Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Hum Reprod ; 36(5): 1326-1338, 2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33681988

RESUMEN

STUDY QUESTION: Can group culture with stage-specific anti-Müllerian hormone (AMH) modulation support human follicular development and oocyte maturation in vitro? SUMMARY ANSWER: In the presence of FSH, AMH supplementation at the secondary-to-early antral stage followed by AMH depletion promotes the coordinated growth and function of human follicles during group culture, thereby yielding mature oocytes. WHAT IS KNOWN ALREADY: Stage-specific AMH modulation promotes in-vitro development of nonhuman primate follicles. The group culture method supports nonhuman primate follicle growth from the primary to antral stage, producing developmentally competent oocytes. STUDY DESIGN, SIZE, DURATION: Ovarian tissue samples were collected from 19 patients of reproductive age (22-47 years old having menstrual cycles) who underwent oophorectomy or hysterectomy for clinical purposes. Tissue pieces were cultured in a matrix-free system for 3 weeks followed by isolation of follicles for the subsequent 6-week individual or group culture. PARTICIPANTS/MATERIALS, SETTING, METHODS: Pieces of ovarian cortical tissue were cultured to support primordial follicle activation and early-stage follicle growth. Secondary follicles isolated from cultured tissue were then randomly assigned to two groups for individual culture: control and AMH modulation, i.e., recombinant human AMH protein supplementation during the secondary-to-early antral stage followed by the addition of neutralizing anti-human AMH antibody. Secondary follicles were also cultured in groups with the same AMH modulation. Follicle survival, growth, steroid hormone and paracrine factor production, steroidogenic protein expression, as well as oocyte maturation and morphology were assessed. MAIN RESULTS AND THE ROLE OF CHANCE: Follicles grew to the secondary stage during 3 weeks of ovarian tissue culture. In-vitro-developed follicles expressed AMH and levels of secreted AMH increased (P < 0.05) in the culture media over time. Secondary follicles isolated from cultured ovarian tissue survived and grew to the antral stage during 6 weeks of individual follicle culture. In-vitro-developed antral follicles produced granulosa and theca cell-derived steroid hormones and paracrine factors, which were detectable in the culture media. Germinal vesicle oocytes obtained from cultured follicles exhibited a perinucleolar chromatin rim configuration. AMH modulation did not alter follicle survival or oocyte maturation relative to those of the control follicles. However, follicle diameters, as well as steroid hormone and paracrine factor production, increased (P < 0.05) in the AMH-modulation group compared with the control group. Secondary follicles isolated from cultured ovarian tissue formed aggregates and grew to the antral stage during 6 weeks of group culture. In-vitro-developed antral follicles expressed steroidogenic enzymes and secreted steroid hormones were detectable in the culture media. Oocytes obtained from cultured follicle aggregates with AMH-modulation progressed to the metaphase II stage after IVM, containing a normal-sized first polar body and meiotic spindle. Oocytes exhibited a typical ultrastructure. LIMITATIONS, REASONS FOR CAUTION: Follicles were obtained from fresh ovarian tissue of adult patients. Oocyte maturation rates were relatively low and oocytes were assessed by morphological evaluation. Owing to the lack of a control group, the beneficial effects of AMH modulation remained undetermined for the group culture in this study. WIDER IMPLICATIONS OF THE FINDINGS: Stage-specific AMH modulation supports human follicular development in the matrix-free group culture, which is consistent with previously reported AMH actions on growing follicles in nonhuman primates. Oocytes generated by in-vitro-developed follicles achieve meiotic maturation with a typical morphology and ultrastructure, which supports in-vitro follicle maturation as a potential approach for fertility preservation in women. STUDY FUNDING/COMPETING INTEREST(S): NICHD R01HD082208 and NIH Office of the Director P51OD011092. The authors have no competing interest to declare. TRIAL REGISTRATION NUMBER: N/A.


Asunto(s)
Hormona Antimülleriana , Folículo Ovárico , Adulto , Femenino , Humanos , Metafase , Oocitos , Ovario
2.
JCI Insight ; 6(4)2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33616080

RESUMEN

A maternal Western-style diet (WSD) is associated with poor reproductive outcomes, but whether this is from the diet itself or underlying metabolic dysfunction is unknown. Here, we performed a longitudinal study using regularly cycling female rhesus macaques (n = 10) that underwent 2 consecutive in vitro fertilization (IVF) cycles, one while consuming a low-fat diet and another 6-8 months after consuming a high-fat WSD. Metabolic data were collected from the females prior to each IVF cycle. Follicular fluid (FF) and oocytes were assessed for cytokine/steroid levels and IVF potential, respectively. Although transition to a WSD led to weight gain and increased body fat, no difference in insulin levels was observed. A significant decrease in IL-1RA concentration and the ratio of cortisol/cortisone was detected in FF after WSD intake. Despite an increased probability of isolating mature oocytes, a 44% reduction in blastocyst number was observed with WSD consumption, and time-lapse imaging revealed delayed mitotic timing and multipolar divisions. RNA sequencing of blastocysts demonstrated dysregulation of genes involved in RNA binding, protein channel activity, mitochondrial function and pluripotency versus cell differentiation after WSD consumption. Thus, short-term WSD consumption promotes a proinflammatory intrafollicular microenvironment that is associated with impaired preimplantation development in the absence of large-scale metabolic changes.


Asunto(s)
Dieta Occidental/efectos adversos , Fertilidad , Reproducción , Tejido Adiposo , Animales , Dieta Alta en Grasa , Desarrollo Embrionario , Femenino , Fertilidad/genética , Líquido Folicular/fisiología , Expresión Génica , Estudios Longitudinales , Macaca mulatta , Modelos Animales , Obesidad , Oocitos/fisiología , Reproducción/genética , Aumento de Peso
3.
Biol Reprod ; 104(3): 602-610, 2021 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-33348377

RESUMEN

Insulin-like growth factors (IGFs) are known for their involvement in endocrine and paracrine regulation of ovarian function. Although IGF2 is the predominant circulating and intraovarian form of IGFs in primate species, the stage-specific follicular expression, action, and regulation of IGF2 are not well defined. Therefore, experiments were conducted to investigate the follicular IGF production in response to steroid hormone regulation and the direct IGF actions on follicular development and function in vitro. Preantral follicles were isolated from rhesus macaque ovaries and cultured to the antral stage in media supplemented with follicle-stimulating hormone and insulin. Follicles were randomly assigned to treatment groups: (a) control, (b) trilostane (a steroid synthesis inhibitor), (c) trilostane + estradiol, (d) trilostane + progesterone, and (e) trilostane + dihydrotestosterone. Media was analyzed for IGF concentrations, which were correlated to follicle growth. Follicles produced IGF2, but not IGF1, at the antral stage. Steroid depletion decreased, whereas steroid replacement increased, IGF2 production by antral follicles. Media IGF2 levels correlated positively with antral follicle diameters. Macaque preantral follicles and granulosa cells were subsequently cultured without (control) and with recombinant human IGF2 supplementation. Follicle survival, growth, and paracrine factor production, as well as granulosa cell proliferation and gonadotropin receptor gene expression, were assessed. IGF2 addition increased follicle survival rates, diameters and inhibin B production, as well as granulosa cell proliferation. These data demonstrate that IGF2 produced by antral follicles, in response to steroid hormone regulation, could act as a paracrine factor that positively impacts preantral follicle development and function in primates.


Asunto(s)
Factor II del Crecimiento Similar a la Insulina/metabolismo , Factor II del Crecimiento Similar a la Insulina/farmacología , Folículo Ovárico/crecimiento & desarrollo , Folículo Ovárico/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Estradiol/análogos & derivados , Estradiol/farmacología , Femenino , Hormona Folículo Estimulante/farmacología , Factor II del Crecimiento Similar a la Insulina/administración & dosificación , Macaca mulatta , Progesterona/farmacología , Técnicas de Cultivo de Tejidos
4.
J Assist Reprod Genet ; 36(2): 229-239, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30430314

RESUMEN

PURPOSE: To determine the effects of PGL1001, a somatostatin receptor isoform-2 (SSTR-2) antagonist, on ovarian follicle development, oocyte fertilization, and subsequent embryo developmental potential in the rhesus macaque. METHODS: Cycling female rhesus macaques (N = 8) received vehicle through one menstrual (control) cycle, followed by daily injections of PGL1001, a SSTR-2 antagonist, for three menstrual (treatment) cycles. Main endpoints include overall animal health and ovarian hormones (e.g., estradiol [E2], progesterone [P4], and anti-Müllerian hormone [AMH]), ovarian circumference, numbers of oocytes and their maturation status following controlled ovarian stimulation (COS), as well as oocyte fertilization and subsequent blastocyst rates that were assessed in control and PGL1001 treatment cycles. Circulating PGL1001 levels were assessed at baseline as well as 6, 60, and 90 days during treatment. RESULTS: PGL1001 treatment did not impact overall animal health, menstrual cycle length, or circulating levels of ovarian hormones (E2, P4, and AMH) in comparison to vehicle treatment during natural cycles. PGL1001 treatment increased (p Ë‚ 0.05) ovarian circumference and the day 8 to day 1 ratio of AMH levels (p Ë‚ 0.05) during a COS protocol, as well as oocyte fertilization rates compared to the vehicle treatment interval. Blastocyst development rates were not significantly different between vehicle and PGL1001 treatment groups. CONCLUSION: Prolonged treatment with PGL1001 appears to be safe and does not affect rhesus macaque general health, menstrual cycle length, or ovarian hormone production. Interestingly, PGL1001 treatment increased the fertilization rate of rhesus macaque oocytes collected following ovarian stimulation.


Asunto(s)
Desarrollo Embrionario/efectos de los fármacos , Oocitos/efectos de los fármacos , Folículo Ovárico/crecimiento & desarrollo , Receptores de Somatostatina/antagonistas & inhibidores , Animales , Hormona Antimülleriana/administración & dosificación , Blastocisto/efectos de los fármacos , Femenino , Fertilización/efectos de los fármacos , Humanos , Macaca mulatta , Folículo Ovárico/efectos de los fármacos , Inducción de la Ovulación/métodos , Progesterona/administración & dosificación , Somatostatina/metabolismo
5.
Biol Reprod ; 98(2): 197-207, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29293939

RESUMEN

Anti-Müllerian hormone (AMH) plays a key role during ovarian follicular development, with local actions associated with a dynamic secretion profile by growing follicles. While results for AMH effects on antral follicle growth and function are consistent among studies in various species, any effects on preantral follicle development remain controversial. Therefore, experiments were conducted to investigate the direct actions and role of AMH during follicle development at the preantral stage. Macaque-specific short-hairpin RNAs (shRNAs) targeting AMH mRNA were incorporated into adenoviral vectors to decrease AMH gene expression in rhesus macaque follicles. Secondary follicles were isolated from adult macaque ovaries and cultured individually in the ultra-low-attachment dish containing defined medium supplemented with follicle-stimulating hormone and insulin for 5 weeks. Follicles were randomly assigned to treatment groups: (a) control, (b) nontargeting control shRNA-vector, (c) AMH shRNA-vector, (d) AMH shRNA-vector + recombinant human AMH, and (e) recombinant human AMH. Follicle survival and growth were assessed. Culture media were analyzed for steroid hormone and paracrine factor concentrations. For in vivo study, the nontargeting control shRNA-vector and AMH shRNA-vector were injected into macaque ovaries. Ovaries were collected 9 days postinjection for morphology and immunohistochemistry assessment. Decreased AMH expression reduced preantral follicle survival and growth in nonhuman primates. Supplemental AMH treatment in the culture media promoted preantral follicle growth to the small antral stage in vitro with increased steroid hormone and paracrine factor production, as well as oocyte maturation. These data demonstrate that AMH is a critical follicular paracrine/autocrine factor positively impacting preantral follicle survival and growth in primates.


Asunto(s)
Hormona Antimülleriana/metabolismo , Folículo Ovárico/crecimiento & desarrollo , Animales , Hormona Antimülleriana/genética , Femenino , Técnicas de Maduración In Vitro de los Oocitos/métodos , Macaca mulatta , Folículo Ovárico/metabolismo
6.
J Assist Reprod Genet ; 35(1): 71-79, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28936565

RESUMEN

PURPOSE: Neutral red (NR) may assist identification of preantral follicles in pieces of cortical tissue prior to cryopreservation in cancer patients requesting fertility preservation. This study is the first to analyze this effect by follicle growth rate after long-term culture in primates. METHODS: Ovarian cortex was obtained from adult rhesus macaques, was cut into fragments, and was incubated with NR. Secondary follicles were readily visualized following NR staining and then were encapsulated into alginate beads and cultured individually for 4 weeks in αMEM media supplemented with 10 ng/ml FSH at 5% O2. RESULTS: The survival rates of secondary follicles during culture were similar between those derived from control tissue (71 ± 13%) and those treated with NR (68 ± 9%). The proportion of surviving follicles that formed an antrum were also similar in both groups (70 ± 17% control; 48 ± 24% NR-treated). Follicle diameters were not different between control follicles (184 ± 5µm) and those stained with NR (181 ± 7 µm) on the day of isolation. The percentages of surviving follicles within three cohorts based on their diameters at week 4 of culture were similar between the control group and NR-stained tissue group, fast-grow follicles (24 ± 6% vs. 13 ± 10%), slow-grow follicles (66 ± 5% vs. 60 ± 9%), or no-grow (10 ± 9% vs. 27 ± 6%), respectively. There were no differences in follicle diameters between groups during the culture period. Pre-exposure of secondary follicles to NR diminished their capacity to produce both estradiol and androstenedione by week 4 of culture, when follicles are exhibiting an antrum. Inhibitory effects of NR on steroid production by slow-grow follicles was less pronounced. CONCLUSIONS: NR does not affect secondary follicle survival, growth, and antrum formation during long-term culture, but steroid hormone production by fast-grow follicles is compromised. NR can be used as a non-invasive tool for in situ identification of viable secondary follicles in ovarian cortex before tissue cryopreservation without affecting follicle survival and growth in vitro. Whether maturation or developmental competence of oocytes derived from antral follicles in 3D culture that were previously isolated from NR-stained tissue is normal or compromised remains to be determined. Likewise, the functional consequences of pre-exposure to NR prior to ovarian cortical tissue cryopreservation and transplantation are unknown.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Macaca mulatta , Rojo Neutro/farmacología , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/crecimiento & desarrollo , Animales , Procesos de Crecimiento Celular/efectos de los fármacos , Supervivencia Celular , Femenino , Folículo Ovárico/citología , Andamios del Tejido
7.
Biol Reprod ; 96(5): 1052-1059, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28444134

RESUMEN

The phosphoinositide 3-kinase/AKT (protein kinase B) signaling pathway negatively regulates follicle activation via the forkhead box O (FOXO) transcription factor in rodents. FOXO3 knockout mice exhibit global activation of primordial follicles leading to early depletion of ovarian follicles and subsequent infertility. Whether a similar mechanism for follicle activation exists in the primate ovary is unclear. In the current study, protein localization of FOXO1, 3, and 4 as well as their upstream regulator, AKT/p-AKT, was examined in rhesus macaque ovaries of three developmental stages: fetal, prepubertal, and adult. FOXO1 protein is expressed in granulosa cells of fetal, prepubertal, and adult ovaries. FOXO3 is distributed sparsely in the mitotically active germ cells, but its expression decreases following follicle formation in the macaque fetal ovary. In addition, FOXO3 is seldom with interanimal variation in the prepubertal ovary and is absent in the adult ovary. FOXO4 is nondetectable in fetal ovaries, although it is expressed in some theca cells of antral follicles and some stromal cells in prepubertal and adult ovaries. Our results suggest that the regulation and/or function of FOXO3 in the primate primordial follicle may differ than that of the rodent. Nevertheless, AKT/p-AKT is expressed in macaque primordial oocytes, suggesting that similar upstream events but different downstream effects may regulate primordial follicle activation in nonhuman primates compared to rodents. Elucidation of the mechanism responsible for follicle activation in primates will be crucial for understanding primary ovarian insufficiency, improving female fertility, and applying techniques for in vitro maturation of follicles for fertility preservation in cancer survivors.


Asunto(s)
Feto/metabolismo , Factores de Transcripción Forkhead/genética , Ovario/metabolismo , Animales , Femenino , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O3/genética , Células de la Granulosa/metabolismo , Inmunohistoquímica , Macaca mulatta , Proteína Oncogénica v-akt/metabolismo , Ovario/crecimiento & desarrollo , Maduración Sexual , Células del Estroma/metabolismo
8.
J Assist Reprod Genet ; 34(2): 291-300, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27900615

RESUMEN

Products that are manufactured for use in a clinical trial, with the intent of gaining US Food and Drug Administration (FDA) approval for clinical use, must be produced under an FDA approved investigational new drug (IND) application. We describe work done toward generating reliable methodology and materials for preserving ovarian cortical tissue through a vitrification kit and reviving this tissue through a warming and recovery kit. We have described the critical steps, procedures, and environments for manufacturing products with the intent of submitting an IND. The main objective was to establish an easy-to-use kit that would ensure standardized procedures for quality tissue preservation and recovery across the 117 Oncofertility Consortium sites around the globe. These kits were developed by breaking down the components and steps of a research protocol and recombining them in a way that considers component stability and use in a clinical setting. The kits were manufactured utilizing current good manufacturing practice (cGMP) requirements and environment, along with current good laboratory practices (cGLP) techniques. Components of the kit were tested for sterility and endotoxicity, and morphological endpoint release criteria were established. We worked with the intended down-stream users of these kits for development of the kit instructions. Our intention is to test these initial kits, developed and manufactured here, for submission of an IND and to begin clinical testing for preserving the ovarian tissue that may be used for future restoration of fertility and/or hormone function in women who have gonadal dysgenesis from gonadotoxic treatment regimens or disease.


Asunto(s)
Investigación Biomédica , Juego de Reactivos para Diagnóstico , Vitrificación , Femenino , Humanos , Folículo Ovárico/fisiología , Estados Unidos , United States Food and Drug Administration
9.
Fertil Steril ; 105(4): 1023-34, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26718060

RESUMEN

OBJECTIVE: To examine the small antral follicle (SAF) cohort in ovaries of adult rhesus monkeys after consumption of a Western-style diet (WSD), with or without chronically elevated androgen levels since before puberty. DESIGN: Cholesterol or T (n = 6 per group) implants were placed SC in female rhesus macaques beginning at 1 year of age (prepubertal), with addition of a WSD (high fat/fructose) at 5.5 years (menarche approximately 2.6 years). Ovaries were collected at 7 years of age. One ovary per female was embedded in paraffin for morphologic and immunohistochemical analyses. The SAFs (<2.5 mm) were dissected from the other ovary obtained at or near menses in a subgroup of females (n = 3 per group) and processed for microarray analyses of the SAF transcriptome. Ovaries of adult monkeys consuming a standard macaque diet (low in fats and sugars) were obtained at similar stages of the menstrual cycle and used as controls for all analyses. SETTING: Primate research center. ANIMAL(S): Adult, female rhesus monkeys (Macaca mulatta). INTERVENTION(S): None. MAIN OUTCOME MEASURES: Histologic analyses, SAF counts and morphology, protein localization and abundance in SAFs, transcriptome in SAFs (messenger RNAs [mRNAs]). RESULT(S): Compared with controls, consumption of a WSD, with and without T treatment, increased the numbers of SAFs per ovary, owing to the presence of more atretic follicles. Numbers of granulosa cells expressing cellular proliferation markers (pRb and pH3) was greater in healthy SAFs, whereas numbers of cells expressing the cell cycle inhibitor (p21) was higher in atretic SAFs. Intense CYP17A1 staining was observed in the theca cells of SAFs from WSD with or without T groups, compared with controls. Microarray analyses of the transcriptome in SAFs isolated from WSD and WSD plus T-treated females and controls consuming a standard diet identified 1,944 genes whose mRNA levels changed twofold or more among the three groups. Further analyses identified several gene pathways altered by WSD and/or WSD plus T associated with steroid, carbohydrate, and lipid metabolism, plus ovarian processes. Alterations in levels of several SAF mRNAs are similar to those observed in follicular cells from women with polycystic ovary syndrome. CONCLUSION(S): These data indicate that consumption of a WSD high in fats and sugars in the presence and absence of chronically elevated T alters the structure and function of SAFs within primate ovaries.


Asunto(s)
Andrógenos/administración & dosificación , Dieta Occidental/efectos adversos , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/patología , Maduración Sexual/fisiología , Factores de Edad , Andrógenos/metabolismo , Animales , Recuento de Células , Femenino , Humanos , Macaca mulatta , Folículo Ovárico/metabolismo , Ovario/efectos de los fármacos , Ovario/metabolismo , Ovario/patología , Maduración Sexual/efectos de los fármacos , Resultado del Tratamiento
10.
J Ovarian Res ; 8: 8, 2015 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-25824473

RESUMEN

BACKGROUND: ADRB-2 was implicated in rodent ovarian functions, including initial follicular growth. In contrast, ADRB-2 expression and function in nonhuman primate and human ovary were not fully known but innervation and significant levels of norepinephrine (NE), which is a ligand at the ADRB-2, were reported in the ovary. METHODS: We studied expression of ADRB-2 in human and rhesus monkey ovary (RT-PCR, immunohistochemistry; laser micro dissection) and measured levels of norepinephrine (NE; ELISA) in monkey follicular fluid (FF). 3D cultures of monkey follicles (4 animals) were exposed to NE or the ADRB-2 agonist isoproterenol (ISO), and follicular development (size) was monitored. Upon termination expression of ADRB-2, FSH receptor and aromatase genes were examined. RESULTS: Immunohistochemistry and RT-PCR of either human follicular granulosa cells (GCs) obtained by laser micro dissection or isolated monkey follicles revealed ADRB-2 in GCs of primordial, primary, secondary and tertiary follicles. Staining of GCs in primordial and primary follicles was intense. In large preantral and antral follicles the staining was heterogeneous, with positive and negative GCs present but GCs lining the antrum of large follicles were generally strongly immunopositive. Theca, interstitial, and ovarian surface epithelial cells were also positive. NE was detected in FF of preovulatory antral monkey follicles (0.37 + 0.05 ng/ml; n = 7; ELISA) but not in serum. We examined preantral follicles ranging from 152 to 366 µm in diameter in a 3D culture in media supplemented with follicle stimulating hormone (FSH). Under these conditions, neither NE, nor ISO, influenced growth rate in a period lasting up to one month. Upon termination of the cultures, all surviving follicles expressed aromatase and FSH receptors, but only about half of them also co-expressed ADRB-2. The ADRB-2 expression was not correlated with the treatment but was positively correlated with the follicular size at the beginning and at the end of the culture period. Hence, expression of ADRB-2 was found in the largest and fastest-in vitro growing follicles. CONCLUSIONS: The results imply ADRB-2-mediated actions in the development of primate follicles. Drugs interfering with ADRB-2 are used to treat medical conditions and may have unexplored effects in the human ovary.


Asunto(s)
Folículo Ovárico/metabolismo , Receptores Adrenérgicos beta 2/biosíntesis , Animales , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunohistoquímica , Captura por Microdisección con Láser , Macaca mulatta , Norepinefrina/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Biol Reprod ; 92(5): 133, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25810474

RESUMEN

Tamoxifen (TAM) is a selective estrogen receptor modulator with tissue-specific effects on estrogen signaling used predominantly for treatment and chemoprevention of breast cancers. Recent studies have shown that TAM prevents infertility and decreases follicular loss from common cancer chemotherapy and radiation therapy in preclinical models. Here we review current and novel uses of selective estrogen receptor modulator s and advantages and challenges for translation of TAM for human fertility preservation.


Asunto(s)
Ovario/efectos de los fármacos , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Tamoxifeno/farmacología , Femenino , Fertilidad/efectos de los fármacos , Humanos
12.
Hum Reprod ; 27(8): 2430-7, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22593432

RESUMEN

BACKGROUND: In non-primates, the epidermal growth factor (EGF) and EGF-related ligands such as amphiregulin (AREG) serve as critical intermediates between the theca/mural cells and the cumulus-oocyte-complex (COC) following the mid-cycle LH surge. Studies were designed in primates (1) to analyze AREG levels in follicular fluid (follicular fluid) obtained from pre-ovulatory follicles, as well as (2) to assess dose-dependent effects of AREG on oocytes from small antral follicles (SAFs) during culture, including meiotic and cytoplasmic maturation. METHODS: Controlled ovulation protocols were performed on rhesus monkeys (n=12) to determine AREG content within the single, naturally selected dominant follicle after an ovulatory stimulus. Using healthy COCs (n=271) obtained from SAFs during spontaneous cycles (n=27), in vitro maturation (IVM) was performed in the absence or presence of physiological concentrations of AREG (10 or 100 ng/ml) with or without gonadotrophins (FSH, 75 mIU/ml; LH, 75 mIU/ml). At the end of the culture period, oocyte meiotic maturation was evaluated and ICSI was performed (n=111), from which fertilization and early embryo development was followed in vitro. RESULTS: AREG levels in follicular fluid from pre-ovulatory follicles increased (P<0.05) following an ovulatory bolus of hCG at 12, 24 and 36 h post-treatment. At 12 h post-hCG, AREG levels in follicular fluid ranged from 4.8 to 121.4 ng/ml. Rhesus macaque COCs incubated with 10 ng/ml AREG in the presence of gonadotrophins displayed an increased percentage of oocytes that progressed to the metaphase II (MII) stage of meiosis (82 versus 56%, P<0.05) and a decreased percentage of metaphase I (MI) oocytes (2 versus 23%, P<0.05) relative to controls, respectively. The percentage of either MI or MII oocytes at the end of the culture period was not different between oocytes cultured with 100 ng/ml AREG or in media alone. Fertilization and first cleavage rates obtained by ICSI of all IVM MII oocytes were 93 and 98%, respectively, and did not vary among treatment groups. Of the MII oocytes that fertilized (n=103), 37 were randomly selected and maintained in culture to assess developmental potential. A total of 13 early blastocysts were obtained, with four embryos developing to expanded blastocysts. CONCLUSIONS: These data indicate that AREG levels increase in rhesus macaque pre-ovulatory follicles after an ovulatory stimulus, and a specific concentration of AREG (10 ng/ml) enhances rhesus macaque oocyte nuclear maturation but not cytoplasmic maturation from SAFs obtained during the natural menstrual cycle. However, owing to the small number of samples in some treatment groups, further studies are now required.


Asunto(s)
Glicoproteínas/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Oocitos/citología , Anfirregulina , Animales , Blastocisto/citología , Células Cultivadas , Citoplasma/metabolismo , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Líquido Folicular/metabolismo , Ligandos , Hormona Luteinizante/metabolismo , Macaca mulatta , Meiosis , Oogénesis , Folículo Ovárico/citología , Ovario/fisiología , Factores de Tiempo
13.
Cryobiology ; 65(1): 1-11, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22569078

RESUMEN

Ovarian tissue cryopreservation is the only proven option for fertility preservation in female cancer patients who are prepubertal or require immediate treatment. However it remains unclear which cryopreservation protocol is best in cases where the tissue may contain cancerous cells, as these should be matured in vitro rather than autografted. This study evaluated different cryoprotectant exposure times and whether the addition of synthetic polymers (Supercool X-1000, Z-1000 and polyvinylpyrrolidone [PVP K-12]) to the vitrification solution is beneficial to tissue morphology, cellular proliferation and subsequent in vitro function of secondary follicles. Pieces of macaque (n=4) ovarian cortex were exposed to vitrification solution containing glycerol (25%, v/v) and ethylene glycol (25%, v/v) for 3 or 8 min, without (V3, V8) or with (VP3, VP8) polymers (0.2% [v/v] X-1000, 0.4% Z-1000 and 0.2% PVP). Fresh and vitrified tissues were fixed for histology and phosphohistone H3 (PPH3) analysis, or used for secondary follicle isolation followed by encapsulated 3D culture. Five-week follicle survival and growth, as well as steroid hormones (estradiol [E(2)], progesterone, androstenedione) were measured weekly. Morphology of the stroma and preantral follicles as well as PPH3 expression, was preserved in all vitrified tissues. Vitrification with polymers and shorter incubation time (VP3) increased in vitro follicle survival and E(2) production compared to other vitrified groups. Thus, a short exposure of macaque ovarian tissue to a vitrification solution containing synthetic polymers preserves morphology and improves in vitro function of secondary follicles.


Asunto(s)
Criopreservación/métodos , Crioprotectores/farmacología , Oocitos/citología , Folículo Ovárico/crecimiento & desarrollo , Ovario/crecimiento & desarrollo , Polímeros/farmacología , Animales , Femenino , Preservación de la Fertilidad/métodos , Hormonas Esteroides Gonadales/metabolismo , Histonas/metabolismo , Macaca mulatta/metabolismo , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Folículo Ovárico/efectos de los fármacos , Ovario/efectos de los fármacos , Factores de Tiempo , Vitrificación
14.
Hum Reprod ; 26(9): 2461-72, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21705370

RESUMEN

BACKGROUND: Ovarian tissue cryopreservation is the only option for preserving fertility in prepubertal girls and cancer patients requiring immediate treatment. Following ovarian tissue cryopreservation, fertility can be restored after tissue transplant or in vitro follicle maturation. METHODS: Macaque (n= 4) ovarian cortex was cryopreserved using slow-rate freezing (slow freezing) or vitrification. Tissues were fixed for histology or phosphohistone H3 (PPH3) analysis, cultured with bromodeoxyuridine (BrdU) or used for three-dimensional secondary follicle culture. Follicular diameter and steroid hormones were measured weekly. RESULTS: Slow freezing induced frequent cryo-injuries while vitrification consistently maintained morphology of the stroma and secondary follicles. PPH3 was similar in fresh and vitrified, but sparse in slow-frozen tissues. BrdU uptake appeared diminished following both methods compared with that in fresh follicles. In vitro follicle survival and growth were greater in fresh than in cryopreserved follicles. Antrum formation appeared similar after vitrification compared with the fresh, but was reduced following slow freezing. Steroid production was delayed or diminished following both methods compared with fresh samples. CONCLUSIONS: Secondary follicle morphology was improved after vitrification relative to slow freezing. Following vitrification, stroma was consistently more compact with intact cells typical to that of fresh tissue. BrdU uptake demonstrated follicle viability post-thaw/warming. For the first time, although not to the extent of fresh follicles, macaque follicles from cryopreserved tissue can survive, grow, form an antrum and produce steroid hormones, indicating some functional preservation. The combination of successful ovarian tissue cryopreservation with in vitro maturation of follicles will offer a major advancement to the field of fertility preservation.


Asunto(s)
Criopreservación/métodos , Macaca mulatta , Folículo Ovárico/fisiología , Ovario/fisiología , Animales , Femenino , Preservación de la Fertilidad , Técnicas de Cultivo de Tejidos
15.
J Assist Reprod Genet ; 27(11): 591-7, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20711751

RESUMEN

INTRODUCTION: we serendipitously observed a protective effect of tamoxifen against depletion of ovarian follicles by 7,12-dimethylbenzanthracene (DMBA), a chemical carcinogen, during a cancer prevention study. Such ovarian protection is being sought as an alternative approach to fertility preservation in human cancer patients. METHODS: rats received tamoxifen (0, 1 mg or 2.5 mg/kg/d) and DMBA (0, 1, 2 mg/kg/wk) or cyclophosphamide (0, 35, 50 mg/kg/wk). Ovarian follicles were quantified and effects on fertility and litter size were tested. Cultured oocytes were exposed to chemotherapy drug doxorubicin, with or without 4-hydroxytamoxifen (4HT). RESULTS: DMBA and cyclophosphamide decreased the number of primordial and total follicles, and this reduction was prevented by tamoxifen. Cyclophosphamide tended to reduce fertility and lessened neonatal survival. Tamoxifen reversed these defects. Doxorubicin caused oocyte fragmentation which was prevented by 4HT. CONCLUSIONS: tamoxifen decreases follicle loss and improves reproductive function following exposure to ovarian toxicants including chemotherapy drugs in the female rat.


Asunto(s)
9,10-Dimetil-1,2-benzantraceno/toxicidad , Antineoplásicos/toxicidad , Ciclofosfamida/toxicidad , Doxorrubicina/toxicidad , Folículo Ovárico/efectos de los fármacos , Sustancias Protectoras/farmacología , Tamoxifeno/farmacología , Animales , Células Cultivadas , Femenino , Fertilidad/efectos de los fármacos , Oocitos/efectos de los fármacos , Ovario/efectos de los fármacos , Ovario/patología , Ratas , Ratas Sprague-Dawley , Tamoxifeno/análogos & derivados
16.
Reprod Toxicol ; 28(1): 32-7, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19490992

RESUMEN

The aryl hydrocarbon receptor (AHR) mediates the effects of many endocrine disruptors and contributes to the loss of fertility in polluted environments. Female rats exposed chronically to environmentally relevant doses of the AHR agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) across their lifespan experience accelerated reproductive senescence preceded by ovarian endocrine disruption. The purpose of this study was to determine the changes in ovarian gene expression that accompany the loss of ovarian function caused by chronic exposure to TCDD. Beginning in utero, female Sprague-Dawley rats received TCDD (1, 5, 50, or 200 ng/kg-week; n=4 per group) or vehicle weekly throughout their lifespan, and were sacrificed on diestrus just prior to loss of reproductive cyclicity at 11 months of age. Microarray analysis was used to determine differences in ovarian gene expression between control and TCDD-treated (200 ng/kg-week) animals. To confirm microarray results, real-time PCR was used to assess changes in gene expression among treatment groups. TCDD treatment decreased (p<0.05) proestrus serum estradiol concentrations with no effect on serum progesterone. In ovaries from rats treated with 200 ng/kg-week TCDD compared to controls, 19 genes of known function were found to be up-regulated, while 31 ovarian genes were found to be down-regulated >or=1.5-fold (p

Asunto(s)
Disruptores Endocrinos/toxicidad , Contaminantes Ambientales/toxicidad , Ovario/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/agonistas , Reproducción/efectos de los fármacos , Factores de Edad , Animales , Animales Recién Nacidos , Relación Dosis-Respuesta a Droga , Estradiol/sangre , Femenino , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Edad Gestacional , Análisis de Secuencia por Matrices de Oligonucleótidos , Ovario/metabolismo , Embarazo , Progesterona/sangre , Ratas , Ratas Sprague-Dawley , Receptores de Hidrocarburo de Aril/metabolismo , Reproducción/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Cancer Prev Res (Phila) ; 1(7): 546-53, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19139004

RESUMEN

Women at increased risk for breast cancer are at increased risk for ovarian cancer as well, reflecting common risk factors and intertwined etiology of the two diseases. We previously developed a rat model of elevated breast and ovarian cancer risk, allowing evaluation of dual-target cancer prevention strategies. Tamoxifen, a Food and Drug Administration-approved breast cancer chemoprevention drug, has been shown to promote ovarian cysts in premenopausal women; however, the effect of tamoxifen on ovarian cancer risk is still controversial. In the current experiment, Fischer 344 rats (n = 8 per treatment group) received tamoxifen (TAM) or vehicle (control) in factorial combination with combined breast and ovarian carcinogen (17beta-estradiol and 7,12 dimethylbenza[a]anthracene, respectively). Mammary and ovarian morphologies were normal in the control and TAM groups. Carcinogen (CARC) treatment induced mammary dysplasia with elevated cell proliferation and reduced estrogen receptor-alpha expression and promoted preneoplastic changes in the ovary. In the CARC + TAM group, tamoxifen reduced preneoplastic changes and proliferation rate in the mammary gland, but not in the ovary, compared with rats treated with carcinogen alone. Putative stem cell markers (Oct-4 and aldehyde dehydrogenase 1) were also elevated in the mammary tissue by carcinogen and this expansion of the stem cell population was not reversed by tamoxifen. Our study suggests that tamoxifen prevents early progression to mammary cancer but has no effect on ovarian cancer progression in this rat model.


Asunto(s)
Neoplasias Mamarias Experimentales/prevención & control , Neoplasias Ováricas/prevención & control , Lesiones Precancerosas/prevención & control , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Tamoxifeno/farmacología , Animales , Carcinógenos/toxicidad , Modelos Animales de Enfermedad , Femenino , Inmunohistoquímica , Neoplasias Mamarias Experimentales/patología , Neoplasias Ováricas/patología , Lesiones Precancerosas/patología , Ratas , Ratas Endogámicas F344 , Factores de Riesgo
18.
Carcinogenesis ; 28(1): 130-5, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16891317

RESUMEN

Women at increased risk for breast cancer are often also at increased risk for ovarian cancer, reflecting common risk factors and intertwined etiologies for both diseases. Unlike breast cancer prevention, primary ovarian cancer prevention has been impractical due to the low incidence, lack of risk and response biomarkers and difficulties in sampling ovarian tissue. Challenges in the development of ovarian cancer prevention drugs, however, may be circumvented through the development of breast cancer prevention strategies that simultaneously decrease ovarian cancer. In the present study, three commonly used mammary cancer carcinogen models [7,12-dimethylbenz[alpha]anthracene (DMBA), N-methyl-N-nitrosourea (MNU) and estradiol (E2)] were combined with local ovarian DMBA administration to induce progression to mammary and ovarian cancer concurrently in the rat. Animals were treated for 3 or 6 months, and tissue histology as well as proliferation, hormonal and inflammation biomarkers were assessed. Mammary and ovarian morphologies (measured as descriptive histology and dysplasia scores) were normal in vehicle controls. Mammary hyperplasia was observed in DMBA/DMBA (mammary carcinogen/ovarian carcinogen) and MNU/DMBA-treated rats; however, ovarian preneoplastic changes were seldom observed after these treatments. All E2/DMBA-treated rats had mammary hyperplasia, atypia, ductal carcinoma in situ and/or invasive adenocarcinoma, while 50% also developed preneoplastic changes in the ovary (ovarian epithelial and stromal hyperplasia and inclusion cyst formation). In both the mammary gland and ovary, decreased estrogen receptor alpha expression was detected, and in the mammary gland elevated Ki-67 and cyclooxygenase-2 expressions were observed. This combined breast and ovarian cancer rat model (systemic E2 treatment and local ovarian DMBA) may be useful for future dual target breast and ovarian cancer prevention studies.


Asunto(s)
Cocarcinogénesis , Modelos Animales de Enfermedad , Neoplasias Mamarias Experimentales/patología , Neoplasias Ováricas/patología , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Adenocarcinoma/inducido químicamente , Adenocarcinoma/patología , Alquilantes/toxicidad , Animales , Carcinógenos/toxicidad , Carcinoma Intraductal no Infiltrante/inducido químicamente , Carcinoma Intraductal no Infiltrante/patología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Progresión de la Enfermedad , Epitelio/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Estrógenos/toxicidad , Femenino , Hiperplasia/inducido químicamente , Hiperplasia/patología , Técnicas para Inmunoenzimas , Antígeno Ki-67/metabolismo , Neoplasias Mamarias Experimentales/inducido químicamente , Metilnitrosourea/toxicidad , Neoplasias Ováricas/inducido químicamente , Lesiones Precancerosas , Ratas , Ratas Endogámicas F344
19.
Biol Reprod ; 76(2): 198-202, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17050859

RESUMEN

The aryl hydrocarbon receptor (AHR) mediates the effects of many endocrine disruptors and contributes to the loss of fertility in polluted environments. While previous work has focused on mechanisms of short-term endocrine disruption and ovotoxicity in response to AHR ligands, we have shown recently that chronic exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induces premature reproductive senescence in female rats without depletion of ovarian follicular reserves. In the current study, premature reproductive senescence was induced using a range of low-dose exposure to TCDD (0, 1, 5, 50, and 200 ng kg(-1) wk(-1)) beginning in utero and continuing until the transition to reproductive senescence. Doses of 50 and 200 ng TCDD kg(-1) wk(-1) delayed the age at vaginal opening and accelerated the loss of normal reproductive cyclicity with age without depletion of follicular reserves. Serum estradiol concentrations were decreased in a dose-dependent fashion (> or = 5 ng kg(-1) wk(-1)) across the estrous cycle in perisenescent rats still displaying normal cyclic vaginal cytology. Serum FSH, LH, and progesterone profiles were unchanged by TCDD. The loss of reproductive cyclicity following chronic exposure to TCDD was not accompanied by decreased responsiveness to GnRH. Ovarian endocrine disruption is the predominant functional change preceding the premature reproductive senescence induced by chronic exposure to low doses of the AHR-specific ligand TCDD.


Asunto(s)
Exposición a Riesgos Ambientales , Contaminantes Ambientales/farmacología , Ovario/efectos de los fármacos , Ovario/fisiopatología , Dibenzodioxinas Policloradas/administración & dosificación , Receptores de Hidrocarburo de Aril/agonistas , Reproducción/efectos de los fármacos , Animales , Estradiol/metabolismo , Estro/metabolismo , Femenino , Hormona Liberadora de Gonadotropina/farmacología , Gonadotropinas/metabolismo , Hipófisis/efectos de los fármacos , Progesterona/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Vagina/efectos de los fármacos , Vagina/patología , Vagina/fisiopatología
20.
J Neurobiol ; 65(2): 192-204, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16118792

RESUMEN

Survival and growth of sympathetic neurons is regulated by nerve growth factor acting through trkA and p75NTR receptors. Sympathetic neurons are also affected by gonadal steroid hormones, particularly estrogen. To determine if estrogen may influence sympathetic neurons via altered neurotrophin receptor expression, we investigated effects of acute or chronic estrogen administration on levels of trkA and p75NTR proteins, numbers of immunoreactive neurons, and numbers of neurons expressing trkA, p75NTR, and estrogen receptor-alpha transcripts. Superior cervical ganglia from ovariectomized or estradiol-treated rats were processed for in situ hybridization or immunohistochemistry, and percentages of stained neurons quantitated or processed for Western blot analysis. In ovariectomized rats, approximately 50% of sympathetic neurons expressed trkA mRNA and protein. Acute estrogen administration did not affect trkA transcript expression, but reduced trkA protein significantly. Chronic treatment did not alter neuronal trkA expression. Approximately 70% of sympathetic neurons in ovariectomized rats expressed p75NTR transcripts and about 50% showed p75NTR immunoreactivity. Acute estrogen did not affect p75NTR expression. However, chronic estrogen reduced p75NTR mRNA and protein expression significantly. Fifty to sixty percent of sympathetic neurons in ovariectomized rats displayed estrogen receptor-alpha mRNA. After acute estrogen administration, estrogen receptor-alpha transcript expression increased by 35%, although this was not maintained chronically. These findings indicate that estrogen can influence sympathetic neuronal neurotrophin receptor expression as well as estrogen receptor-alpha. Reduced trkA expression after acute estrogen may transiently predispose neurons to degenerative events, while diminished p75NTR expression by chronic estrogen administration may exert long-term effects on survival or axonal outgrowth in sympathetic neurons.


Asunto(s)
Estradiol/farmacología , Receptor de Factor de Crecimiento Nervioso/genética , Receptor trkA/genética , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiología , Animales , Western Blotting , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Expresión Génica/efectos de los fármacos , Ovariectomía , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Receptor de Factor de Crecimiento Nervioso/metabolismo , Receptor trkA/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...