertilidade , Humanos , Pesquisa , Sociedades
7.
Biol Reprod ; 98(1): 54-62, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29177503

RESUMO

Anti-Müllerian hormone (AMH) is both a gonadal hormone and a putative paracrine regulator of neurons, the uterus, and the placenta. A mouse line with neuronal expression of AMH (Thy1.2-AMH) was generated to examine the role of paracrine AMH in the brain. The mice had normal behavior, but unexpectantly AMH was present in the circulation of the transgenic mice. Thy1.2-AMHTg/0 studs sired pups with a normal frequency, when mated with wild-type dams. In stark contrast, Thy1.2-AMHTg/0 dams rarely gave birth, with evidence of spontaneous midgestational abortion. This leads to the hypothesis that AMH influences the capacity of dams to carry concepti to term. This hypothesis was tested by mating AMH-deficient (Amh-/-), Thy1.2-AMHTg/0, and wild-type dams when 49-, 80-, and 111 days old, using proven wild-type studs. The litter sizes from the first two matings and the number of fetuses present on the 10th day of gestation of the third mating were recorded. Thy1.2-AMHTg/0 dams carried near normal numbers of midterm fetuses, but typically produced no pups, indicating that extensive late resorption of fetuses was occurring. Amh-/- dams exhibited a lesser reduction in litter size than the Thy1.2-AMHTg/0 dams, with no evidence of enhanced loss of fetuses. In conclusion, this study provides the first evidence that high AMH levels can cause a miscarriage phenotype and that the absence of AMH affects reproductive output.


Assuntos
Hormônio Antimülleriano/metabolismo , Animais , Animais Recém-Nascidos , Hormônio Antimülleriano/genética , Encéfalo/crescimento & desenvolvimento , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Tamanho da Ninhada de Vivíparos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Folículo Ovariano/fisiologia , Gravidez , Proteínas Recombinantes
8.
Mol Reprod Dev ; 84(7): 626-637, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28500669

RESUMO

The hypothesis that, in contrast to other transforming growth factor-beta (TGFß) superfamily ligands, the dose-response curve of Anti-Müllerian hormone (AMH) is unmodulated was tested by examining whether known TGFB superfamily modulators affect AMH signaling, using a P19/BRE luciferase reporter assay. AMHC and AMHN,C activated the reporter with an EC50 of approximately 0.5 nM. Follistatins (FS) produced concentration-dependent increases in AMHC - and AMHN,C -initiated reporter activity, with FS288 being more potent than FS315; however, the maximum bioactivity of AMH was not altered by either follistatin. Thirteen other TGFß regulators (Chordin, Chordin-like 1, Chordin-like 2, Differential screening-selected gene aberrative in neuroblastoma [DAN], Decorin, Endoglin, Follistatin-like 1, Follistatin-like 3, Follistatin-like 4, Noggin, α2 macroglobulin, TGFß receptor 3, Von Willebrand factor C domain-containing 2) had little or no effect. Surface plasmon resonance analysis showed no significant association between FS288 and AMHC , suggesting that FS288 indirectly regulates AMH signaling. Activin A, a direct target of FS288, did not itself induce reporter activity in P19 cells, but did prevent the FS288-induced increase in AMH signaling. Hence, local concentrations of FS288 and Activin A may influence the response of some cell types to AMH.


Assuntos
Hormônio Antimülleriano/química , Folistatina/química , Transdução de Sinais , Ressonância de Plasmônio de Superfície , Animais , Hormônio Antimülleriano/genética , Linhagem Celular , Folistatina/genética , Folistatina/metabolismo , Humanos , Camundongos
9.
Am J Physiol Endocrinol Metab ; 305(10): E1241-7, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24045871

RESUMO

Anti-Müllerian hormone (AMH) in blood is a marker of ovarian status in women and the presence of cryptic testes in babies. Despite this, the molecular form of AMH in blood has not been verified. AMH is synthesized as an inert proprotein precursor (proAMH), which can be cleaved to yield NH2-terminal (AMHN) and COOH-terminal (AMHC) fragments, that can complex noncovalently (AMHN,C). Developing males have 10-fold more AMH than young adults. We report here that human blood is a mixture of inactive proAMH and receptor-binding AMHN,C. The AMH in the blood of boys, men, and premenopausal women was immunoprecipitated using antibodies to the NH2- and COOH-terminal peptides. The precipitated proteins were then analyzed by Western blots, using recombinant proteins as markers. The glycosylation status of AMH was verified using deglycosylating enzymes. The NH2-terminal antibody precipitated a major protein that migrated alongside rhproAMH and was detected by anti-AMHN and anti-AMHC. This antibody also precipitated significant levels of AMHN and AMHC from all participants. Antibodies specific to AMHC precipitated rhAMHC but did not precipitate AMHC from human blood. Hence, all the AMHC in human blood appears to be bound to AMHN. Both AMHN and proAMH were glycosylated, independent of age and sex. In conclusion, boys and young adults have the same form of AMH, with a significant proportion being the inactive precursor. This raises the possibility that the endocrine functions of AMH are partly controlled by its cleavage in the target organ. The presence of proAMH in blood may confound the use of AMH for diagnosis.


Assuntos
Hormônio Antimülleriano/sangue , Fragmentos de Peptídeos/sangue , Adulto , Hormônio Antimülleriano/química , Pré-Escolar , Feminino , Células HEK293 , Humanos , Masculino , Pessoa de Meia-Idade , Complexos Multiproteicos/sangue , Complexos Multiproteicos/química , Fragmentos de Peptídeos/química , Ligação Proteica , Precursores de Proteínas/sangue , Precursores de Proteínas/química , Estrutura Terciária de Proteína , Adulto Jovem
10.
Endocrinology ; 164(8)2023 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-37337678

RESUMO

Serum anti-Müllerian hormone (AMH) is a biomarker for predicting antral follicle counts but there is no clear consensus on whether AMH is indicative of primordial follicle counts in humans. Mice were used as a model species in this study to obtain accurate follicle counts across the reproductive phase of life. Serum AMH was measured in 62 female C57Bl6/J mice aged 25 to 401 days. Primordial and primary follicles were determined by stereological counts and all secondary and antral follicles were counted in serial histological sections. Serum AMH was most strongly correlated with small- and medium-sized antral follicles. Immunohistochemistry and stepwise multiple regression confirmed that these follicle development stages are the key determinants of serum AMH, with little contribution from other stages. Primordial follicles were not found to have strong correlations with serum AMH or antral follicle counts, particularly in younger females, but the strength of the association appeared to increase with age. This result is likely attributed to high interindividual variation in primordial follicle activation and preantral follicle survival rates. Recent large studies in human populations have shown similar results but the primary limitation of these studies was that primordial follicle counts were determined from ovarian cortical biopsies, where regional variation in follicle distribution may affect the quality of the data. In the present study, whole ovaries were surveyed, eliminating this limitation. The findings indicate that primordial follicle counts are not closely related with either serum AMH or antral follicle counts in females in the early phase of the reproductive phase of life.


Assuntos
Hormônio Antimülleriano , Hormônios Peptídicos , Animais , Feminino , Camundongos , Hormônio Foliculoestimulante , Folículo Ovariano , Ovário
12.
Endocrine ; 76(1): 189-197, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34988934

RESUMO

PURPOSE: The ratio of the anti-Müllerian hormone (AMH) precursor (proAMH) to active AMH (AMHN,C) is higher in childhood than in adulthood but has never been quantified during adolescence. The ratio of proAMH to total AMH (AMH prohormone index, API) was examined during the puberty in healthy girls. The API was also compared between girls with and without polycystic ovary syndrome (PCOS) to determine if there were differences that could assist in PCOS diagnosis during adolescence. METHODS: Total AMH and proAMH were measured by immunoassay in a single-centre, cross-sectional observational study; 61 controls and 29 girls with PCOS were included in the study (age range 8-21 years). The API was calculated as proAMH as a percentage of total AMH. Differences in API between control and PCOS subjects and across age-groups were examined by Welch's ANOVA. The relationship between API and a range of metabolic parameters was examined by Pearson correlation. RESULTS: The API in healthy females increased between the ages of 10~15 years and declined from 15~20 years (p < 0.001). The API was negatively correlated with body mass index in the control (p = 0.04) and PCOS groups (p = 0.007). The API was associated with factors related to adiposity and lipid metabolism. The API was not significantly different in control girls and girls with PCOS. CONCLUSIONS: Higher API during adolescence suggests that proteolytic activation of proAMH is suppressed during this life stage. API was not different between control girls and girls with PCOS indicating that it is not useful in diagnosis of PCOS during adolescence.


Assuntos
Hormônios Peptídicos , Síndrome do Ovário Policístico , Adolescente , Adulto , Hormônio Antimülleriano/metabolismo , Biomarcadores/metabolismo , Índice de Massa Corporal , Criança , Estudos Transversais , Feminino , Humanos , Síndrome do Ovário Policístico/metabolismo , Fator de Crescimento Transformador beta , Adulto Jovem
13.
J Endocrinol ; 255(1): R1-R26, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-35980384

RESUMO

Polycystic ovary syndrome (PCOS) is a common endocrine condition characterised by a range of reproductive, endocrine, metabolic and psychological abnormalities. Reports estimate that around 10% of women of reproductive age are affected by PCOS, representing a significant prevalence worldwide, which poses a high economic health burden. As the origin of PCOS remains largely unknown, there is neither a cure nor mechanism-based treatments leaving patient management suboptimal and focused solely on symptomatic treatment. However, if the underlying mechanisms underpinning the development of PCOS were uncovered then this would pave the way for the development of new interventions for PCOS. Recently, there have been significant advances in our understanding of the underlying pathways likely involved in PCOS pathogenesis. Key insights include the potential involvement of androgens, insulin, anti-Müllerian hormone and transforming growth factor beta in the development of PCOS. This review will summarise the significant scientific discoveries on these factors that have enhanced our knowledge of the mechanisms involved in the development of PCOS and discuss the impact these insights may have in shaping the future development of effective strategies for women with PCOS.


Assuntos
Resistência à Insulina , Síndrome do Ovário Policístico , Androgênios/metabolismo , Hormônio Antimülleriano/metabolismo , Feminino , Humanos , Insulina , Síndrome do Ovário Policístico/metabolismo
14.
J Endocrinol ; 253(2): 53-62, 2022 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-35099409

RESUMO

Female anti-Müllerian hormone (AMH) overexpressing (Thy1.2-AMHTg/0) mice experience fetal resorption (miscarriage) by mid-gestation. This study examined whether the ovary, uterine implantation sites and hypothalamus are potential sites of AMH action, as AMH type-2 receptor (AMHR2) expression is reported in each tissue. Pregnancy in Thy1.2-AMHTg/0 mice was compared to wild-type (WT) mice via histological examination of implantation sites, hormone assays, embryo culture and embryo transfer. Uterine AMH and AMHR2 expression was examined by RT-qPCR and immunohistochemistry. The first signs of fetal resorption in the Thy1.2-AMHTg/0 dams occurred at embryonic day 9.5 (E9.5) with 100% of fetuses resorbing by E13.5. Cultured embryos from Thy1.2-AMHTg/0 dams had largely normal developmental rates but a small proportion experienced a minor developmental delay relative to embryos from WT dams. However, embryos transferred from WT donor females always failed to survive to term when transferred into Thy1.2-AMHTg/0 dams. Amh and Amhr2 mRNA was detected in the gravid uterus but at very low levels relative to expression in the ovaries. Progesterone and estradiol levels were not significantly different between WT and Thy1.2-AMHTg/0 dams during pregnancy but luteinizing hormone (LH) levels were significantly elevated in Thy1.2-AMHTg/0 dams at E9.5 and E13.5 relative to WT dams. Collectively, these experiments suggest that AMH overexpression does not cause fetal resorption through an effect on oocytes or preimplantation embryo development. The Thy1.2-AMHTg/0 fetal resorption phenotype is nearly identical to that of transgenic LH overexpression models, suggesting that neuroendocrine mechanisms may be involved in the cause of the miscarriage.


Assuntos
Aborto Espontâneo , Hormônio Antimülleriano , Aborto Espontâneo/metabolismo , Animais , Hormônio Antimülleriano/genética , Hormônio Antimülleriano/metabolismo , Transferência Embrionária , Feminino , Reabsorção do Feto/metabolismo , Humanos , Camundongos , Oócitos/metabolismo , Gravidez
15.
J Neuroinflammation ; 8: 172, 2011 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-22152221

RESUMO

BACKGROUND: Metallothionein-I and -II (MT-I/II) is produced by reactive astrocytes in the injured brain and has been shown to have neuroprotective effects. The neuroprotective effects of MT-I/II can be replicated in vitro which suggests that MT-I/II may act directly on injured neurons. However, MT-I/II is also known to modulate the immune system and inflammatory processes mediated by the immune system can exacerbate brain injury. The present study tests the hypothesis that MT-I/II may have an indirect neuroprotective action via modulation of the immune system. METHODS: Wild type and MT-I/II(-/-) mice were administered cryolesion brain injury and the progression of brain injury was compared by immunohistochemistry and quantitative reverse-transcriptase PCR. The levels of circulating leukocytes in the two strains were compared by flow cytometry and plasma cytokines were assayed by immunoassay. RESULTS: Comparison of MT-I/II(-/-) mice with wild type controls following cryolesion brain injury revealed that the MT-I/II(-/-) mice only showed increased rates of neuron death after 7 days post-injury (DPI). This coincided with increases in numbers of T cells in the injury site, increased IL-2 levels in plasma and increased circulating leukocyte numbers in MT-I/II(-/-) mice which were only significant at 7 DPI relative to wild type mice. Examination of mRNA for the marker of alternatively activated macrophages, Ym1, revealed a decreased expression level in circulating monocytes and brain of MT-I/II(-/-) mice that was independent of brain injury. CONCLUSIONS: These results contribute to the evidence that MT-I/II(-/-) mice have altered immune system function and provide a new hypothesis that this alteration is partly responsible for the differences observed in MT-I/II(-/-) mice after brain injury relative to wild type mice.


Assuntos
Lesões Encefálicas/imunologia , Lesões Encefálicas/patologia , Leucócitos/imunologia , Macrófagos/imunologia , Metalotioneína/imunologia , Animais , Astrócitos/metabolismo , Lesões Encefálicas/sangue , Quimiocinas/imunologia , Citocinas/imunologia , Contagem de Leucócitos , Macrófagos/citologia , Masculino , Metalotioneína/genética , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/metabolismo
17.
Endocrine ; 71(1): 225-232, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32918141

RESUMO

PURPOSE: Anti-Müllerian hormone (AMH) levels fall during pregnancy but the amount of time required for AMH levels to return to normal has not been accurately determined. We have previously shown that AMH levels have yet to return to normal in some women at 3-months postpartum. In this study, AMH levels were examined at 1- and 5-months postpartum to examine whether AMH levels had returned to normal within this interval. METHODS: Longitudinal study involving 38 pregnant women, with serum samples taken in the first trimester, third trimester, 1-month postpartum, 5-months postpartum and 4-6 years postpartum. Participants were recruited from a tertiary maternity clinic (single centre). All women in the study were intending to breastfeed exclusively for at least 5 months, with all 38 participants achieving this at 1-month postpartum and 36/38 after 5 months. RESULTS: Serum AMH concentrations had not returned to expected non-pregnant levels by 1-month postpartum. At 5-months postpartum, mean AMH concentrations were similar to expected non-pregnant levels but the rank order of AMH concentrations was still dissimilar to the non-pregnant state. CONCLUSIONS: The regulation of AMH secretion appears to be distinctly different in non-pregnant, pregnant and postpartum populations. This may affect the conclusions that can be drawn from AMH measurements in women during pregnancy and the postpartum period.


Assuntos
Hormônio Antimülleriano , Período Pós-Parto , Feminino , Humanos , Estudos Longitudinais , Gravidez , Primeiro Trimestre da Gravidez , Terceiro Trimestre da Gravidez
18.
Fertil Steril ; 116(2): 546-557, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33775397

RESUMO

OBJECTIVE: To characterize and evaluate the variation in serum concentrations of oocyte-secreted growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15) throughout the menstrual cycle in women from young to advanced reproductive ages. DESIGN: Cross-sectional, observational, and exploratory study. SETTING: Multicenter university-based clinical practices and laboratories. PATIENT(S): Serum was collected every 1-3 days throughout the menstrual cycle from 3 cohorts of healthy, ovulatory women: menses to late luteal phase (21-29 years of age; n = 16; University of Otago) and across one interovulatory interval (18-35 years of age; n = 10; and 45-50 years of age; n = 15; University of Saskatchewan). INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): To detect the changes in serum GDF9 and BMP15 across the cycle, mean concentration and variance were statistically modeled using a generalized additive model of location, shape and scale (GAMLSS). Follicle-stimulating hormone, luteinizing hormone, estradiol, progesterone, and anti-Müllerian hormone were also assessed. RESULT(S): GDF9 and BMP15 were detectable in 54% and 73% of women and varied 236-fold and 52-fold between women, respectively. Across the menstrual cycle, there were minimal changes in GDF9 or BMP15 within a woman for all cohorts, with no significant differences detected in the modeled mean concentrations. However, modeled variances were highest in the luteal phases of all women for BMP15 immediately after ovulation, regardless of age. CONCLUSION(S): Serial changes in GDF9 or BMP15 concentrations across the cycle were not statistically detected and are likewise similar across the reproductive lifespan. Further research is required to fully elucidate the utility of these oocyte biomarkers at diagnosing fertility potential and/or disease.


Assuntos
Proteína Morfogenética Óssea 15/sangue , Fator 9 de Diferenciação de Crescimento/sangue , Ciclo Menstrual/sangue , Adulto , Biomarcadores/sangue , Estudos Transversais , Feminino , Humanos , Pessoa de Meia-Idade
19.
J Endocrinol ; 237(2): 153-163, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29540452

RESUMO

Anti-Müllerian hormone (AMH) is an ovarian regulator that affects folliculogenesis. AMH inhibits the developmental activation of the dormant primordial follicles and the oocyte within. In more mature follicles, AMH reduces granulosa cell sensitivity to follicle-stimulating hormone (FSH). We examined the effects of AMH overexpression on the stages of ovarian folliculogenesis, and the development of embryos, with a transgenic mouse that overexpresses human AMH in central nervous system neurons under the control of the mouse Thy1.2 promoter (Thy1.2-AMHTg mice). These mice are severely sub-fertile, despite relatively normal ovulation rates. The embryos of Thy1.2-AMHTg females exhibited delayed preimplantation development and extensive mid-gestation fetal resorption. Young Thy1.2-AMHTg mouse ovaries exhibited only a slight reduction in the rate of primordial follicle activation but large declines in the number of developing follicles surviving past the primary stage. It was expected that Thy1.2-AMHTg mice would retain more primordial follicles as they aged, but at 5 months, their number was significantly reduced relative to wild-type females. These data indicate that moderate elevations in AMH levels can severely restrict reproductive output and the number of developing follicles in the ovary. This evidence suggests that early antral follicles are a target for AMH signaling, which may regulate early follicle survival.


Assuntos
Hormônio Antimülleriano/genética , Folículo Ovariano/fisiologia , Animais , Hormônio Antimülleriano/fisiologia , Sobrevivência Celular/genética , Células Cultivadas , Técnicas de Cultura Embrionária , Perda do Embrião/genética , Perda do Embrião/patologia , Embrião de Mamíferos , Feminino , Humanos , Tamanho da Ninhada de Vivíparos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Ovulação/genética , Ovulação/fisiologia , Gravidez
20.
J Endocrinol ; 233(1): R1-R13, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28130407

RESUMO

The mammalian ovary has a finite supply of oocytes, which are contained within primordial follicles where they are arrested in a dormant state. The number of primordial follicles in the ovary at puberty is highly variable between females of the same species. Females that enter puberty with a small ovarian reserve are at risk of a shorter reproductive lifespan, as their ovarian reserve is expected to be depleted faster. One of the roles of anti-Müllerian hormone (AMH) is to inhibit primordial follicle activation, which slows the rate at which the ovarian reserve is depleted. A simple interpretation is that the function of AMH is to conserve ovarian reserve. However, the females with the lowest ovarian reserve and the greatest risk of early reserve depletion have the lowest levels of AMH. In contrast, AMH apparently strongly inhibits primordial follicle activation in females with ample ovarian reserve, for reasons that remain unexplained. The rate of primordial follicle activation determines the size of the developing follicle pool, which in turn, determines how many oocytes are available to be selected for ovulation. This review discusses the evidence that AMH regulates the size of the developing follicle pool by altering the rate of primordial follicle activation in a context-dependent manner. The expression patterns of AMH across life are also consistent with changing requirements for primordial follicle activation in the ageing ovary. A potential role of AMH in the fertility of ageing females is proposed herein.


Assuntos
Hormônio Antimülleriano/metabolismo , Reserva Ovariana/fisiologia , Ovário/metabolismo , Ovulação/fisiologia , Animais , Feminino , Humanos , Folículo Ovariano/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA