Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
J Assist Reprod Genet ; 40(3): 443-454, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36542312

ABSTRACT

PURPOSE: As a secondary report to elucidate the diverse spectrum of oncofertility practices for childhood cancer around the globe, we present and discuss the comparisons of oncofertility practices for childhood cancer in limited versus optimum resource settings based on data collected in the Repro-Can-OPEN Study Part I & II. METHODS: We surveyed 39 oncofertility centers including 14 in limited resource settings from Africa, Asia, and Latin America (Repro-Can-OPEN Study Part I), and 25 in optimum resource settings from the USA, Europe, Australia, and Japan (Repro-Can-OPEN Study Part II). Survey questions covered the availability of fertility preservation and restoration options offered in case of childhood cancer as well as their degree of utilization. RESULTS: In the Repro-Can-OPEN Study Part I & II, responses for childhood cancer and calculated oncofertility scores showed the following characteristics: (1) higher oncofertility scores in optimum resource settings than in limited resource settings for ovarian and testicular tissue cryopreservation; (2) frequent utilization of gonadal shielding, fractionation of anticancer therapy, oophoropexy, and GnRH analogs; (3) promising utilization of oocyte in vitro maturation (IVM); and (4) rare utilization of neoadjuvant cytoprotective pharmacotherapy, artificial ovary, in vitro spermatogenesis, and stem cells reproductive technology as they are still in preclinical or early clinical research settings. CONCLUSIONS: Based on Repro-Can-OPEN Study Part I & II, we presented a plausible oncofertility best practice model to help optimize care for children with cancer in various resource settings. Special ethical concerns should be considered when offering advanced and innovative oncofertility options to children.


Subject(s)
Fertility Preservation , Neoplasms , Male , Female , Humans , Fertility Preservation/methods , Cryopreservation , Neoplasms/complications , Neoplasms/therapy , Surveys and Questionnaires , Australia
3.
J Assist Reprod Genet ; 39(8): 1693-1712, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35870095

ABSTRACT

Since 2007, the Oncofertility Consortium Annual Conference has brought together a diverse network of individuals from a wide range of backgrounds and professional levels to disseminate emerging basic and clinical research findings in fertility preservation. This network also developed enduring educational materials to accelerate the pace and quality of field-wide scientific communication. Between 2007 and 2019, the Oncofertility Consortium Annual Conference was held as an in-person event in Chicago, IL. The conference attracted approximately 250 attendees each year representing 20 countries around the world. In 2020, however, the COVID-19 pandemic disrupted this paradigm and precluded an in-person meeting. Nevertheless, there remained an undeniable demand for the oncofertility community to convene. To maintain the momentum of the field, the Oncofertility Consortium hosted a day-long virtual meeting on March 5, 2021, with the theme of "Oncofertility Around the Globe" to highlight the diversity of clinical care and translational research that is ongoing around the world in this discipline. This virtual meeting was hosted using the vFairs ® conference platform and allowed over 700 people to participate, many of whom were first-time conference attendees. The agenda featured concurrent sessions from presenters in six continents which provided attendees a complete overview of the field and furthered our mission to create a global community of oncofertility practice. This paper provides a synopsis of talks delivered at this event and highlights the new advances and frontiers in the fields of oncofertility and fertility preservation around the globe from clinical practice and patient-centered efforts to translational research.


Subject(s)
COVID-19 , Fertility Preservation , Neoplasms , COVID-19/epidemiology , Humans , Pandemics
4.
Ann Oncol ; 30(11): 1760-1775, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31418765

ABSTRACT

Oncofertility is a new interdisciplinary field at the intersection of oncology and reproductive medicine that expands fertility options for young cancer patients. The most common forms of hematological malignancies that occur in girls and young women and therefore necessitate oncofertility care are acute lymphocytic leukemia, acute myeloid leukemia, non-Hodgkin's lymphoma, and Hodgkin's lymphoma. Aggressive gonadotoxic anticancer regimens including alkylating chemotherapy and total body irradiation are used often in treating girls and young women with hematological malignancies. The risks of gonadotoxicity and subsequent iatrogenic premature ovarian insufficiency and fertility loss depend mainly on the type and stage of the disease, dose of anticancer therapy as well as the age of the patient at the beginning of treatment. To avoid or at least mitigate the devastating complications of anticancer therapy-induced gonadotoxicity, effective and comprehensive strategies that integrate different options for preserving and restoring fertility ranging from established to experimental strategies should be offered before, during, and after chemotherapy or radiotherapy. A multidisciplinary approach that involves strong coordination and collaboration between hemato-oncologists, gynecologists, reproductive biologists, research scientists, and patient navigators is essential to guarantee high standard of care.


Subject(s)
Fertility Preservation/methods , Hematologic Neoplasms/therapy , Medical Oncology/methods , Primary Ovarian Insufficiency/etiology , Reproductive Medicine/methods , Adolescent , Adult , Age Factors , Antineoplastic Agents, Alkylating/adverse effects , Cancer Survivors , Child , Child, Preschool , Female , Fertility/drug effects , Fertility/radiation effects , Fertility Preservation/standards , Hematologic Neoplasms/mortality , Humans , Infant , Infant, Newborn , Intersectoral Collaboration , Medical Oncology/organization & administration , Medical Oncology/standards , Patient Care Team/organization & administration , Patient Care Team/standards , Reproductive Medicine/organization & administration , Reproductive Medicine/standards , Standard of Care , Survival Rate , Treatment Outcome , Whole-Body Irradiation/adverse effects , Young Adult
5.
Sci Rep ; 5: 17709, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26633657

ABSTRACT

The gonadotoxic effects of chemotherapy and radiation may result in premature ovarian failure in premenopausal oncology patients. Although autotransplantation of ovarian tissue has led to successful live births, reintroduction of latent malignant cells inducing relapse is a significant concern. In this report, we investigated the design of biomaterial grafts for transplantation of isolated ovarian follicles as a means to preserve fertility. Primordial and primary ovarian follicles from young female mice were extracted and encapsulated into biomaterials for subsequent transplantation into adult mice. Among the formulations tested, aggregated follicles encapsulated within fibrin had enhanced survival and integration with the host tissue following transplantation relative to the fibrin-alginate and fibrin-collagen composites. All mice transplanted with fibrin-encapsulated follicles resumed cycling, and live births were achieved only for follicles transplanted within VEGF-loaded fibrin beads. The extent to which these procedures reduce the presence of metastatic breast cancer cells among the isolated follicles was evaluated, with significantly reduced numbers of cancer cells present relative to intact ovaries. This ability to obtain live births by transplanting isolated primordial and primary follicles, while also reducing the risk of re-seeding disease relative to ovarian tissue transplantation, may ultimately provide a means to preserve fertility in premenopausal oncology patients.


Subject(s)
Infertility, Female/therapy , Neoplasm Recurrence, Local/prevention & control , Ovarian Follicle/transplantation , Primary Ovarian Insufficiency/therapy , Transplants/transplantation , Animals , Biocompatible Materials/therapeutic use , Disease Models, Animal , Female , Humans , Infertility, Female/pathology , Live Birth , Mice , Neoplasm Recurrence, Local/pathology , Pregnancy , Primary Ovarian Insufficiency/pathology
6.
Zygote ; 23(4): 475-84, 2015 Aug.
Article in English | MEDLINE | ID: mdl-24666604

ABSTRACT

The aim of this study was to evaluate the influence of two-dimensional (2D) and three-dimensional (3D) alginate culture systems on in vitro development of pre-antral caprine follicles. In addition, the influence of the reproductive age of the ovary donor on the in vitro culture success was investigated. Pre-antral follicles from pre-pubertal or adult goats were isolated and cultured directly on a plastic surface (2D) or encapsulated in an alginate-based matrix (3D). After 18 days, the oocytes underwent in vitro maturation (IVM) and in vitro fertilization (IVF) to produce embryos. The 3D system showed higher rates of follicle survival, lower rates of oocyte extrusion, and a greater number of recovered oocytes for IVM and IVF (P < 0.05). Only pre-antral follicles from adult animals produced MII oocytes and embryos. The estradiol concentrations increased from day 2 to day 12 of culture in all groups tested (P < 0.05). Conversely, progesterone concentrations were lower in 3D-cultured follicles than in 2D-cultured follicles, with differences on days 2 and 6 of culture (P < 0.05). We provide compelling evidence that a 2D or 3D alginate in vitro culture system offers a promising approach to achieving full in vitro development of caprine pre-antral follicles to produce mature oocytes that are capable of fertilization and viable embryos.


Subject(s)
Cell Culture Techniques/methods , Oocytes/physiology , Ovarian Follicle/growth & development , Age Factors , Alginates , Animals , Cell Culture Techniques/instrumentation , Cell Survival , Estradiol/metabolism , Female , Fertilization in Vitro , Glucuronic Acid , Goats , Hexuronic Acids , In Vitro Oocyte Maturation Techniques/methods , Male , Oocytes/cytology , Ovarian Follicle/physiology , Puberty
7.
Mol Hum Reprod ; 20(11): 1077-89, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25143461

ABSTRACT

Rapid cellular zinc influx regulates early mammalian development during the oocyte-to-egg transition through modulation of the meiotic cell cycle. Despite the physiological necessity of this zinc influx, the molecular mechanisms that govern such accumulation are unknown. Here we show that the fully grown mammalian oocyte does not employ a transcriptionally based mechanism of zinc regulation involving metal response element-binding transcription factor-1 (MTF-1), as demonstrated by a lack of MTF-1 responsiveness to environmental zinc manipulation. Instead, the mammalian oocyte controls zinc uptake through two maternally derived and cortically distributed zinc transporters, ZIP6 and ZIP10. Targeted disruption of these transporters using several approaches during meiotic maturation perturbs the intracellular zinc quota and results in a cell cycle arrest at a telophase I-like state. This arrest phenocopies established models of zinc insufficiency during the oocyte-to-egg transition, indicating the essential function of these maternally expressed transporters. Labile zinc localizes to punctate cytoplasmic structures in the human oocyte, and ZIP6 and ZIP10 are enriched in the cortex. Altogether, we demonstrate a mechanism of metal regulation required for female gamete development that may be evolutionarily conserved.


Subject(s)
Cation Transport Proteins/physiology , Zinc/metabolism , Adolescent , Adult , Animals , Biological Transport/genetics , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Cycle/genetics , Female , Gene Expression Regulation , Homeostasis , Humans , Mice, Inbred Strains , Oocytes/metabolism , Response Elements , Telophase
8.
Mol Hum Reprod ; 19(12): 828-37, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24026057

ABSTRACT

In women, ovary and adrenal gland produce androgens. Androgens are essential drivers of the primordial to antral follicle development, prior to serving as substrate for estrogen production in the later stages of folliculogenesis. Androgens play a crucial role in the follicular-stromal intertalk by fine tuning the extracellular matrix and vessel content of the ovarian stroma. Local auto-and paracrine factors regulate androgen synthesis in the pre-antral follicle. Androgen excess is a hallmark of polycystic ovary syndrome and is a key contributor in the exaggerated antral follicle formation, stromal hyperplasia and hypervascularity. Hyperandrogenaemia overrides the follicular-stromal dialog, resulting in follicular arrest and disturbed ovulation. On the other hand, androgen deficiency is likely to have a negative impact on fertility as well, and further research is needed to examine the benefits of androgen-replacement therapy in subfertility.


Subject(s)
Androgens/physiology , Ovarian Follicle/growth & development , Polycystic Ovary Syndrome/etiology , Female , Humans , Infertility, Female/complications , Infertility, Female/metabolism , Ovarian Diseases/complications , Ovarian Diseases/metabolism , Ovarian Diseases/pathology , Ovarian Follicle/metabolism , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/pathology , Receptors, Androgen/metabolism
9.
Cell Death Differ ; 20(8): 987-97, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23598363

ABSTRACT

Non-proliferating oocytes within avascular regions of the ovary are exquisitely susceptible to chemotherapy. Early menopause and sterility are unintended consequences of chemotherapy, and efforts to understand the oocyte apoptotic pathway may provide new targets for mitigating this outcome. Recently, the c-Abl kinase inhibitor imatinib mesylate (imatinib) has become the focus of research as a fertoprotective drug against cisplatin. However, the mechanism by which imatinib protects oocytes is not fully understood, and reports of the drug's efficacy have been contradictory. Using in vitro culture and subrenal grafting of mouse ovaries, we demonstrated that imatinib inhibits the cisplatin-induced apoptosis of oocytes within primordial follicles. We found that, before apoptosis, cisplatin induces c-Abl and TAp73 expression in the oocyte. Oocytes undergoing apoptosis showed downregulation of TAp63 and upregulation of Bax. While imatinib was unable to block cisplatin-induced DNA damage and damage response, such as the upregulation of p53, imatinib inhibited the cisplatin-induced nuclear accumulation of c-Abl/TAp73 and the subsequent downregulation of TAp63 and upregulation of Bax, thereby abrogating oocyte cell death. Surprisingly, the conditional deletion of Trp63, but not ΔNp63, in oocytes inhibited apoptosis, as well as the accumulation of c-Abl and TAp73 caused by cisplatin. These data suggest that TAp63 is the master regulator of cisplatin-induced oocyte death. The expression kinetics of TAp63, c-Abl and TAp73 suggest that cisplatin activates TAp63-dependent expression of c-Abl and TAp73 and, in turn, the activation of TAp73 by c-Abl-induced BAX expression. Our findings indicate that imatinib protects oocytes from cisplatin-induced cell death by inhibiting c-Abl kinase, which would otherwise activate TAp73-BAX-mediated apoptosis. Thus, imatinib and other c-Abl kinase inhibitors provide an intriguing new way to halt cisplatin-induced oocyte death in early follicles and perhaps conserve the endocrine function of the ovary against chemotherapy.


Subject(s)
Antineoplastic Agents/adverse effects , Apoptosis/physiology , Cisplatin/adverse effects , Oocytes/physiology , Platinum/adverse effects , Signal Transduction/physiology , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzamides/pharmacology , Cells, Cultured , Cisplatin/pharmacology , DNA Damage/drug effects , DNA Damage/physiology , Dose-Response Relationship, Drug , Female , Imatinib Mesylate , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Models, Animal , Nuclear Proteins/drug effects , Nuclear Proteins/physiology , Oocytes/drug effects , Oogenesis/drug effects , Oogenesis/physiology , Piperazines/pharmacology , Platinum/pharmacology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/drug effects , Pyrimidines/pharmacology , Signal Transduction/drug effects , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/physiology , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/physiology
10.
Ultramicroscopy ; 128: 24-31, 2013 May.
Article in English | MEDLINE | ID: mdl-23500508

ABSTRACT

A dedicated analytical scanning transmission electron microscope (STEM) with dual energy dispersive spectroscopy (EDS) detectors has been designed for complementary high performance imaging as well as high sensitivity elemental analysis and mapping of biological structures. The performance of this new design, based on a Hitachi HD-2300A model, was evaluated using a variety of biological specimens. With three imaging detectors, both the surface and internal structure of cells can be examined simultaneously. The whole-cell elemental mapping, especially of heavier metal species that have low cross-section for electron energy loss spectroscopy (EELS), can be faithfully obtained. Optimization of STEM imaging conditions is applied to thick sections as well as thin sections of biological cells under low-dose conditions at room and cryogenic temperatures. Such multimodal capabilities applied to soft/biological structures usher a new era for analytical studies in biological systems.


Subject(s)
Erythrocytes/ultrastructure , Islets of Langerhans/ultrastructure , Microscopy, Electron, Scanning Transmission/instrumentation , Microscopy, Electron, Scanning Transmission/methods , Spectrometry, X-Ray Emission/instrumentation , Spectroscopy, Electron Energy-Loss/instrumentation , Tobacco Mosaic Virus/ultrastructure , Animals , Cryoelectron Microscopy/methods , Humans , Male , Metals, Heavy/analysis , Spectrometry, X-Ray Emission/methods , Spectroscopy, Electron Energy-Loss/methods , Spermatozoa/cytology , Spermatozoa/ultrastructure
11.
Reproduction ; 145(1): 19-32, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23108112

ABSTRACT

In vitro follicle growth in alginate hydrogels is a unique and versatile method for studying ovarian and follicle biology that may also have implications for fertility preservation. Current culture systems support the development of isolated mouse follicles from the secondary stage onward. However, it has been a challenge to grow smaller follicles in vitro due to the dissociation of the oocyte from companion somatic cells. Recent work has demonstrated that coculturing primary follicles with mouse embryonic fibroblasts or ovarian stromal cells supports follicle survival and growth. In this study, we demonstrate that follicles themselves can exert a beneficial coculture effect. When primary follicles were cultured in groups of five or ten (multiple follicle culture), there was increased growth and survival. The multiple follicle culture approach maintained follicle integrity and resulted in the formation of antral stage follicles containing meiotically competent gametes. The growth and survival of primary follicles were highly number dependent, with the most significant enhancement observed when the largest number of follicles was grown together. Our data suggest that the follicle unit is necessary to produce the secreted factors responsible for the supportive effects of multiple follicle culture, as neither denuded oocytes, oocyte-secreted factors, nor granulosa cells alone were sufficient to support early follicle growth in vitro. Therefore, there may be signaling from both the oocyte and the follicle that enhances growth but requires both components in a feedback mechanism. This work is consistent with current in vivo models for follicle growth and thus advances the movement to recapitulate the ovarian environment in vitro.


Subject(s)
Coculture Techniques , Ovarian Follicle/cytology , Paracrine Communication/physiology , Animals , Cell Communication/physiology , Cell Survival/physiology , Cells, Cultured , Female , Mice , Mice, Inbred Strains , Models, Animal , Oocytes/cytology , Oocytes/physiology , Ovarian Follicle/physiology
12.
Hum Reprod ; 27(6): 1801-10, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22456922

ABSTRACT

BACKGROUND: In vitro follicle growth is a promising fertility preservation strategy in which ovarian follicles are cultured to produce mature and fertilization-competent oocytes. However, in primates, there has been limited success with in vitro follicle growth starting from primordial and primary follicles because adequate isolation methods and culture strategies have not been established. Understanding how to use primordial follicles for fertility preservation has significant implications because these follicles are the most abundant in the ovary, are found in all females and are fairly resistant to cryopreservation and chemotherapeutics. METHODS: In the primate ovary, primordial follicles are concentrated near the collagen-rich ovarian cortex. To obtain these follicles, we separated the ovarian cortex prior to enzymatic digestion and enriched the primordial follicle concentration by using a novel double filtration system. To test the hypothesis that a rigid physical environment, as found in vivo, is optimal for survival, primordial follicles were cultured in different concentrations of alginate for up to 6 days. Follicle survival and morphology were monitored throughout the culture. RESULTS: We found that primate ovarian tissue can be maintained for up to 24 h at 4°C without compromising tissue or follicle health. Hundreds of intact and viable primordial follicles were isolated from each ovary independent of animal age. Follicle survival and morphology were more optimal when follicles were cultured in 2% alginate compared with 0.5% alginate. CONCLUSIONS: By mimicking the rigid ovarian environment through the use of biomaterials, we have established conditions that support primordial follicle culture. These results lay the foundations for studying the basic biology of primordial follicles in a controlled environment and for using primordial follicles for fertility preservation methods.


Subject(s)
Macaca mulatta , Ovarian Follicle/physiology , Tissue Culture Techniques/veterinary , Alginates , Animals , Culture Media , Female , Glucuronic Acid , Hexuronic Acids , Ovarian Follicle/growth & development , Ovary/physiology , Tissue Culture Techniques/methods , Tissue and Organ Harvesting/methods , Tissue and Organ Harvesting/veterinary
13.
Hum Reprod ; 26(9): 2473-85, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21669966

ABSTRACT

BACKGROUND: Ovarian tissue cryopreservation is an emerging fertility preservation option, and culturing follicles isolated from this tissue to obtain mature gametes may ultimately be the best solution for patients for whom transplantation is contraindicated. It is unclear, however, how patient-specific variables (including age, weight and menstrual cycle stage) impact follicle growth and quality during three-dimensional culture. METHODS: We used a mouse model to systematically determine how these variables impact in vitro follicle growth. We characterized metabolic and hormonal profiles of mice at specific ages, weights and cycle stages and secondary follicles from these cohorts were isolated and cultured. We then assessed follicle survival, growth and function, as well as meiotic competence and spindle morphology of the resulting oocytes. RESULTS: We found that older mice and mice with increased body weight had higher serum cholesterol, abnormal glucose tolerance and lower levels of circulating Anti-Müllerian hormone compared with younger and leaner controls. Secondary follicles isolated from different cohorts and grown in vitro had indistinguishable growth trajectories. However, the follicles isolated from older and heavier mice and those in diestrus had altered hormone profiles. These follicles contained oocytes with reduced meiotic competence and produced oocytes with greater spindle defects. CONCLUSIONS: These results suggest that the original physical environment of the follicle within the ovary can impact its function when isolated and cultured. These findings are valuable as we begin to use in vitro follicle growth technology for a heterogeneous fertility preservation patient population.


Subject(s)
Estrous Cycle , Ovarian Follicle/physiology , Tissue Culture Techniques , Age Factors , Animals , Anti-Mullerian Hormone/blood , Body Weight , Cholesterol/blood , Female , Fertility Preservation , Glucose/metabolism , Glucose Tolerance Test , Meiosis/physiology , Mice , Mice, Inbred Strains , Oocytes/cytology , Oocytes/physiology , Oocytes/ultrastructure , Spindle Apparatus/ultrastructure
14.
Reproduction ; 142(1): 113-22, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21502334

ABSTRACT

The present study examined the influences of the physical and hormonal microenvironment on in vitro growth and steroidogenesis of dog follicles. Follicles were enzymatically isolated and individually encapsulated in 0.5% (w/v; n=17) or 1.5% (n=10) alginate and cultured with 0.5 IU/ml equine chorionic gonadotropin for 192 h. In a separate experiment, follicles were encapsulated in 0.5% alginate and cultured with 0 (n=22), 1 (n=23), 10 (n=20) or 100 (n=21) µg/ml FSH for 240 h. Follicle diameter and steroid production were assessed every 48 h in both studies. Follicles encapsulated in the 0.5% alginate grew faster (P<0.05) than those cultured in the 1.5% concentration. Oestradiol (E(2)) and progesterone (P(4)) increased consistently (P<0.05) over time, and follicles in the 1.5% alginate produced more (P<0.05) P(4) than those in the 0.5% solution. Follicles cultured in the highest FSH concentration (100 µg/ml) increased 100% in size after 240 h compared with 50 to 70% in lower dosages. E(2) concentration remained unchanged over time (P>0.05) across FSH dosages. However, P(4) increased (P<0.05) as culture progressed and with increasing FSH concentration. Results demonstrate that dog follicles cultured in alginate retain structural integrity, grow in size and are hormonally active. Lower alginate and increasing FSH concentrations promote in vitro follicle growth. However, the absence of an E(2) rise in follicles cultured in FSH alone suggests the need for LH supplementation to support theca cell differentiation and granulosa cell function.


Subject(s)
Dogs , Estradiol/metabolism , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Progesterone/metabolism , Tissue Culture Techniques/veterinary , Alginates/chemistry , Alginates/metabolism , Animals , Cell Survival , Chemical Phenomena , Chorionic Gonadotropin/metabolism , Female , Follicle Stimulating Hormone/metabolism , Glucuronic Acid/chemistry , Glucuronic Acid/metabolism , Hexuronic Acids/chemistry , Hexuronic Acids/metabolism , Hydrogel, Polyethylene Glycol Dimethacrylate , Kinetics , Luteinizing Hormone/metabolism , Osmolar Concentration , Ovarian Follicle/cytology , Reproductive Techniques/veterinary
16.
Hum Reprod Update ; 16(4): 395-414, 2010.
Article in English | MEDLINE | ID: mdl-20124287

ABSTRACT

BACKGROUND Female cancer patients are offered 'banking' of gametes before starting fertility-threatening cancer therapy. Transplants of fresh and frozen ovarian tissue between healthy fertile and infertile women have demonstrated the utility of the tissue banked for restoration of endocrine and fertility function. Additional methods, like follicle culture and isolated follicle transplantation, are in development. METHODS Specialist reproductive medicine scientists and clinicians with complementary expertise in ovarian tissue culture and transplantation presented relevant published literature in their field of expertise and also unpublished promising data for discussion. As the major aims were to identify the current gaps prohibiting advancement, to share technical experience and to orient new research, contributors were allowed to provide their opinioned expert views on future research. RESULTS Normal healthy children have been born in cancer survivors after orthotopic transplantation of their cryopreserved ovarian tissue. Longevity of the graft might be optimized by using new vitrification techniques and by promoting rapid revascularization of the graft. For the in vitro culture of follicles, a successive battery of culture methods including the use of defined media, growth factors and three-dimensional extracellular matrix support might overcome growth arrest of the follicles. Molecular methods and immunoassay can evaluate stage of maturation and guide adequate differentiation. Large animals, including non-human primates, are essential working models. CONCLUSIONS Experiments on ovarian tissue from non-human primate models and from consenting fertile and infertile patients benefit from a multidisciplinary approach. The new discipline of oncofertility requires professionalization, multidisciplinarity and mobilization of funding for basic and translational research.


Subject(s)
Fertility , Ovarian Follicle/growth & development , Ovarian Follicle/transplantation , Tissue Culture Techniques , Tissue Preservation/methods , Animals , Cats , Female , Humans , Mice , Primates , Rats , Tissue Banks
17.
Steroids ; 74(12): 913-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19631674

ABSTRACT

Estrogen non-responsive estrogen receptor alpha (ERalpha) knock-in (ENERKI) mice have a mutation (glycine 525 to leucine, G525L) in the ligand-binding domain of ERalpha. The mutant ERalpha protein has a significantly lower affinity and response to endogenous estrogens, while not altering growth factor activated ligand-independent pathways. ENERKI females demonstrated signs of early follicle development as determined by a significant increase in antral follicle formation by 20 days of age. Adult ENERKI females were infertile, had hemorrhagic ovarian follicular cysts, and failed to develop corpora lutea in response to a superovulation regimen. These results illustrate the importance of ERalpha ligand-induced signaling for ovarian development and for estrogen feedback on the hypothalamus and pituitary. Although ERalpha ligand-induced signaling by endogenous estrogens is lost in ENERKI females, the ERalpha selective agonist propyl pyrazole triol (PPT), a synthetic nonsteroidal compound, is still able to activate G525L ERalphain vivo to increase uterine weight. To test whether PPT could restore ligand-dependent receptor activation, ENERKI females were treated with PPT and evaluated for spontaneous ovulation, ovarian hemorrhagic cysts, and LH serum levels. Daily PPT treatments beginning on day 4 of life prevented formation of ovarian hemorrhagic cysts in adult ENERKI animals. In accordance with this result, preputial gland weight and LH levels were also lowered in these animals, indicating PPT treatments most likely led to restoration of ERalpha negative feedback of the hypothalamic-pituitary axis.


Subject(s)
Estrogen Receptor alpha/genetics , Ovary/abnormalities , Ovary/growth & development , Animals , Drug Administration Schedule , Estrogen Receptor alpha/metabolism , Female , Gene Knock-In Techniques , Hemorrhage/prevention & control , Humans , Infertility/genetics , Ligands , Male , Mice , Ovarian Follicle/growth & development , Ovary/physiopathology , Phenols , Phenotype , Pregnancy , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Sexual Behavior, Animal , Signal Transduction/genetics , Superovulation/genetics
18.
J Endocrinol ; 178(1): 137-48, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12844345

ABSTRACT

The presence of activin A and its effects have previously been documented in the adrenal gland, particularly in the human fetal adrenal gland and the rat adrenal gland. The primary signaling pathway of activin involves interactions between receptor and intracellular (Smad) proteins that have not been completely described in the adrenal gland. In this study, we demonstrate that the components of the activin signaling cascade are present in two complementary models, the fetal rat adrenal gland and the human adrenocortical cell line, H295R, by means of RT-PCR, western analysis, and immunoprecipitation techniques. Using the cell line, activin signaling was analyzed using an activin-responsive reporter gene, p3TP-luc, and luciferase assays to assess transcriptional activity with co-expression of the different activin receptors and Smads to demonstrate the functionality of the signaling cascade. In the fetal rat adrenal gland, the relative amounts of mRNA of the type II receptors, RII and RIIB, were regulated by gestational age, such that the RIIB levels increased after birth while RII levels fell. Using immunodetection techniques, the activin receptors and the different Smad proteins were detected in the rat fetal adrenal glands. Notably, the presence of Smad4 protein is significantly increased after birth in the rat adrenal gland. RT-PCR established a similar profile in the H295R cells. Using p3TP-luc, the H295R cells show transcriptional activation of this activin-responsive reporter in the presence of activin A. Co-expression of type I and type II receptors as well as Smads, results in ligand-independent transcriptional activity in addition to an activin-stimulated response. In determining activin's effects on adrenal function, adrenal steroid production was evaluated by incubation of the H295R cells with increasing doses of activin A and inhibin A, resulting in a detectable increase in P450c17 expression. Co-incubation of activin A with follistatin diminishes this response. These results are consistent with a role for activin A in the adrenal gland by demonstrating that the elements of the activin signaling pathway are present, intact, and functional. This suggests that in the adrenal gland the components of the activin receptor/Smad pathway are dynamically changing in the transition from fetal to neonatal life, and are important to the function of this organ.


Subject(s)
Activins/metabolism , Adrenal Glands/embryology , Adrenal Glands/metabolism , Signal Transduction/physiology , Activin Receptors, Type II/metabolism , Activins/genetics , Adrenal Cortex/metabolism , Animals , Animals, Newborn , Blotting, Western/methods , Cell Line , DNA-Binding Proteins/metabolism , Gestational Age , Humans , Precipitin Tests , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Smad Proteins , Steroid 17-alpha-Hydroxylase/metabolism , Steroid 21-Hydroxylase/metabolism , Trans-Activators/metabolism , Transcription, Genetic
20.
J Endocrinol ; 174(1): 71-83, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12098665

ABSTRACT

Follicle-stimulating hormone (FSH) stimulates ovarian follicle development and the production of protein hormones including inhibin A and inhibin B. The inhibins are dimeric proteins (alpha-beta(A) or alpha-beta(B)) secreted by growing follicles that suppress FSH in a classical endocrine negative feedback loop. Siberian hamsters, Phodopus sungorus, exhibit seasonal variation in FSH levels. Given the role of inhibin in FSH regulation, we hypothesized that ovarian inhibin expression differs between animals reared in long (16 h light:8 h darkness) and short (6 h light:18 h darkness) photoperiods. To examine inhibin expression in animals housed under long or short photoperiods, hamster inhibin alpha-, beta(A)-, and beta(B)-subunits were cloned and used to detect and localize inhibin subunit mRNA in developing follicles. Ovarian inhibin alpha-subunit mRNA levels were significantly higher in long day-exposed (LD) than in short day-exposed (SD) hamsters. In addition, dimeric inhibin, as well as inhibin alpha-, beta(A)-, and beta(B)-subunit protein levels were higher in the LD than in the SD hamster ovaries.


Subject(s)
Inhibins/metabolism , Photoperiod , Amino Acid Sequence , Animals , Cricetinae , DNA, Complementary/metabolism , Female , In Situ Hybridization , Ovarian Follicle/metabolism , Phodopus , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...