Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
Reprod Biol Endocrinol ; 22(1): 51, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38671458

ABSTRACT

BACKGROUND: Ovarian damage and follicle loss are major side effects of chemotherapy in young female patients with cancer. However, effective strategies to prevent these injuries are still lacking. The purpose of this study was to verify low-intensity pulsed ultrasound (LIPUS) can reduce ovarian injury caused by chemotherapy and to explore its underlying mechanisms in mice model. METHODS: The mice were randomly divided into the Control group, Cisplatin group, and Cisplatin + LIPUS group. The Cisplatin group and Cisplatin + LIPUS group were intraperitoneally injected with cisplatin every other day for a total of 10 injections, and the Control group was injected with saline. On the second day of each injection, the Cisplatin + LIPUS group received irradiation, whereas the other two groups received sham irradiation. We used a variety of biotechnologies to detect the differences in follicle count, granulosa cell apoptosis, fibrosis, transcriptome level, oxidative damage, and inflammation in differently treated mice. RESULT: LIPUS was able to reduce primordial follicle pool depletion induced by cisplatin and inhibit the apoptosis of granulosa cells. Transcriptomic results confirmed that LIPUS can reduce ovarian tissue injury. We demonstrated that LIPUS can relieve ovarian fibrosis by inhibiting TGF-ß1/Smads pathway. Meanwhile, it can reduce the oxidative damage and reduced the mRNA levels of proinflammatory cytokines caused by chemotherapy. CONCLUSION: LIPUS can reduce the toxic effects of chemotherapy drugs on ovaries, inhibit ovarian fibrosis, reduce the inflammatory response, and redcue the oxidative damage, reduce follicle depletion and to maintain the number of follicle pools.


Subject(s)
Antineoplastic Agents , Cisplatin , Ovary , Ultrasonic Waves , Animals , Female , Mice , Cisplatin/adverse effects , Ovary/drug effects , Ovary/radiation effects , Ovary/pathology , Antineoplastic Agents/adverse effects , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Apoptosis/radiation effects , Ovarian Follicle/drug effects , Ovarian Follicle/radiation effects , Ultrasonic Therapy/methods
2.
Reprod Biomed Online ; 43(5): 799-809, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34602345

ABSTRACT

RESEARCH QUESTION: What is the impact of radiation exposure on oocyte quality and female fertility? DESIGN: Prepubertal mice underwent whole-body irradiation with a single dose (0.02, 0.1, 0.5, 2, 8 Gy) of gamma- or X-rays. Oocytes were quantified in irradiated (n = 36) and sham-treated (n = 8) mice. After a single exposure to 2 Gy, formation of DNA double-strand breaks (n = 10), activation of checkpoint kinase (Chk2) (n = 10) and dynamics of follicular growth (n = 18) were analysed. Fertility assessment was performed in adult irradiated mice and controls from the number of pups per mouse (n = 28) and the fetal abortion rate (n = 24). Ploidy of mature oocytes (n = 20) was analysed after CREST immunostaining, and uterine sections were examined. RESULTS: Radiation exposure induced a massive loss of primordial follicles with LD50 below 50 mGy for both gamma and X-rays. Growing follicles survived doses up to 8 Gy. This difference in radiosensitivity was not due to a different amount of radio-induced DNA damage, and Chk2 was activated in all oocytes. Exposure to a 2 Gy dose abolished the long-term fertility of females due to depletion of the ovarian reserve. Detailed analysis indicates that surviving oocytes were able to complete folliculogenesis and could be fertilized. This transient fertility allowed irradiated females to produce a single litter albeit with a high rate of fetal abortion (23%, P = 0.0096), related to altered ploidy in the surviving oocytes (25.5%, P = 0.0035). CONCLUSIONS: The effects of radiation on surviving oocyte quality question natural conception as a first-line approach in cancer survivors. Together, the data emphasize the need for fertility preservation before radiation exposure and call for reassessment of the use of cryopreserved oocytes.


Subject(s)
Fertility Preservation/methods , Oocytes/physiology , Oocytes/radiation effects , Ovary/radiation effects , Primary Ovarian Insufficiency/etiology , Abortion, Spontaneous , Aneuploidy , Animals , DNA/radiation effects , DNA Damage , Disease Models, Animal , Dose-Response Relationship, Radiation , Female , Gamma Rays , Mice , Mice, Inbred C57BL , Ovarian Follicle/radiation effects , Ovarian Reserve/radiation effects , Sexual Maturation/radiation effects , Whole-Body Irradiation , X-Rays
3.
J Assist Reprod Genet ; 38(10): 2745-2756, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34453231

ABSTRACT

PURPOSE: The huge loss of ovarian follicles after transplantation of frozen/thawed ovarian tissue is considered a major drawback on the efficacy of the procedure. Here we investigate whether Er:YAG laser treatment prior to xenotransplantation can improve re-vascularization and subsequently follicle survival in human ovarian tissue. METHODS: A total of 99 frozen/thawed human ovarian cortex pieces were included of which 72 pieces from 12 woman were transplanted to immunodeficient mice. Tissues from each woman were included in both an 8-day and an 8-week duration study and treated with either full-beam laser (L1) or fractionated laser (L2), or served as untreated controls. Vascularization of the ovarian xenografts were evaluated after 8 days by qPCR and murine Cd31 immunohistochemical analysis. Follicle densities were evaluated histologically 8 weeks after xenografting. RESULTS: Gene expression of Vegf/VEGF was upregulated after L1 treatment (p=0.002, p=0.07, respectively), whereas Angpt1, Angpt2, Tnf-α, and Il1-ß were significantly downregulated. No change in gene expression was found in Cd31/CD31, ANGPT1, ANGPT2, ANGTPL4, XBP1, or LRG1 after any of the laser treatments. The fraction of Cd31 positive cells were significantly reduced after L1 and L2 treatment (p<0.0001; p=0.0003, respectively), compared to controls. An overall negative effect of laser treatment was detected on follicle density (p=0.03). CONCLUSIONS: Er:YAG laser treatment did not improve re-vascularization or follicle survival in human ovarian xenografts after 8 days and 8 weeks grafting, respectively. However, further studies are needed to fully explore the potential angiogenic effects of controlled tissue damage using different intensities or lasers.


Subject(s)
Cryopreservation/methods , Fertility Preservation/methods , Lasers, Solid-State/therapeutic use , Ovarian Follicle/blood supply , Ovarian Follicle/cytology , Ovary/transplantation , Transplantation, Heterologous/methods , Animals , Female , Heterografts , Humans , Mice , Ovarian Follicle/radiation effects , Ovary/radiation effects
4.
J Ovarian Res ; 14(1): 26, 2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33546719

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) is the most common complex endocrine disorder affecting approximately 2-20% of reproductive aged females. Tumour Treating Fields (100-300 kHz) is a recent innovative, non-invasive therapeutic approach to cancer therapy. This frequency as an alternative therapy for the management of polycystic ovaries has not yet been explored. OBJECTIVES: To explore the effect of full-body exposure of 150 kHz Electromagnetic Radiation (EMR), on the development of polycystic ovaries in an estradiol valerate-induced PCO rat model. METHOD: Twenty-one female adult rats were divided into three groups (n = 7 each): control, Estradiol Valerate (EV) and EV + EMR groups. The EV + EMR group was subjected to full body exposure at 150 kHz EMR continuously for eight consecutive weeks. Estradiol valerate was administered orally to induce polycystic ovaries in EV and EV + EMR groups. Body and ovarian weights were recorded and analysed. The regularity of the estrous cycle was assessed in all three groups. The histological study of ovarian tissue was carried out by haematoxylin and eosin staining. The serum concentration levels of Luteinizing Hormone (LH), Follicle-Stimulating Hormone (FSH) and testosterone were measured using the ELISA method. RESULTS: The body and ovary weights did not differ significantly between the EV and EV + EMR groups. The estrous cycle was found to be irregular in both the EV and EV + EMR groups. Ovarian histology revealed near normal morphology with little or no degenerative and morphological changes in developing follicles in the exposed group. Histometrical analysis showed an increased number of developing follicles and a significant reduction in the number and size of follicular cysts (p < 0.05) in the EV + EMR group. Hormonal analysis revealed no significant difference in the testosterone and FSH levels between the EV + EMR and EV groups. However, the LH, LH/FSH ratio decreased significantly in the EV + EMR group compares to the EV group. CONCLUSION: The 150 kHz EMR appear to have little or no degenerative and morphological changes in the developing follicles, an increased number of typical developing follicles and a significant reduction in the number and size of the follicular cysts (p < 0.05).


Subject(s)
Electromagnetic Radiation , Estrous Cycle/radiation effects , Ovarian Follicle/radiation effects , Ovary/radiation effects , Polycystic Ovary Syndrome/pathology , Animals , Body Weight , Disease Models, Animal , Estradiol/toxicity , Estrogens/toxicity , Estrous Cycle/drug effects , Female , Follicle Stimulating Hormone/blood , Luteinizing Hormone/blood , Organ Size , Ovarian Follicle/drug effects , Ovary/drug effects , Ovary/pathology , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/radiotherapy , Rats , Rats, Sprague-Dawley , Testosterone/blood
5.
J Reprod Immunol ; 144: 103272, 2021 04.
Article in English | MEDLINE | ID: mdl-33465522

ABSTRACT

Radioprotective effects of Resveratrol is well known in normal cells exposed to the damaging effects of ionizing radiation however, its potential radioprotective effect on ovarian follicle formation and development is still uncertain. Astonishingly, it has been reported that PARP contributed to the pathogenesis of immune-mediated ovarian injury. In this paper, Resveratrol was tested for its inflammatory, anti-cervical carcinoma activity, and checked its targets poly (ADP-ribose) polymerase 1 (PARP-1) induced premature ovarian failure with a potent enzymatic modulatory activity. Through high-throughput virtual screening method, Resveratrol was screened to find its target. That the compound strongly inhibited cervical carcinoma HT-3 cell. The cell proliferation was evaluated by an CCK-8 assay, and the cell apoptosis was assessed by a flow cytometry. Rat model of premature ovarian failure was used to introduce resveratrol preparation and rtPCR was done to measure expression of apoptosis related markers. We report resveratrol as a potential target for PARP-1 and its modulator from a high-throughput virtual screening method. Resveratrol was measured its anti-cervical carcinoma activity by using an CCK-8 assay, which suggested that the compound strongly inhibited HT-3 cell proliferation, the IC50 value is 0.65 µM. In addition, the compound induced HT-3 cell apoptosis in a dose-response manner. Resveratrol preserves the entire ovarian follicle pool manifested by increasing serum anti-Müllerian hormone (AMH) levels. Study suggest that resveratrol restored ovarian function through increasing AMH levels, and diminishing ovarian inflammation, predominantly modulation of PPAR-1 and inhibition of inflammatory cytokines. Resveratrol was identified targets for PARP-1 from a high-throughput virtual screening method, strongly inhibited PARP-1 protein and HT-3 cell proliferation. Resveratrol is a promising PARP-1 modulator with anti-cervical carcinoma activity, which deserves further investigation.


Subject(s)
Carcinoma/drug therapy , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Primary Ovarian Insufficiency/drug therapy , Resveratrol/pharmacology , Uterine Cervical Neoplasms/drug therapy , Animals , Anti-Mullerian Hormone/blood , Apoptosis/drug effects , Carcinoma/immunology , Carcinoma/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Disease Models, Animal , Female , High-Throughput Screening Assays , Humans , Molecular Docking Simulation , Ovarian Follicle/drug effects , Ovarian Follicle/immunology , Ovarian Follicle/pathology , Ovarian Follicle/radiation effects , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Primary Ovarian Insufficiency/blood , Primary Ovarian Insufficiency/immunology , Rats , Resveratrol/therapeutic use , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/pathology , Whole-Body Irradiation/adverse effects
6.
Int J Radiat Biol ; 97(3): 367-375, 2021.
Article in English | MEDLINE | ID: mdl-33320730

ABSTRACT

PURPOSE: The sequelae of premature loss of ovarian function can undoubtedly have undesirable effects for a woman although radiotherapy is one of the most relevant treatment modalities for various types of malignancies. The aim of this study was to determine the effect of different doses of radiation on ovarian folliculogenesis, inflammation, and apoptotic markers. MATERIALS AND METHODS: For this purpose, 40 healthy Wistar albino female rats divided into four groups: 1) Control group; 2) those that were exposed to total body 1 Gy of gamma rays; 3) those that were exposed to the total body 5 Gy of gamma rays, and 4) those that were exposed to total body 10 Gy of gamma rays. External irradiation to the total body was given with gamma irradiation delivered by the Co60 teletherapy machine. The day after radiation application the rats were sacrificed and the ovaries were removed in all groups. Histopathologic examination, follicle counting, and classification were performed in the ovarian tissues. The expression of AMH, TNF-α, IL1-ß, Bax, and Bcl-2 was detected. The stained sections were examined for caspase 3 positive apoptotic cell numbers. RESULTS: The recorded results revealed that increased radiation dose induced obvious ovarian injuries that were indicated by histopathological, and immunohistochemical alterations, including elevation of ovarian injury markers. A significantly lower number of total and primordial follicles was detected with increasing radiation dose compared with the control group. According to our immunohistochemical results, 10 Gy of gamma rays group had the lowest AMH expression levels, while had the highest TNF-α, IL1-ß expression level compared to the control group. When the groups were evaluated in terms of apoptosis, it was seen that the number of caspase 3 positive cells and Bax immunoreactivity intensity increased with radiation dose. In contrast, Bcl-2 immunoreactivity intensity decreased with increasing radiation dose compared with the control group. CONCLUSIONS: We demonstrate here that dose rate plays an important role when estimating the relation between exposure to an increased dose of ionizing radiation and the risk of ovarian disease. According to these results, certain factors have to be optimized before introducing them into clinics.


Subject(s)
Ovarian Follicle/radiation effects , Animals , Anti-Mullerian Hormone/blood , Apoptosis , Dose-Response Relationship, Radiation , Female , Interleukin-1beta/analysis , Ovarian Follicle/pathology , Radiotherapy Dosage , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/analysis
7.
Lasers Med Sci ; 36(4): 703-713, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33111207

ABSTRACT

Androgenetic alopecia (AGA) is a global challenge, affecting a large number of people worldwide. Efficacy of the existed treatments can barely meet the demands of patients. Patients who are poorly responding to those treatments are seeking for a more effective and suitable technique to treat their disease. Low-level light therapy (LLLT) is a newly developed technique, which has been proved to stimulate hair growth. Based on the function principle of LLLT in other domains and refer to the published literatures, we write this review to neaten and elucidate the possible mechanism of LLLT in the treatment of AGA. A review of published literature which is associated with keywords LLLT, photobiomodulation, AGA, treatment, hair growth, and mechanism was performed to elucidate the proposed mechanism of LLLT in the treatment of AGA. The present study shows that LLLT can accelerate hair growth in AGA patients. The proposed mechanism of LLLT in treating AGA may vary among different specialists. But we can summarize the consensual mechanisms as follows; low-level light absorbed by chromophores can lead to the production of nitric oxide (NO) and the modulation of reactive oxygen species (ROS). These mobilized molecules subsequently activate redox-related signaling pathways in hair follicle cells and perifollicular cells. Finally, these activated cells participate in the regrowth of hair follicle. Even though the efficacy of LLLT in the treatment of AGA in both men and women has already been confirmed, the present studies focusing on discovering LLLT are still inadequate and unsystematic. More studies are needed to standardize the optimum treatment parameters applied in promoting hair growth and determine the long-term safety and efficacy of LLLT. Current recognitions about the mechanisms of LLLT, mainly focused on the molecules that may take effect, neglected different cellular components that are functional in the hair follicle macro-environment.


Subject(s)
Alopecia/radiotherapy , Low-Level Light Therapy , Alopecia/metabolism , Female , Humans , Ovarian Follicle/metabolism , Ovarian Follicle/pathology , Ovarian Follicle/radiation effects , Reactive Oxygen Species/metabolism , Treatment Outcome
8.
Prog Biophys Mol Biol ; 145: 10-18, 2019 08.
Article in English | MEDLINE | ID: mdl-30500339

ABSTRACT

It is known that LLLT has beneficial effects on several pathological conditions including wound healing, pain and inflammation. LLLT modulates biological processes, including cell proliferation, apoptosis and angiogenesis. In the present study, we examined the effect of local application of LLLT on follicular dynamics, ovarian reserve, AMH expression, progesterone levels, apoptosis, angiogenesis, and reproductive outcome in adult mice. LLLT (200 J/cm2) increased the percentage of primary and preantral follicles, whilst decreasing the percentage of corpora lutea compared to control ovaries. LLLT-treated ovaries did not exhibit any changes regarding the number of primordial follicles. We observed a higher percentage of AMH-positive follicles (in early stages of development) in LLLT-treated ovaries compared to control ovaries. LLLT reduced the P4 concentration and the apoptosis in early antral follicles compared to control ones. LLLT caused a reduction in the endothelial cell area and an increase in the periendothelial cell area in the ovary. Additionally, LLLT was able to improve oocyte quality. Our findings suggest that local application of LLLT modulates follicular dynamics by regulating apoptosis and the vascular stability in mouse ovary. In conclusion, these data indicate that LLLT might become a novel and useful tool in the treatment of several pathologies, including female reproductive disorders.


Subject(s)
Anti-Mullerian Hormone/biosynthesis , Apoptosis/radiation effects , Low-Level Light Therapy , Neovascularization, Physiologic/radiation effects , Ovary/radiation effects , Animals , Cell Line , Cell Proliferation/radiation effects , Corpus Luteum/radiation effects , Female , Fertilization in Vitro/radiation effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovarian Follicle/cytology , Ovarian Follicle/radiation effects , Ovary/blood supply , Ovary/cytology , Ovary/metabolism , Progesterone/biosynthesis , Superovulation/radiation effects
9.
Reproduction ; 155(6): 553-562, 2018 06.
Article in English | MEDLINE | ID: mdl-29636407

ABSTRACT

Radiation damage due to total body irradiation (TBI) or targeted abdominal radiation can deplete ovarian follicles and accelerate reproductive aging. We characterized a mouse model of low-dose TBI to investigate how radiation affects the follicular and stromal compartments of the ovary. A single TBI dose of either 0.1 Gy or 1 Gy (Cesium-137 γ) was delivered to reproductively adult CD1 female mice, and sham-treated mice served as controls. Mice were euthanized either 2 weeks or 5 weeks post exposure, and ovarian tissue was harvested. To assess the ovarian reserve, we classified and counted the number of morphologically normal follicles in ovarian histologic sections for all experimental cohorts using an objective method based on immunohistochemistry for an oocyte-specific protein (MSY2). 0.1 Gy did not affect that total number of ovarian follicles, whereas 1 Gy resulted in a dramatic loss. At two weeks, there was a significant reduction in all preantral follicles, but early antral and antral follicles were still present. By five weeks, there was complete depletion of all follicle classes. We examined stromal quality using histologic stains to visualize ovarian architecture and fibrosis and by immunohistochemistry and quantitative microscopy to assess cell proliferation, cell death and vasculature. There were no differences in the ovarian stroma across cohorts with respect to these markers, indicating that this compartment is more radio-resistant relative to the germ cells. These findings have implications for reproductive health and the field of fertility preservation because the radiation doses we examined mimic scatter doses experienced in typical therapeutic regimens.


Subject(s)
Ovarian Follicle/pathology , Stromal Cells/cytology , Whole-Body Irradiation , Animals , Cells, Cultured , Female , Mice , Ovarian Follicle/radiation effects , Stromal Cells/radiation effects
10.
Gen Comp Endocrinol ; 260: 90-99, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29317212

ABSTRACT

The early stages of ovarian folliculogenesis generally progress independent of gonadotropins, whereas later stages require signaling initiated by FSH. In Siberian hamsters, cycles of folliculogenesis are mediated by changes in photoperiod which depress the hypothalamic pituitary gonadal axis. Reduced gonadotropins lead to decreases in mature follicle development and ovulation; however, early stages of folliculogenesis have not been explored in regressed ovaries. We hypothesized that intraovarian factors that contribute predominantly to later stages of folliculogenesis would react to changes in photoperiod, whereas factors contributing to earlier stages would not change. To probe if the early stages of folliculogenesis continue in the photoinhibited ovary while late stages decline, we measured the mRNA abundance of factors that interact with FSH signaling (Fshr, Igf1, Cox2) and factors that can function independently of FSH (c-Kit, Kitl, Foxo3, Figla, Nobox, Sohlh1, Lhx8). While plasma FSH, antral follicles, and corpora lutea numbers declined with exposure to inhibitory photoperiod, the numbers of primordial, primary, and secondary follicles did not change. Expression of factors that interact with FSH signaling changed with changes in photoperiod; however, expression of factors that do not interact with FSH were not significantly altered. These results suggest that the photoinhibited ovary is not completely quiescent, as factors important for follicle selection and early follicle growth are still expressed in regressed ovaries. Instead, the lack of gonadotropin support that characterizes the non-breeding season appears to inhibit only final stages of folliculogenesis in Siberian hamsters.


Subject(s)
Anovulation/genetics , Follicle Stimulating Hormone/metabolism , Ovarian Follicle/physiology , Ovulation/genetics , Photoperiod , Transcriptome , Animals , Anovulation/metabolism , Cricetinae , Female , Follicle Stimulating Hormone/genetics , Gene Expression Profiling , Gonadotropins/genetics , Gonadotropins/metabolism , Light , Ovarian Follicle/metabolism , Ovarian Follicle/radiation effects , Ovary/metabolism , Ovary/radiation effects , Ovulation/radiation effects , Phodopus , RNA, Messenger/genetics , Receptors, FSH/genetics , Receptors, FSH/metabolism , Seasons , Signal Transduction/genetics , Signal Transduction/radiation effects , Time Factors , Transcriptome/radiation effects
11.
Lasers Med Sci ; 32(9): 1971-1979, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28801854

ABSTRACT

Folliculogenesis is a cycle that produces the majority of oocyte. Any disruption to this cycle leads to ovulation diseases, like polycystic ovarian syndrome (PCOS). Treatments include drugs and surgery; lasers have also been used complementarily. Meanwhile, still there is no definite treatment for PCOS. This study investigated the photo-bio stimulation effect of near-infrared and red low-level laser on producing follicles and compared the result with result of using common drug, clomiphene. Therefore, the aim of this study was to propose the use of lasers autonomously treatment. So, there was one question: how do lasers affect folliculogenesis cycle in rat's ovary tissue? In this study, 28 rats were assigned to four groups as follows: control (CT), clomiphene drug (D), red laser (RL), and near-infrared laser (NIRL). Afterwards, 14 rats of RL and NIRL groups received laser on the first 2 days of estrous cycle, each 6 days, for 48 days. During treatment period, each rat received energy density of 5 J/cm2. Seven rats in D group received clomiphene. After the experiment, lasers' effects at two wavelengths of 630 and 810 nm groups have been investigated and compared with clomiphene and CT groups. Producing different follicles to complement folliculogenesis cycle increased in NIRL and RL groups, but this increase was significant only in the NIRL group. This indicates that NIRL increases ovarian activity to produce oocyte that certainly can be used in future studies for finding a cure to ovarian negligence to produce more oocyte and treat diseases caused by it like PCOS.


Subject(s)
Clomiphene/pharmacology , Infrared Rays , Low-Level Light Therapy , Ovarian Follicle/drug effects , Ovarian Follicle/radiation effects , Animals , Female , Fertility Agents, Female/pharmacology , Hormones/pharmacology , Ovarian Follicle/pathology , Ovarian Follicle/physiology , Rats, Wistar
12.
Biomed Res Int ; 2017: 4176170, 2017.
Article in English | MEDLINE | ID: mdl-28607932

ABSTRACT

Ionizing radiation may cause irreversible ovarian failure, which, therefore, calls for an effective radioprotective reagent. The aim of the present study was to evaluate the potential radioprotective effect of N-acetylcysteine (NAC) on ionizing radiation induced ovarian failure and loss of ovarian reserve in mice. Kun-Ming mice were either exposed to X-irradiation (4 Gy), once, and/or treated with NAC (300 mg/kg), once daily for 7 days before X-irradiation. We examined the serum circulating hormone levels and the development of ovarian follicles as well as apoptosis, cell proliferation, and oxidative stress 24 hours after X-irradiation. In addition, morphological observations on the endometrial luminal epithelium and the fertility assessment were performed. We found that NAC successfully restored the ovarian and uterine function, enhanced the embryo implantation, improved the follicle development, and altered the abnormal hormone levels through reducing the oxidative stress and apoptosis level in granulosa cells while promoting the proliferation of granulosa cells. In conclusion, the radioprotective effect of NAC on mice ovary from X-irradiation was assessed, and our results suggested that NAC can be a potential radioprotector which is capable of preventing the ovarian failure occurrence and restoring the ovarian reserve.


Subject(s)
Acetylcysteine/administration & dosage , Ovarian Follicle/physiopathology , Primary Ovarian Insufficiency/drug therapy , Radiation-Protective Agents/administration & dosage , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Disease Models, Animal , Embryo Implantation/drug effects , Embryo Implantation/radiation effects , Endometrium/drug effects , Endometrium/radiation effects , Female , Humans , Mice , Ovarian Follicle/drug effects , Ovarian Follicle/radiation effects , Ovarian Reserve/drug effects , Ovarian Reserve/radiation effects , Primary Ovarian Insufficiency/physiopathology , Radiation, Ionizing
13.
Hum Reprod ; 31(8): 1816-26, 2016 08.
Article in English | MEDLINE | ID: mdl-27251203

ABSTRACT

STUDY QUESTION: Do charged iron particles, components of space radiation, cause premature ovarian failure? SUMMARY ANSWER: Exposure to charged iron particles causes ovarian DNA damage, oxidative damage and apoptosis, resulting in premature ovarian failure. WHAT IS KNOWN ALREADY: The ovary is very sensitive to follicle destruction by low linear energy transfer (LET) radiation, such as X-rays and γ-rays. However, it is completely unknown whether high-LET radiation, such as charged iron particles, also destroys ovarian follicles. STUDY DESIGN, SIZE, DURATION: Twelve week old C57BL/6J female mice were exposed to single doses of 0, 5, 30 or 50 cGy (n = 8/group) charged iron particles (LET = 179 keV/µm) at energy of 600 MeV/u. Two groups were irradiated at the highest dose, one fed AIN-93M chow and the other fed AIN-93M chow supplemented with 150 mg/kg diet alpha lipoic acid (ALA). PARTICIPANTS/MATERIALS, SETTING, METHODS: We quantified the numbers of ovarian follicles, measured serum follicle stimulating hormone (FSH) and luteinizing hormone (LH) concentrations, and analyzed histone H2AX phosphorylation, oxidative damage and apoptosis markers in the ovarian follicles. MAIN RESULTS AND THE ROLE OF CHANCE: H2AX phosphorylation, lipid peroxidation, protein nitration and apoptosis were highly induced in ovarian follicles at 6 h and remained increased 1 week after irradiation. As a result, numbers of healthy ovarian follicles were significantly and dose-dependently depleted at 1 and 8 weeks post-irradiation, with 57, 84 and 99% decreases in primordial follicles at 8 weeks at the 5, 30 and 50 cGy doses, respectively (P < 0.05 versus 0 cGy). Consistent with near-total depletion of ovarian follicles in the 50 cGy group, serum concentrations of FSH and LH were significantly elevated at 8 weeks. Dietary supplementation with ALA partially prevented the adverse ovarian effects of 50 cGy iron particles. LIMITATIONS, REASONS FOR CAUTION: About 21% of the estimated radiation dose from exposure to galactic cosmic rays during a multi-year Mars mission will be due to high-LET particles, of which iron is only one. The effects of galactic cosmic rays, which contain a mixture of multiple charged particles, as well as protons, neutrons, and helium ions, may differ from the effects of iron alone. WIDER IMPLICATIONS OF THE FINDINGS: We show for the first time that charged high-LET ions are highly damaging to the ovary even at low doses, causing premature ovarian failure. In addition to raising concerns for female astronauts, these findings raise concerns for ovarian damage due to clinical uses of high-LET particles for cancer treatment. In addition to causing infertility, premature ovarian failure has adverse implications for the functions of heart, brain, bone and muscle later in life. STUDY FUNDING/COMPETING INTERESTS: This work was supported by a National Aeronautics and Space Administration grant NNX14AC50G to U.L. B.M. was partially supported by a National Space Biomedical Research Institute First Award, PF04302. Additional support was received from the University of California Irvine Center for Occupational and Environmental Health. The authors have no conflicts of interests.


Subject(s)
Cosmic Radiation , Iron , Ovarian Follicle/radiation effects , Oxidative Stress/radiation effects , Thioctic Acid/administration & dosage , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Female , Follicle Stimulating Hormone/blood , Histones/metabolism , Lipid Peroxidation/drug effects , Lipid Peroxidation/radiation effects , Luteinizing Hormone/blood , Mice , Mice, Inbred C57BL , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Oxidative Stress/drug effects , Phosphorylation/drug effects , Phosphorylation/radiation effects , Radiation Dosage
14.
Int J Radiat Biol ; 92(6): 329-37, 2016 06.
Article in English | MEDLINE | ID: mdl-27007703

ABSTRACT

The effects on human health of electromagnetic field (EMF) have begun to be seriously questioned with the entry into daily life of devices establishing EMF, such as cell phones, wireless fidelity, and masts. Recent studies have reported that exposure to EMF, particularly during pregnancy, affects the developing embryo/fetus. The aim of this study was therefore to examine the effects of exposure to continuous 900-Megahertz (MHz) EMF applied in the prenatal period on ovarian follicle development and oocyte differentiation. Six pregnant Sprague Dawley rats were divided equally into a non-exposed control group (CNGr) and a group (EMFGr) exposed to continuous 900-MHz EMF for 1 h daily, at the same time every day, on days 13-21 of pregnancy. New groups were established from pups obtained from both groups after birth. One group consisting of female pups from CNGr rats was adopted as newborn CNGr (New-CNGr, n = 6), and another group consisting of female pups from EMFGr rats was adopted as newborn EMFGr (New-EMFGr, n = 6). No procedure was performed on New-CNGr or New-EMFGr rats. All rat pups were sacrificed on the postnatal 34th day, and their ovarian tissues were removed. Follicle count, histological injury scoring and morphological assessment with apoptotic index criteria were performed with sections obtained following routine histological tissue preparation. Follicle count results revealed a statistically significant decrease in primordial and tertiary follicle numbers in New-EMFGr compared to New-CNGr (p < 0.05), while atretic follicle numbers and apoptotic index levels increased significantly (p < 0.05). Histopathological examination revealed severe follicle degeneration, vasocongestion, a low level of increased stromal fibrotic tissue and cytoplasmic vacuolization in granulosa cell in New-EMFGr. Prenatal exposure to continuous 900-MHz EMF for 1 h each day from days 13-21 led to a decrease in ovarian follicle reservoirs in female rat pups at the beginning of the prepubertal period.


Subject(s)
Electromagnetic Fields/adverse effects , Ovarian Follicle/pathology , Ovarian Follicle/radiation effects , Ovarian Reserve/radiation effects , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/pathology , Aging/pathology , Aging/radiation effects , Animals , Dose-Response Relationship, Radiation , Female , Microwaves/adverse effects , Ovarian Follicle/physiopathology , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Radiation Dosage , Rats , Rats, Sprague-Dawley
15.
J Adolesc Young Adult Oncol ; 4(4): 174-83, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26697267

ABSTRACT

PURPOSE: Fertility preservation in a pediatric and teen female population is challenging because standard technologies of egg and embryo freezing may not be possible due to premenarcheal status. Ovarian tissue cryopreservation (OTC) with the intent of future ovarian tissue transplantation or in vitro follicle growth may be the only option to preserve fertility. The purpose of this study was to add to the general understanding of primordial follicle dynamics in young patients. METHODS: First, the unique infrastructure of the Oncofertility Consortium National Physicians Cooperative (OC-NPC) is described, which simultaneously drives clinical fertility preservation and basic research to explore and expand the reproductive options for those in need. Then, the OC-NPC research resource is used to perform a histological evaluation of ovarian tissue from 24 participants younger than 18 years of age. RESULTS: Primordial follicles, which comprise the ovarian reserve, were observed in all participant tissues, irrespective of variables, including age, diagnosis, previous treatment history, tissue size, and tissue processing methods. Primordial follicles were present in ovarian tissue, even in participants who had a previous history of exposure to chemotherapy and/or radiation treatment regimens, which placed them at risk for iatrogenic infertility or premature ovarian failure. CONCLUSION: Primordial follicles were observed in ovarian tissue from all participants examined, despite population and tissue heterogeneity. These results increase the understanding of human follicle dynamics and support OTC as a promising fertility preservation modality in the young female population. Future studies to evaluate follicle quality within these tissues are warranted.


Subject(s)
Cryopreservation/methods , Fertility Preservation/methods , Ovarian Follicle/anatomy & histology , Adolescent , Age Factors , Antineoplastic Agents/pharmacology , Child , Child, Preschool , Female , Humans , Infant , Neoplasms/drug therapy , Neoplasms/radiotherapy , Ovarian Follicle/drug effects , Ovarian Follicle/pathology , Ovarian Follicle/radiation effects , Specimen Handling/methods
16.
PLoS One ; 10(10): e0140055, 2015.
Article in English | MEDLINE | ID: mdl-26465611

ABSTRACT

Radiotherapy is one of the standard cytotoxic therapies for cancer. However, it has a profound impact on ovarian function leading to premature ovarian failure and infertility. Since none of the currently available methods for fertility preservation guarantees future fertility, the need for an effective radioprotective agent is highly intensified. The present study investigated the mechanisms of the potential radioprotective effect of growth hormone (GH) on γ irradiation-induced ovarian failure and the impact of the insulin like growth factor 1 (IGF-1) in the underlying protection. Immature female Sprague-Dawley rats were either exposed to single whole body irradiation (3.2 Gy) and/or treated with GH (1 mg/kg s.c). Experimental γ-irradiation produced an array of ovarian dysfunction that was evident by assessment of hormonal changes, follicular development, proliferation marker (PCNA), oxidative stress as well as apoptotic markers. In addition, IGF-1/IGF-1R axis expression was assessed using real-time PCR and immunolocalization techniques. Furthermore, after full maturity, fertility assessment was performed. GH significantly enhanced follicular development and restored anti-Mullerian hormone serum level as compared with the irradiated group. In addition, GH significantly ameliorated the deleterious effects of irradiation on oxidative status, PCNA and apoptosis. Interestingly, GH was shown to enhance the ovarian IGF-1 at transcription and translation levels, a property that contributes significantly to its radioprotective effect. Finally, GH regained the fertility that was lost following irradiation. In conclusion, GH showed a radioprotective effect and rescued the ovarian reserve through increasing local IGF-1 level and counteracting the oxidative stress-mediated apoptosis.


Subject(s)
Growth Hormone/pharmacology , Ovarian Follicle/drug effects , Ovarian Follicle/radiation effects , Radiation Injuries , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Biomarkers , Body Weight , Female , Gamma Rays/adverse effects , Human Growth Hormone/pharmacology , Humans , Insulin-Like Growth Factor I/metabolism , Male , Organ Size , Ovarian Follicle/metabolism , Ovarian Follicle/pathology , Ovary/drug effects , Ovary/metabolism , Ovary/pathology , Ovary/radiation effects , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Rats , Receptor, IGF Type 1/metabolism
17.
Arq Bras Endocrinol Metabol ; 58(2): 132-43, 2014 Mar.
Article in Portuguese | MEDLINE | ID: mdl-24830590

ABSTRACT

This article is a review on different aspects of premature ovarian failure (POF) defined as the development of hypogonadism in women before 40 years of age. The review will discuss the etiopathogeny, autoimmune and iatrogenic causes, abnormalities of chromosome X, as well as clinical manifestations, diagnosis, and treatment. Most of the women with this disorder do not have menstrual history, specific of POF development, but infertility associated with the diagnosis is the most problematic aspect of the disease.


Subject(s)
Chromosome Aberrations , Primary Ovarian Insufficiency , Adult , Chromosomes, Human, X/genetics , Diagnosis, Differential , Female , Fragile X Mental Retardation Protein/genetics , Humans , Hypogonadism/etiology , Incidence , Infertility, Female/etiology , Infertility, Female/therapy , Middle Aged , Ovarian Follicle/drug effects , Ovarian Follicle/radiation effects , Ovary/diagnostic imaging , Primary Ovarian Insufficiency/drug therapy , Primary Ovarian Insufficiency/epidemiology , Primary Ovarian Insufficiency/etiology , Ultrasonography
18.
Indian J Exp Biol ; 52(5): 496-503, 2014 May.
Article in English | MEDLINE | ID: mdl-24851412

ABSTRACT

Resonance experiment was employed to investigate the mechanism of photoperiodic time measurement during initiation of ovarian growth and functions in the subtropical population of female tree sparrow (Passer montanus) at Shillong (Latitude 25 degrees 34 'N, Longitude 91 degrees 53 'E). Photosensitive birds were subjected to various resonance light dark cycles of different durations such as: 12-(6L:6D), 24-(6L:18D), 36-(6L:30D), 48-(6L:42D), 60-(6L:54D) and 72-(6L:66D) h along with a control group under long days (14L:10D) for 35 days. Birds, exposed to long days, exhibited ovarian growth confirming their photosensitivity at the beginning of the experiment. The birds experiencing resonance light/dark cycles of 12, 36 and 60 h responded well while those exposed to 24, 48 and 72 h cycles did not. Serum levels of estradiol-17beta ran almost parallel to changes in the follicular size. Further, histomorphometric analyses of ovaries of the birds subjected to various resonance light dark cycles revealed distinct correlation with the ovarian growth and the serum levels of estradiol-17beta. No significant change in body weight was observed in the birds under any of the light regimes. The results are in agreement with the avian external coincidence model of photoperiodic time measurement and indicate that an endogenous circadian rhythm is involved during the initiation of the gonadal growth and functions in the female tree sparrow.


Subject(s)
Circadian Rhythm/physiology , Ovarian Follicle/physiology , Photoperiod , Sparrows/physiology , Animals , Estradiol/blood , Female , Ovarian Follicle/radiation effects , Sparrows/blood
19.
Ultrasound Med Biol ; 40(7): 1535-44, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24785439

ABSTRACT

One of the challenges in ovarian transplantation is ischemia-reperfusion damage. When transitional tissue faces an acute and critical condition in terms of blood supply (immediately after organ transplantation), treatment with low-intensity pulsed ultrasound (LIPUS) seems to be very beneficial. The aim of this study was to evaluate the effects of ultrasound therapy on heterotopic transplanted mouse ovarian tissue. Adult female Naval Medical Research Institute mice were divided into three groups. In the experimental groups, the transplanted ovary was exposed 5 min daily to ultrasound with an intensity of 0.3 W/cm(2), frequency of 3 MHz and pulse mode of 1:4. The grafted ovaries were assessed with the usual histology and immunohistochemistry techniques. Results indicate that more CD31 angiogenic factor was expressed in irradiated animals than in control animals, and ultrasound therapy resulted in better follicular preservation, especially after 14 d. In conclusion, therapeutic ultrasound may accelerate and increase re-angiogenesis and can help to promote ovarian follicular growth.


Subject(s)
Apoptosis/radiation effects , Neovascularization, Physiologic/radiation effects , Ovarian Follicle/growth & development , Ovarian Follicle/radiation effects , Ovary/transplantation , Ultrasonic Therapy/methods , Animals , Female , High-Energy Shock Waves , Mice , Ovarian Follicle/cytology , Treatment Outcome
20.
PLoS One ; 9(4): e93311, 2014.
Article in English | MEDLINE | ID: mdl-24691397

ABSTRACT

Exposure to radiation during fetal development induces testicular germ cell tumors (TGCT) and reduces spermatogenesis in mice. However, whether DNA damaging chemotherapeutic agents elicit these effects in mice remains unclear. Among such agents, cyclophosphamide (CP) is currently used to treat breast cancer in pregnant women, and the effects of fetal exposure to this drug manifested in the offspring must be better understood to offer such patients suitable counseling. The present study was designed to determine whether fetal exposure to CP induces testicular cancer and/or gonadal toxicity in 129 and in 129.MOLF congenic (L1) mice. Exposure to CP on embryonic days 10.5 and 11.5 dramatically increased TGCT incidence to 28% in offspring of 129 mice (control value, 2%) and to 80% in the male offspring of L1 (control value 33%). These increases are similar to those observed in both lines of mice by radiation. In utero exposure to CP also significantly reduced testis weights at 4 weeks of age to ∼ 70% of control and induced atrophic seminiferous tubules in ∼ 30% of the testes. When the in utero CP-exposed 129 mice reached adulthood, there were significant reductions in testicular and epididymal sperm counts to 62% and 70%, respectively, of controls. In female offspring, CP caused the loss of 77% of primordial follicles and increased follicle growth activation. The results indicate that i) DNA damage is a common mechanism leading to induction of testicular cancer, ii) increased induction of testis cancer by external agents is proportional to the spontaneous incidence due to inherent genetic susceptibility, and iii) children exposed to radiation or DNA damaging chemotherapeutic agents in utero may have increased risks of developing testis cancer and having reduced spermatogenic potential or diminished reproductive lifespan.


Subject(s)
Antineoplastic Agents, Alkylating/adverse effects , Cyclophosphamide/adverse effects , Maternal Exposure , Ovarian Follicle/drug effects , Prenatal Exposure Delayed Effects , Spermatogenesis/drug effects , Testicular Neoplasms/etiology , Animals , Disease Models, Animal , Female , Male , Mice , Ovarian Follicle/radiation effects , Ovary/drug effects , Ovary/metabolism , Ovary/pathology , Ovary/radiation effects , Pregnancy , Sperm Count , Spermatogenesis/radiation effects , Testis/drug effects , Testis/metabolism , Testis/pathology , Testis/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...