Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Reprod Sci ; 29(5): 1449-1456, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34731458

RESUMO

Reproductive health can be affected by obesity through various mechanisms. Obesity-associated inflammatory markers and altered hormones can have direct and indirect impacts on female reproductive health. However, little is known about obesity prevalence and trend among infertile women and obesity association with infertility in reproductive-age women of the U.S. In a cross-sectional study, we sought to conduct a secondary analysis of National Survey of Family Growth (NSFG) data from 2011 to 2019. A total of 6,035 infertile and non-pregnant fecund women aged 20-44 years were included in a nationally representative sample of the U.S. The weighted prevalence of obesity (BMI ≥ 30) among infertile women was 41.63% for the period 2011-2019. Obese women had 62% higher odds of infertility (95% CI 1.24, 2.17) compared to non-obese women of reproductive age (20-44 years). However, there was no specific trend of obesity among infertile and non-pregnant fecund women aged 20-44 years in the U.S. from 2011 to 2019. Given the negative impact of obesity on female reproductive health and in light of our results, it is important to counsel obese infertile reproductive-age women of the U.S. about behavioral changes that include weight management. Future longitudinal studies are needed to evaluate the risk of infertility among obese women.


Assuntos
Infertilidade Feminina , Estudos Transversais , Feminino , Humanos , Infertilidade Feminina/complicações , Infertilidade Feminina/epidemiologia , Obesidade/complicações , Obesidade/epidemiologia , Estados Unidos/epidemiologia
5.
BMC Genomics ; 18(1): 280, 2017 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-28381255

RESUMO

BACKGROUND: Infertility in dairy cattle is a concern where reduced fertilization rates and high embryonic loss are contributing factors. Studies of the paternal contribution to reproductive performance are limited. However, recent discoveries have shown that, in addition to DNA, sperm delivers transcription factors and epigenetic components that are required for fertilization and proper embryonic development. Hence, characterization of the paternal contribution at the time of fertilization is warranted. We hypothesized that sire fertility is associated with differences in DNA methylation patterns in sperm and that the embryonic transcriptomic profiles are influenced by the fertility status of the bull. Embryos were generated in vitro by fertilization with either a high or low fertility Holstein bull. Blastocysts derived from each high and low fertility bulls were evaluated for morphology, development, and transcriptomic analysis using RNA-Sequencing. Additionally, DNA methylation signatures of sperm from high and low fertility sires were characterized by performing whole-genome DNA methylation binding domain sequencing. RESULTS: Embryo morphology and developmental capacity did not differ between embryos generated from either a high or low fertility bull. However, RNA-Sequencing revealed 98 genes to be differentially expressed at a false discovery rate < 1%. A total of 65 genes were upregulated in high fertility bull derived embryos, and 33 genes were upregulated in low fertility derived embryos. Expression of the genes CYCS, EEA1, SLC16A7, MEPCE, and TFB2M was validated in three new pairs of biological replicates of embryos. The role of the differentially expressed gene TFB2M in embryonic development was further assessed through expression knockdown at the zygotic stage, which resulted in decreased development to the blastocyst stage. Assessment of the epigenetic signature of spermatozoa between high and low fertility bulls revealed 76 differentially methylated regions. CONCLUSIONS: Despite similar morphology and development to the blastocyst stage, preimplantation embryos derived from high and low fertility bulls displayed significant transcriptomic differences. The relationship between the paternal contribution and the embryonic transcriptome is unclear, although differences in methylated regions were identified which could influence the reprogramming of the early embryo. Further characterization of paternal factors delivered to the oocyte could lead to the identification of biomarkers for better selection of sires to improve reproductive efficiency.


Assuntos
Blastocisto/metabolismo , Metilação de DNA , Desenvolvimento Embrionário/genética , Fertilidade/genética , Espermatozoides/metabolismo , Transcriptoma , Animais , Bovinos , Ilhas de CpG , Epigenômica/métodos , Feminino , Fertilização in vitro , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Sequenciamento de Nucleotídeos em Larga Escala , Masculino , Oócitos/metabolismo , Gravidez , Reprodutibilidade dos Testes
6.
BMC Cancer ; 17(1): 102, 2017 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-28166758

RESUMO

BACKGROUND: Ovarian cancer remains the most deadly gynecological cancer with a poor aggregate survival rate; however, the specific rates are highly dependent on the stage of the disease upon diagnosis. Current screening and imaging tools are insufficient to detect early lesions and are not capable of differentiating the subtypes of ovarian cancer that may benefit from specific treatments. METHOD: As an alternative to current screening and imaging tools, we utilized wavelength dependent collagen-specific Second Harmonic Generation (SHG) imaging microscopy and optical scattering measurements to probe the structural differences in the extracellular matrix (ECM) of normal stroma, benign tumors, endometrioid tumors, and low and high-grade serous tumors. RESULTS: The SHG signatures of the emission directionality and conversion efficiency as well as the optical scattering are related to the organization of collagen on the sub-micron size scale and encode structural information. The wavelength dependence of these readouts adds additional characterization of the size and distribution of collagen fibrils/fibers relative to the interrogating wavelengths. We found a strong wavelength dependence of these metrics that are related to significant structural differences in the collagen organization and are consistent with the dualistic classification of type I and II serous tumors. Moreover, type I endometrioid tumors have strongly differing ECM architecture than the serous malignancies. The SHG metrics and optical scattering measurements were used to form a linear discriminant model to classify the tissues, and we obtained high accuracy (>90%) between high-grade serous tumors from the other tissue types. High-grade serous tumors account for ~70% of ovarian cancers, and this delineation has potential clinical applications in terms of supplementing histological analysis, understanding the etiology, as well as development of an in vivo screening tool. CONCLUSIONS: SHG and optical scattering measurements provide sub-resolution information and when combined provide superior diagnostic power over clinical imaging modalities. Additionally the measurements are able to delineate the different subtypes of ovarian cancer and may potentially assist in treatment protocols. Understanding the altered collagen assembly can supplement histological analysis and provide new insight into the etiology. These methods could become an in vivo screening tool for earlier detection which is important since ovarian malignancies can metastasize while undetectable by current clinical imaging resolution.


Assuntos
Matriz Extracelular/patologia , Neoplasias Ovarianas/diagnóstico , Microscopia de Geração do Segundo Harmônico/métodos , Feminino , Humanos , Gradação de Tumores/métodos , Neoplasias Ovarianas/patologia
7.
J Ovarian Res ; 9(1): 47, 2016 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-27488614

RESUMO

BACKGROUND: Improved cancer therapeutics and enhanced cancer survivorship have emphasized the severe long-term side effects of chemotherapy. Specifically, studies have linked many chemotherapy agents with primary ovarian insufficiency, although an exact insult model has not yet been determined. To investigate and ultimately solve this problem, a novel device for extended study of mammalian ovaries in vitro was developed. METHODS: A bioreactor was fabricated for bovine ovarian culture that provides intravascular delivery of media to the ovary through isolation and cannulation of a main ovarian artery branch. Whole ovaries were cultured in vitro using three methods: (1) continuously supplied fresh culture media, (2) recirculated culture media, or (3) continuously supplied fresh culture media supplemented with 500 nM doxorubicin for 24 or 48 h. TUNEL assay was used to assess apoptotic cell percentages in the three groups as compared to uncultured baseline ovaries. RESULTS: The ovary culture method was shown to maintain cell viability by effectively delivering nutrient-enriched pH-balanced media at a constant flow rate. Lower apoptosis observed in ovaries cultured in continuously supplied fresh culture media illustrates that this culture device and method are the first to sustain whole bovine ovary viability for 48 h. Meanwhile, the increase in the percentage of cell apoptosis with doxorubicin treatment indicates that the device can provide an alternative model for testing chemotherapy and chemoprotection treatments to prevent primary ovarian insufficiency in cancer patients. CONCLUSIONS: An ovarian bioreactor with consistent culture media flow through an ovarian vasculature-assisted approach maintains short-term whole bovine ovary viability.


Assuntos
Meios de Cultura/química , Técnicas de Cultura de Órgãos/instrumentação , Técnicas de Cultura de Órgãos/métodos , Ovário/citologia , Animais , Reatores Biológicos/veterinária , Bovinos , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/farmacologia , Feminino , Humanos , Técnicas de Cultura de Órgãos/veterinária
8.
Reprod Biol ; 15(4): 247-56, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26679166

RESUMO

Our objective was to determine the effects of prostaglandin F2α (PGF2α) and withdrawal of luteotropic stimulants (forskolin or hCG) on expression of chemokines and prostaglandin-endoperoxide synthase 2 (PTGS2) in luteinized human granulosa cells. Human granulosa cells were collected from 12 women undergoing oocyte retrieval and were luteinized in vitro with forskolin or hCG. In first experiment, granulosa-lutein cells were treated with PGF2α, the primary luteolytic hormone in most species. In second experiment, granulosa cells that had been luteinized for 8 d had luteotropins withdrawn for 1, 2, or 3 d. Treatment with PGF2α induced mRNA for chemokine (c-x-c motif) ligand 2 (CXCL2) and CXC ligand 8 (CXCL8; also known as interleukin-8) in granulosa cells luteinized for 8 d but not in cells that were only luteinized for 2 d. Similarly, luteinization of human granulosa cells for 8 d with forskolin or hCG followed by withdrawal of luteotropic stimulants, not only decreased P4 production, but also increased mRNA concentrations for CXCL8, CXCL-2 (after forskolin withdrawal), and PTGS2. These results provide evidence for two key steps in differentiation of luteolytic capability in human granulosa cells. During 8 d of luteinization, granulosa cells acquire the ability to respond to luteolytic factors, such as PGF2α, with induction of genes involved in immune function and PG synthesis. Finally, a decline in luteotropic stimuli triggers similar pathways leading to induction of PTGS2 and possibly intraluteal PGF2α production, chemokine expression, leukocyte infiltration and activation, and ultimately luteal regression.


Assuntos
Quimiocinas/biossíntese , Corpo Lúteo/fisiologia , Dinoprosta/biossíntese , Luteólise/fisiologia , Hormônios Adeno-Hipofisários/metabolismo , Prostaglandinas/biossíntese , Células Cultivadas , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Gonadotropina Coriônica/farmacologia , Colforsina/farmacologia , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Feminino , Regulação da Expressão Gênica/fisiologia , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Hormônios Adeno-Hipofisários/genética , Progesterona/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
9.
PLoS One ; 10(11): e0142588, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26544188

RESUMO

Advances in cancer treatment utilizing multiple chemotherapies have dramatically increased cancer survivorship. Female cancer survivors treated with doxorubicin (DXR) chemotherapy often suffer from an acute impairment of ovarian function, which can persist as long-term, permanent ovarian insufficiency. Dexrazoxane (Dexra) pretreatment reduces DXR-induced insult in the heart, and protects in vitro cultured murine and non-human primate ovaries, demonstrating a drug-based shield to prevent DXR insult. The present study tested the ability of Dexra pretreatment to mitigate acute DXR chemotherapy ovarian toxicity in mice through the first 24 hours post-treatment, and improve subsequent long-term fertility throughout the reproductive lifespan. Adolescent CD-1 mice were treated with Dexra 1 hour prior to DXR treatment in a 1:1 mg or 10:1 mg Dexra:DXR ratio. During the acute injury period (2-24 hours post-injection), Dexra pretreatment at a 1:1 mg ratio decreased the extent of double strand DNA breaks, diminished γH2FAX activation, and reduced subsequent follicular cellular demise caused by DXR. In fertility and fecundity studies, dams pretreated with either Dexra:DXR dose ratio exhibited litter sizes larger than DXR-treated dams, and mice treated with a 1:1 mg Dexra:DXR ratio delivered pups with birth weights greater than DXR-treated females. While DXR significantly increased the "infertility index" (quantifying the percentage of dams failing to achieve pregnancy) through 6 gestations following treatment, Dexra pretreatment significantly reduced the infertility index following DXR treatment, improving fecundity. Low dose Dexra not only protected the ovaries, but also bestowed a considerable survival advantage following exposure to DXR chemotherapy. Mouse survivorship increased from 25% post-DXR treatment to over 80% with Dexra pretreatment. These data demonstrate that Dexra provides acute ovarian protection from DXR toxicity, improving reproductive health in a mouse model, suggesting this clinically available drug may provide ovarian protection for cancer patients.


Assuntos
Antineoplásicos/efeitos adversos , Dexrazoxano/uso terapêutico , Doxorrubicina/efeitos adversos , Ovário/efeitos dos fármacos , Substâncias Protetoras/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Ensaio Cometa , Dano ao DNA , Avaliação Pré-Clínica de Medicamentos , Feminino , Fertilidade/efeitos dos fármacos , Tamanho da Ninhada de Vivíparos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos , Ovário/fisiologia , Fosforilação
10.
Clin Med Rev Case Rep ; 2(2)2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26523290

RESUMO

Uterine artery embolization for intractable postpartum hemorrhage saves lives while preserving fertility. The procedure-related risks of uterine infarction and ovarian insufficiency are rare. A primparous patient underwent bilateral internal hypogastric artery embolization to control severe postpartum hemorrhage following primary cesarean section. The bleeding continued, and a repeat aortogram demonstrated significant filling of the uterus from an anomalous proximal take off of the right uterine artery and from the left ovarian artery. Further embolization was required to control the bleeding. The patient developed acute primary ovarian insufficiency within two weeks of the procedure and subsequently presented with uterine infarction necessitating hysterectomy. This case demonstrates the increased risk of acute ovarian insufficiency and uterine infarction following uterine artery embolization for postpartum hemorrhage in the settings of aberrant pelvic vasculature.

11.
J Reprod Med ; 60(7-8): 345-53, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26380495

RESUMO

OBJECTIVE: To assess subsequent utilization of fertility treatment in reproductive-age women with cervical cancer (CC) who underwent ovarian transposition (OT) to preserve fertility prior to pelvic radiation. STUDY DESIGN: This is a case series of 216 CC patients seen in a comprehensive cancer center. Sixteen patients underwent OT for fertility preservation prior to pelvic radiation. Patients were assessed for utilization of fertility treatment, follicle-stimulating hormone (FSH) levels as a measure of ovarian reserve, and functional assessment of chronic illness therapy-cervix cancer (FACT-CX) to assess quality of life after OT. RESULTS: Of the patients, 94% of patients [corrected] maintained regular menstrual cycles 3 years after ovarian transposition (OT) [corrected] surgery (15/16). When measured (n = 5), serum FSH was normal at baseline and showed a transient elevation at 3 months following chemoradiation, with a return to normal levels at 6 months (means, 6.33 ± 2.94, 48.44 ± 18.63, and 12.52 ± 8.25 mIU/mL, respectively). Only 1 patient in this series attempted fertility treatment (in vitro fertilization) following OT, and she did not become pregnant. FACT-CX indicated that quality of life did not change significantly over the 6 months' duration following OT and chemoradiation therapy. CONCLUSION: OT preserves menstrual cycle regularity without negatively impacting patients' quality of life. The utility of OT as an effective fertility preservation option is hampered by the low utilization rate of in vitro fertilization and lack of ovarian reserve assessment following OT.


Assuntos
Preservação da Fertilidade/estatística & dados numéricos , Fertilização in vitro/estatística & dados numéricos , Ovário/cirurgia , Neoplasias do Colo do Útero/epidemiologia , Neoplasias do Colo do Útero/radioterapia , Neoplasias do Colo do Útero/cirurgia , Adulto , Feminino , Preservação da Fertilidade/métodos , Humanos , Pessoa de Meia-Idade , Estudos Retrospectivos , Resultado do Tratamento , Adulto Jovem
12.
Clin Med Rev Case Rep ; 2(2): 4, 2015 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-26366440

RESUMO

Fertility sparing surgery is advocated for reproductive-age women with benign and borderline ovarian tumors. The hormonal milieu of pregnancy may, however, complicate the decision making process. The patient presented in the third trimester with a rapidly growing tumor that was diagnosed as benign steroid cell tumor by intraoperative frozen section. Fertility-sparing surgery with right oophorectomy and partial left oophorectomy was performed. The final pathology examination demonstrated signet cells staining positive for mucin, which is pathognomonic for Krukenberg tumors. Krukenberg cells were overlooked in the frozen section due to the predominance of hormonally active luteinized ovarian stroma cells. This case highlights the challenges associated with fertility sparing surgery in women presenting with ovarian tumors in pregnancy and the limitations of frozen section in providing an accurate diagnosis.

13.
J Pediatr Adolesc Gynecol ; 28(2): 95-101, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25850590

RESUMO

OBJECTIVE: Female childhood cancer survivors are at an increased risk of reproductive health impairment. We compared reproductive health outcomes with the recommended standard in a cohort of childhood cancer survivors. STUDY DESIGN AND PARTICIPANTS: A retrospective chart review of 222 female childhood cancer survivors aged 21 years or younger that presented to a tertiary referral center between 1997-2008 was initiated. The main outcome measures were the compliance with the American Society of Clinical Oncology guidelines for childhood cancer survivor management of reproductive health. In particular, we evaluated menstrual cycle regularity, fertility preservation counseling, and endocrine profile, as defined by follicle stimulating hormone (FSH) and anti-mullerian hormone (AMH) levels as surrogate markers for ovarian reserve. Secondary outcomes were to study the contribution of survivor clinics in enforcing these guidelines. RESULTS: Of 136 patients older than 13 years at their last visit, 58 patients (43%) had FSH data available and none had AMH data. Patients were stratified into 3 groups according to FSH levels. Forty of 58 patients (69%) have normal ovarian reserve (FSH level < 10), 10 of 58 patients (17%) have decreased ovarian reserve (FSH levels 10-40), and 8 of 58 patients (14%) have premature menopause, defined as FSH > 40. Most patients with amenorrhea have elevated FSH levels indicating primary ovarian insufficiency, while 3 patients (2.2%) have low FSH levels consistent with hypothalamic amenorrhea. None of the patients were counseled on fertility preservation. CONCLUSIONS: Reproductive health follow-up in children with cancer, including FSH and AMH measurement when indicated, should be established and strictly adhered.


Assuntos
Fidelidade a Diretrizes/estatística & dados numéricos , Neoplasias/terapia , Serviços de Saúde Reprodutiva/estatística & dados numéricos , Saúde Reprodutiva , Adolescente , Adulto , Criança , Pré-Escolar , Estudos de Coortes , Aconselhamento , Feminino , Preservação da Fertilidade , Humanos , Ciclo Menstrual , Estudos Retrospectivos , Sobreviventes , Adulto Jovem
14.
Biol Reprod ; 92(3): 73, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25609833

RESUMO

Preservation of ovarian function following chemotherapy for nonovarian cancers is a formidable challenge. For prepubescent girls, the only option to prevent chemotherapy damage to the ovary is ovarian tissue cryopreservation, an experimental procedure requiring invasive surgeries to harvest and reimplant tissue, which carries the risk of cancer reintroduction. Drugs that block the primary mechanism of chemotherapy insult, such as dexrazoxane (Dexra) in the context of anthracycline chemotherapy, provide a novel approach for ovarian protection and have the potential to overcome current limitations to oncofertility treatment. Dexra is a catalytic topoisomerase 2 inhibitor that protects the mouse ovary from acute doxorubicin (DXR) chemotherapy toxicity in vitro by preventing DXR-induced DNA damage and subsequent gammaH2AX activation. To translate acute DXR ovarian insult and Dexra protection from mouse to nonhuman primate, freshly obtained marmoset ovarian tissue was cultured in vitro and treated with vehicle or 20 µM Dexra 1 h prior to 50 nM DXR. Cultured ovarian tissue was harvested at 2, 4, or 24 h post-DXR treatment. Dexra prevented DXR-induced DNA double-strand breaks as quantified by the neutral comet assay. DXR treatment for 24 h increased gammaH2AX phosphorylation, specifically increasing the number of foci-positive granulosa cells in antral follicles, while Dexra pretreatment inhibited DXR-induced gammaH2AX phosphorylation foci formation. Additionally, Dexra pretreatment trended toward attenuating DXR-induced AKT1 phosphorylation and caspase-9 activation as assayed by Western blots of ovarian tissue lysates. The combined findings suggest Dexra prevents primary DXR-induced DNA damage, the subsequent cellular response to DNA damage, and may diminish early apoptotic signaling in marmoset ovarian tissue. This study provides initial translation of Dexra protection against acute ovarian DXR toxicity from mice to marmoset monkey tissue.


Assuntos
Antineoplásicos/efeitos adversos , Callithrix , Dexrazoxano/farmacologia , Doxorrubicina/efeitos adversos , Ovário/efeitos dos fármacos , Inibidores da Topoisomerase II/farmacologia , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Caspase 9/metabolismo , Dano ao DNA/efeitos dos fármacos , Doxorrubicina/farmacologia , Feminino , Técnicas In Vitro , Modelos Animais , Ovário/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Técnicas de Cultura de Tecidos
15.
PLoS One ; 9(9): e108174, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25251158

RESUMO

Increasing numbers of female patients survive cancer, but succumb to primary ovarian insufficiency after chemotherapy. We tested the hypothesis that Bortezomib (Bort) protects ovaries from doxorubicin (DXR) chemotherapy by treating female mice with Bort 1 hour prior to DXR. By preventing DXR accumulation in the ovary, Bort attenuated DXR-induced DNA damage in all ovarian cell types, subsequent γH2AFX phosphorylation, and resulting apoptosis in preantral follicles. Bort pretreatment extended the number of litters per mouse, improved litter size and increased pup weight following DXR treatment, thus increasing the duration of post-chemotherapy fertility and improving pup health. As a promising prophylactic ovoprotective agent, Bort does not interfere with cancer treatment, and is currently used as a chemotherapy adjuvant. Bort-based chemoprotection may preserve ovarian function in a non-invasive manner that avoids surgical ovarian preservation, thus diminishing the health complications of premature menopause following cancer treatment.


Assuntos
Antibióticos Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico , Ácidos Borônicos/uso terapêutico , Doxorrubicina/efeitos adversos , Ovário/efeitos dos fármacos , Insuficiência Ovariana Primária/induzido quimicamente , Insuficiência Ovariana Primária/prevenção & controle , Pirazinas/uso terapêutico , Animais , Peso ao Nascer/efeitos dos fármacos , Bortezomib , Dano ao DNA/efeitos dos fármacos , Feminino , Fertilidade/efeitos dos fármacos , Humanos , Tamanho da Ninhada de Vivíparos/efeitos dos fármacos , Camundongos , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/patologia , Neoplasias Ovarianas/tratamento farmacológico , Ovário/patologia , Insuficiência Ovariana Primária/patologia
16.
Front Genet ; 5: 91, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24795753

RESUMO

MicroRNAs (miRNA) are short non-coding RNAs which act to regulate expression of genes driving numerous cellular processes. These RNAs are secreted within exosomes from cells into the extracellular environment where they may act as signaling molecules. In addition, they are relatively stable and are specifically expressed in association to certain cancers making them strong candidates as biological markers. Moreover, miRNAs have been detected in body fluids including urine, milk, saliva, semen, and blood plasma. However, it is unknown whether they are secreted by embryonic cells into the culture media. Given that miRNAs are expressed throughout embryonic cellular divisions and embryonic genome activation, we hypothesized that they are secreted from the embryo into the extracellular environment and may play a role in the developmental competence of bovine embryos. To test this hypothesis, bovine embryos were cultured individually from day 5 to day 8 of development in an in vitro fertilization system and gene expression of 5 miRNAs was analyzed in both embryos and culture media. Differential miRNA gene expression was observed between embryos that developed to the blastocyst stage and those that failed to develop from the morula to blastocyst stage, deemed degenerate embryos. MiR-25, miR-302c, miR-196a2, and miR-181a expression was found to be higher in degenerate embryos compared to blastocyst embryos. Interestingly, these miRNAs were also found to be expressed in the culture media of both bovine and human pre-implantation embryos. Overall, our results show for the first time that miRNAs are secreted from pre-implantation embryos into culture media and that miRNA expression may correlate with developmental competence of the embryo. Expression of miRNAs in in vitro culture media could allow for the development of biological markers for selection of better quality embryos and for subsequent successful pregnancy.

17.
PLoS One ; 7(8): e42293, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22876313

RESUMO

Primary ovarian insufficiency (POI) is one of the many unintended consequences of chemotherapy faced by the growing number of female cancer survivors. While ovarian repercussions of chemotherapy have long been recognized, the acute insult phase and primary sites of damage are not well-studied, hampering efforts to design effective intervention therapies to protect the ovary. Utilizing doxorubicin (DXR) as a model chemotherapy agent, we defined the acute timeline for drug accumulation, induced DNA damage, and subsequent cellular and follicular demise in the mouse ovary. DXR accumulated first in the core ovarian stroma cells, then redistributed outwards into the cortex and follicles in a time-dependent manner, without further increase in total ovarian drug levels after four hours post-injection. Consistent with early drug accumulation and intimate interactions with the blood supply, stroma cell-enriched populations exhibited an earlier DNA damage response (measurable at 2 hours) than granulosa cells (measurable at 4 hours), as quantified by the comet assay. Granulosa cell-enriched populations were more sensitive however, responding with greater levels of DNA damage. The oocyte DNA damage response was delayed, and not measurable above background until 10-12 hours post-DXR injection. By 8 hours post-DXR injection and prior to the oocyte DNA damage response, the number of primary, secondary, and antral follicles exhibiting TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling)-positive granulosa cells plateaued, indicating late-stage apoptosis and suggesting damage to the oocytes is subsequent to somatic cell failure. Primordial follicles accumulate significant DXR by 4 hours post-injection, but do not exhibit TUNEL-positive granulosa cells until 48 hours post-injection, indicating delayed demise. Taken together, the data suggest effective intervention therapies designed to protect the ovary from chemotherapy accumulation and induced insult in the ovary must act almost immediately to prevent acute insult as significant damage was seen in stroma cells within the first two hours.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Doxorrubicina/toxicidade , Ovário/citologia , Ovário/efeitos dos fármacos , Animais , Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Doxorrubicina/farmacologia , Feminino , Células da Granulosa/efeitos dos fármacos , Histonas/metabolismo , Camundongos , Folículo Ovariano/citologia , Folículo Ovariano/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Insuficiência Ovariana Primária/induzido quimicamente , Fatores de Tempo
18.
Biol Reprod ; 86(3): 96, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22190700

RESUMO

Doxorubicin (DXR) is a frontline chemotherapy agent implicated in unintended ovarian failure in female cancer survivors. The fertility preservation techniques currently available for cancer patients are often time and cost prohibitive and do not necessarily preserve endocrine function. There are no drug-based ovary protection therapies clinically available. This study provides the first investigation using dexrazoxane (Dexra) to limit DXR insult in ovarian tissue. In KK-15 granulosa cells, a 3-h DXR treatment increased double-strand (ds) DNA breaks 40%-50%, as quantified by the neutral comet assay, and dose-dependent cytotoxicity. Dexra exhibited low toxicity in KK-15 cells, inducing no DNA damage and less than 20% cell loss. Cotreating KK-15 cells with Dexra prevented acute DXR-induced dsDNA damage. Similarly, Dexra attenuated the DXR-induced 40%-65% increase in dsDNA breaks in primary murine granulosa cells and cells from in vitro cultured murine ovaries. DXR can cause DNA damage either through a topoisomerase II-mediated pathway, based on DXR intercalation into DNA, or through oxidative stress. Cotreating KK-15 cells with 2 µM Dexra was sufficient to prevent DXR-induced, but not H(2)O(2)-induced, DNA damage. These data indicated the protective effects are likely due to Dexra's inhibition of topoisomerase II catalytic activity. This putative protective agent attenuated downstream cellular responses to DXR, preventing H2AFX activation in KK-15 cells and increasing viability as demonstrated by increasing the DXR lethal dose in KK-15 cells 5- to 8-fold (LD(20)) and primary murine granulosa cells 1.5- to 2-fold (LD(50)). These data demonstrate Dexra protects ovarian cells from DXR insult and suggest that it is a promising tool to limit DXR ovarian toxicity in vivo.


Assuntos
Antibióticos Antineoplásicos/efeitos adversos , Antineoplásicos/farmacologia , Doxorrubicina/efeitos adversos , Infertilidade Feminina/prevenção & controle , Ovário/efeitos dos fármacos , Razoxano/farmacologia , Animais , Antibióticos Antineoplásicos/farmacologia , Linhagem Celular , Células Cultivadas , Dano ao DNA/efeitos dos fármacos , DNA Topoisomerases Tipo II/efeitos dos fármacos , DNA Topoisomerases Tipo II/fisiologia , Relação Dose-Resposta a Droga , Doxorrubicina/farmacologia , Feminino , Células da Granulosa/citologia , Células da Granulosa/efeitos dos fármacos , Histonas/antagonistas & inibidores , Histonas/fisiologia , Peróxido de Hidrogênio/farmacologia , Camundongos , Camundongos Endogâmicos , Modelos Animais , Ovário/citologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia
19.
J Steroid Biochem Mol Biol ; 126(3-5): 78-86, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21600284

RESUMO

Selective estrogen receptor modulators (SERMs) demonstrate differential endometrial cancer (EC) risk. While tamoxifen (TAM) use increases the risk of endometrial hyperplasia and malignancy, raloxifene (RAL) has neutral effects on the uterus. How TAM increases the risk of EC and why TAM and RAL differentially modulate the risk for EC, however, remain elusive. Here, we tested the hypothesis that TAM increases the risk for EC, at least in part, by enhancing the local estrogen biosynthesis and directing estrogen metabolism towards the formation of genotoxic and hormonally active estrogen metabolites. In addition, the differential effects of TAM and RAL in EC risk are attributed to their differential effect on estrogen metabolism/metabolites. The endometrial cancer cell line (Ishikawa cells) and the nonmalignant immortalized human endometrial glandular cell line (EM1) were used for the study. The profile of estrogen/estrogen metabolites (EM), depurinating estrogen-DNA adducts, and the expression of estrogen-metabolizing enzymes in cells treated with 17ß-estradiol (E2) alone or in combination with TAM or RAL were investigated using high performance liquid chromatography-electrospray ionization-tandem mass spectrometry (HPLC-ESI-MS(2)), ultraperformance liquid chromatography/tandem mass spectrometry (UPLC-MS/MS), and Western blot analysis, respectively. TAM significantly increased the total EM and enhanced the formation of hormonally active and carcinogenic estrogen metabolites, 4-hydroxestrone (4-OHE1) and 16α-hydroxyestrone, with concomitant reduction in the formation of antiestrogenic and anticarcinogenic 2-hydroxyestradiol and 2-methoxyestradiol. Furthermore, TAM increased the formation of depurinating estrogen-DNA adducts 4-OHE1 [2]-1-N7Guanine and 4-OHE1 [2]-1-N3 Adenine. TAM-induced alteration in EM and depurinating DNA adduct formation is associated with altered expression of estrogen metabolizing enzymes CYP1A1, CYP1B1, COMT, NQO1, and SF-1 as revealed by Western blot analysis. In contrast to TAM, RAL has minimal effect on EM, estrogen-DNA adduct formation, or estrogen-metabolizing enzymes expression. These data show that TAM perturbs the balance of estrogen-metabolizing enzymes and alters the disposition of estrogen metabolites, which can explain, at least in part, the mechanism for TAM-induced EC. These results also implicate the differential effect of TAM and RAL on estrogen metabolism/metabolites as a potential mechanism for their disparate effects on the endometrium.


Assuntos
Carcinoma/etiologia , Neoplasias do Endométrio/etiologia , Estrogênios/metabolismo , Cloridrato de Raloxifeno/farmacologia , Tamoxifeno/farmacologia , Carcinoma/induzido quimicamente , Células Cultivadas , Adutos de DNA/metabolismo , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Neoplasias do Endométrio/induzido quimicamente , Estradiol/metabolismo , Estradiol/farmacologia , Antagonistas de Estrogênios/efeitos adversos , Antagonistas de Estrogênios/farmacologia , Feminino , Humanos , Redes e Vias Metabólicas/efeitos dos fármacos , Cloridrato de Raloxifeno/efeitos adversos , Fatores de Risco , Moduladores Seletivos de Receptor Estrogênico/efeitos adversos , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Tamoxifeno/efeitos adversos
20.
Reprod Sci ; 18(9): 858-67, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21421897

RESUMO

The estrogen metabolite 2-methoxyestradiol (2-ME2) is one of the most potent antiangiogenic and proapoptotic endogenous steroids. Herein, we investigate the effects of 2-ME2 on angiogenesis of cultured primary ovine uterine artery endothelial cells (UAECs) from nonpregnant follicular (F-UAECs), nonpregnant luteal (L-UAECs), and pregnant ewes (P-UAECs). Uterine artery endothelial cells were treated with vehicle control, 10(-8) mol/L 17ß-estradiol (17ßE2), or 10(-9) to 10(-6) mol/L 2-ME2. Angiogenesis, apotosis, and cell morphology were assessed by capillary tube formation, flowcytometry, and immunohistochemistry. 17ßE2 stimulated while 10(-6) mol/L 2-ME2 inhibited capillary tube formation in F-UAECs (P < .05). The inhibitory effects of 2-ME2 on angiogenesis were minimal in L-UAECs and were absent in P-UAECs when compared to controls. 10(-6) mol/L 2-ME2 increased apoptosis and inhibited microtubular structure equally in pregnant and nonpregnant UAECs when compared to control or 17ßE2 treatments. Thus, 2-ME2 inhibit capillary tube formation in F-UAECs while L-UAECs and P-UAECs are relatively unresponsive to the inhibitory effects of 2ME2 indicating that the pregnancy phenotypic state of the UAECs may modulate the action of 2-ME2 on capillary angiogenesis.


Assuntos
Estradiol/análogos & derivados , Prenhez/fisiologia , Artéria Uterina/efeitos dos fármacos , Útero/irrigação sanguínea , Útero/efeitos dos fármacos , 2-Metoxiestradiol , Animais , Apoptose/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Estradiol/farmacologia , Feminino , Microscopia de Contraste de Fase , Microtúbulos/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Gravidez , Ovinos , Útero/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...