Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 352
Filter
1.
Radiat Res ; 200(4): 321-330, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37702414

ABSTRACT

Testicular injury is a well-documented acute effect of radiation exposure, though little is known about recovery years after irradiation, especially at higher doses. We examined the testes from 143 irradiated and control male rhesus monkeys, who were part of the Radiation Late Effects Cohort over a four-year period. Irradiated animals were exposed to doses ranging from 3.5 to 8.5 Gy of total-body irradiation. The testes were assessed using computed tomography (CT) volumetry, serum testosterone, and histology for deceased members of the cohort. Irradiated animals exhibited dose-dependent testicular atrophy as well as decreased serum testosterone during the winter breeding season when compared to age-matched unirradiated controls. No significant difference in summer testosterone levels was observed. Volumetric and histologic evidence of testicular recovery was present approximately three years postirradiation for animals who received ≤8 Gy. The study demonstrates dose-dependent testicular injury after total-body irradiation and provides evidence for volumetric and spermatogonial recovery even at lethal doses of total-body irradiation in rhesus monkeys.


Subject(s)
Spermatogonia , Testis , Humans , Animals , Male , Macaca mulatta , Testis/radiation effects , Spermatogonia/radiation effects , Dose-Response Relationship, Radiation , Testosterone
2.
Int J Mol Sci ; 22(23)2021 Nov 27.
Article in English | MEDLINE | ID: mdl-34884637

ABSTRACT

The adverse effects of radiation are proportional to the total dose and dose rate. We aimed to investigate the effects of radiation dose rate on different organs in mice. The mice were subjected to low dose rate (LDR, ~3.4 mGy/h) and high dose rate (HDR, ~51 Gy/h) radiation. LDR radiation caused severe tissue toxicity, as observed in the histological analysis of testis. It adversely influenced sperm production, including sperm count and motility, and induced greater sperm abnormalities. The expression of markers of early stage spermatogonial stem cells, such as Plzf, c-Kit, and Oct4, decreased significantly after LDR irradiation, compared to that following exposure of HDR radiation, in qPCR analysis. The compositional ratios of all stages of spermatogonia and meiotic cells, except round spermatid, were considerably reduced by LDR in FACS analysis. Therefore, LDR radiation caused more adverse testicular damage than that by HDR radiation, contrary to the response observed in other organs. Therefore, the dose rate of radiation may have differential effects, depending on the organ; it is necessary to evaluate the effect of radiation in terms of radiation dose, dose rate, organ type, and other conditions.


Subject(s)
Spermatogenesis/radiation effects , Testis/radiation effects , Animals , Dose-Response Relationship, Radiation , Gamma Rays , Male , Mice , Models, Animal , Radiation Dosage , Spermatids/cytology , Spermatids/radiation effects , Spermatogonia/cytology , Spermatogonia/radiation effects , Spermatozoa/cytology , Spermatozoa/radiation effects , Testis/cytology
3.
Biomed Environ Sci ; 34(10): 789-802, 2021 Oct 20.
Article in English | MEDLINE | ID: mdl-34782045

ABSTRACT

OBJECTIVE: The expression patterns of ribosomal large subunit protein 23a (RPL23a) in mouse testes and GC-1 cells were analyzed to investigate the potential relationship between RPL23a expression and spermatogonia apoptosis upon exposure to X-ray. METHODS: Male mice and GC-1 cells were irradiated with X-ray, terminal dUTP nick end-labelling (TUNEL) was performed to detect apoptotic spermatogonia in vivo. Apoptotic rate and cell cycle phase of GC-1 cells were analyzed with flow cytometry. Protein interactions were detected by Immunoprecipitation and protein localization as studied by immunofluorescence. Immunoblotting and real-time PCR were applied to analyze to protein and gene expression. RESULTS: Ionizing radiation (IR) increased spermatogonia apoptosis, the expression of RPL11, MDM2 and p53, and decreased RPL23a expression in mice spermatogonia in vivo and in vitro. RPL23a knockdown weakened the interaction between RPL23a and RPL11, leading to p53 accumulation. Moreover, knockdown and IR decreased RPL23a that induces spermatogonia apoptosis via RPL23a-RPL11-MDM2-p53 pathway in GC-1 cells. CONCLUSION: These results suggested that IR reduced RPL23a expression, leading to weakened the RPL23a-RPL11 interactions, which may have activated p53, resulting in spermatogonia apoptosis. These results provide insights into environmental and clinical risks of radiotherapy following exposure to IR in male fertility. The graphical abstract was available in the web of www.besjournal.com.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation , Ribosomal Proteins/genetics , Spermatogonia/radiation effects , Animals , Male , Mice , Ribosomal Proteins/metabolism , Signal Transduction , Spermatogonia/metabolism
4.
Article in English | WPRIM (Western Pacific) | ID: wpr-921331

ABSTRACT

Objective@#The expression patterns of ribosomal large subunit protein 23a (RPL23a) in mouse testes and GC-1 cells were analyzed to investigate the potential relationship between RPL23a expression and spermatogonia apoptosis upon exposure to X-ray.@*Methods@#Male mice and GC-1 cells were irradiated with X-ray, terminal dUTP nick end-labelling (TUNEL) was performed to detect apoptotic spermatogonia @*Results@#Ionizing radiation (IR) increased spermatogonia apoptosis, the expression of RPL11, MDM2 and p53, and decreased RPL23a expression in mice spermatogonia @*Conclusion@#These results suggested that IR reduced RPL23a expression, leading to weakened the RPL23a-RPL11 interactions, which may have activated p53, resulting in spermatogonia apoptosis. These results provide insights into environmental and clinical risks of radiotherapy following exposure to IR in male fertility. The graphical abstract was available in the web of www.besjournal.com.


Subject(s)
Animals , Male , Mice , Apoptosis/genetics , Gene Expression Regulation , Ribosomal Proteins/metabolism , Signal Transduction , Spermatogonia/radiation effects
5.
Ecotoxicol Environ Saf ; 201: 110831, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32535367

ABSTRACT

The risk of exposure to ionizing radiation (IR) environments has increased with the development of nuclear technology. IR exposure induces excessive apoptosis of the spermatogonia, which leads to male infertility. Spermatogonia apoptosis may be involved in ribosomal stress triggered by DNA damage following exposure to IR because ribosomal proteins (RPs) directly interact with mouse double minute 2 homolog (MDM2) to induce apoptosis. This study aimed to use comparative proteomics and transcriptomics approach to screen the differential RPs and ribosomal mRNAs in mouse testes following high linear energy transfer (LET) carbon ion radiation (CIR). The expression of ribosomal large subunit protein 27a (Rpl27a) decreased at both protein and mRNA levels in the spermatogonia in vivo. After 6 h of CIR, the immunofluorescence signal of 8-oxo-dG and phosphorylated ataxia-telangiectasia-mutated protein (ATM)/histone H2Ax increased, but that of Rpl27a decreased in the spermatogonia of p53 wild-type and knockout mouse testes. Moreover, the nucleolin was scattered throughout the nucleoplasm after CIR. These results suggested that CIR-induced DNA damage might trigger ribosomal stress, and the reduction in the expression of Rpl27a was associated with DNA damage in the spermatogonia. Similarly, in vitro, the immunofluorescence signal of 8-oxo-dG increased in the GC-1 cells after CIR. Moreover, the expression of Rpl27a was regulated by DNA damage because the co-transfection of ATM and Rpl27a or inhibition of ATM-treated CIR could restore the expression of Rpl27a. Furthermore, the reduction in the expression of Rpl27a led to weakened binding of E2F transcription factor 1 (E2F1) and p53 to MDM2, causing p53 activation and E2F1 degradation in p53 wild-type and knockdown GC-1 cells. This study proposed that heavy ion radiation-induced DNA damage mediated spermatogonia apoptosis via the Rpl27a-Rpl5-MDM2-p53/E2F1 signaling pathway. The results provided the underlying molecular mechanisms of spermatogonia apoptosis following exposure to high LET radiation.


Subject(s)
Apoptosis/radiation effects , DNA Damage , Proto-Oncogene Proteins c-mdm2/metabolism , Radiation, Ionizing , Ribosomal Proteins/metabolism , Spermatogonia/radiation effects , Animals , Apoptosis/genetics , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Heavy Ions , Humans , Male , Mice , Proto-Oncogene Proteins c-mdm2/genetics , Ribosomal Proteins/genetics , Signal Transduction , Spermatogonia/metabolism , Spermatogonia/pathology , Tumor Suppressor Protein p53/genetics
6.
Sci Rep ; 10(1): 37, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31913321

ABSTRACT

The exposure of germ cells to radiation introduces mutations in the genomes of offspring, and a previous whole-genome sequencing study indicated that the irradiation of mouse sperm induces insertions/deletions (indels) and multisite mutations (clustered single nucleotide variants and indels). However, the current knowledge on the mutation spectra is limited, and the effects of radiation exposure on germ cells at stages other than the sperm stage remain unknown. Here, we performed whole-genome sequencing experiments to investigate the exposure of spermatogonia and mature oocytes. We compared de novo mutations in a total of 24 F1 mice conceived before and after the irradiation of their parents. The results indicated that radiation exposure, 4 Gy of gamma rays, induced 9.6 indels and 2.5 multisite mutations in spermatogonia and 4.7 indels and 3.1 multisite mutations in mature oocytes in the autosomal regions of each F1 individual. Notably, we found two types of deletions, namely, small deletions (mainly 1~12 nucleotides) in non-repeat sequences, many of which showed microhomology at the breakpoint junction, and single-nucleotide deletions in mononucleotide repeat sequences. The results suggest that these deletions and multisite mutations could be a typical signature of mutations induced by parental irradiation in mammals.


Subject(s)
Genome , Mutation , Oocytes/physiology , Spermatogonia/physiology , Animals , Animals, Newborn , Female , Gamma Rays , Male , Mice , Mice, Inbred C57BL , Mutation Rate , Oocytes/radiation effects , Radiation Effects , Radiation, Ionizing , Spermatogonia/radiation effects , Whole Genome Sequencing
7.
Mol Cells ; 42(11): 794-803, 2019 Nov 30.
Article in English | MEDLINE | ID: mdl-31697875

ABSTRACT

Ultraviolet light (UV)-induced cellular response has been studied by numerous investigators for many years. Long noncoding RNAs (lncRNAs) are emerging as new regulators of diverse cellular process; however, little is known about the role of lncRNAs in the cellular response to UV treatment. Here, we demonstrate that levels of lncRNA-HOTTIP significantly increases after UV stimulation and regulates the UV-mediated cellular response to UV through the coordinate activation of its neighboring gene Hoxa13 in GC-1 cells (spermatogonia germ cell line). UV-induced, G2/M-phase arrest and early apoptosis can be regulated by lncRNA-HOTTIP and Hoxa13. Furthermore, lncRNA-HOTTIP can up-regulate γ-H2AX and p53 expression via Hoxa13 in UV-irradiated GC-1 cells. In addition, p53 has the ability to regulate the expression of both lncRNA-HOTTIP and Hoxa13 in vitro and in vivo. Our results provide new data regarding the role lncRNAs play in the UV response in spermatogenic cells.


Subject(s)
DNA Damage , DNA Repair , RNA, Long Noncoding/genetics , Spermatogonia/radiation effects , Animals , Apoptosis/genetics , Cell Line , G2 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation/radiation effects , Histones/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mice, Inbred C57BL , Spermatogonia/cytology , Spermatogonia/metabolism , Tumor Suppressor Protein p53/genetics , Ultraviolet Rays
8.
Int J Radiat Biol ; 95(10): 1414-1420, 2019 10.
Article in English | MEDLINE | ID: mdl-30648901

ABSTRACT

Purpose: We have proposed a mathematical model (WAM model) expressing increment of the dose-rate dependent mutation frequency caused by artificial radiations. In this model, it is defined that the pool of mutant cells in dynamic equilibrium in organisms. We verified the accuracy of the WAM prediction of mutation frequency in mice. Materials and methods: The theoretical values calculated by the WAM model were compared with the experimental values obtained from the large mouse genetics program at the Oak Ridge National Laboratory (ORNL). Results: Most of all the theoretical values in acute and chronic irradiation conditions nearly coincided with the experimental values. However, the theoretical value of the chronic conditions at the dose-rate of 0.8 R/min was significantly higher than its experimental value. This discordance was able to be minimized in the WAM assumption, when the period from the end of exposure to start mating was two weeks longer. Conclusions: As a result of comparison between experimental and theoretical data, the certainty of the WAM model was confirmed in mice and it was shown that the genetic influence varies depending on the dose-rate.


Subject(s)
Dose-Response Relationship, Radiation , Mutation Rate , Radiation Dosage , Animals , Cell Death , Cell Proliferation/radiation effects , DNA Mutational Analysis , Male , Mice , Models, Genetic , Radiation Protection , Radiobiology/methods , Reproducibility of Results , Spermatogonia/radiation effects
9.
Andrology ; 7(1): 2-7, 2019 01.
Article in English | MEDLINE | ID: mdl-30411532

ABSTRACT

BACKGROUND: Spermatogenesis is a process of dynamic cell differentiation. Ionizing radiation impairs spermatogenesis, and spermatogonia are more radiosensitive than spermatocytes or spermatids. Consistent with this assumption and due to improvement in tumor curability, nowadays, fertility preservation represents a public health need. OBJECTIVES: To discuss radiotherapy-induced risk to male fertility and raise oncologic awareness of male fertility in daily clinical practice. MATERIALS AND METHODS: PubMed and Clinicaltrials.gov databases were searched for papers in English. RESULTS: We provide an overview of clinical landscape. Four main issues were proposed: (i) spermatogenesis and radiobiological general concepts; (ii) impairment of spermatogenesis; (iii) impairment of testosterone-producing Leydig cells; (iv) clinical radiotherapy evidence in oncology. CONCLUSION: This review can be useful in daily clinical work and offer some directions for future research.


Subject(s)
Infertility, Male/etiology , Leydig Cells/radiation effects , Radiation Injuries/pathology , Spermatogenesis/radiation effects , Spermatogonia/radiation effects , Chernobyl Nuclear Accident , Fertility Preservation/methods , Humans , Male , Neoplasms/radiotherapy , Quality of Life/psychology
10.
Vopr Kurortol Fizioter Lech Fiz Kult ; 95(1): 35-40, 2018 Apr 09.
Article in Russian | MEDLINE | ID: mdl-29652044

ABSTRACT

BACKGROUND: it has been demonstrated in various experimental studies that radiation exposure produces a negative impact on the processes of spermatogenesis associated with the disturbances of the microcirculation processes in the testes and the development of cellular and intracellular disintegration expressed as destructive changes in the cells leading to their death. AIM: The objective of the present study was to detect the ultrastructural abnormalities in the cells of Sertoli and spermatogonia under conditions of their exposure to radiation and to identify the peculiarities of their regeneration under the influence of the therapeutic and prophylactic application of low-intensity ultra-high frequency (UHF) electromagnetic radiation (EMR) and low-intensity low-frequency magnetic field (MF). MATERIAL AND METHODS: The experiments were carried out on 28 non-pedigree mature male rats with the body weight 180-220 g that were divided into four groups. The first study group was comprised of the animals exposed to radiation followed by the application of low-intensity ultra-high frequency UHF electromagnetic radiation EMR. The rats in the second study group experienced effects of radiation and low-intensity low-frequency MF. The animals of the third (control) group were exposed to radiation alone, and those comprising the fourth group 1 (only radiation exposure) were considered to be intact. RESULTS: The studies with the use of electron microscopy showed that the therapeutic and prophylactic application of low-intensity ultra-high frequency (UHF) electromagnetic radiation and low-intensity low-frequency magnetic field caused the decrease in the number and the severity of post-radiation defects in the treated cells together with the increase of the number and size of mitochondria as well as hyperplasia of ribosomes; moreover, it promoted cellular and intracellular regeneration. UHF electromagnetic radiation had a more pronounced stimulating effect on the regeneration processes as compared with low-frequency MF. Particularly active processes of intracellular regeneration evolved in Sertoli cells; they were manifested as the increase in the number and size of mitochondria, enhanced hyperplasia of ribosomes, and formation of polysomes and new membranes of the granular endoplasmic reticulum. In spermatogonia, intracellular regeneration was less pronounced than in the Sertoli cells but was accompanied by enhanced cell regeneration and a greater number of reserve stem/progenitor cells. CONCLUSIONS: The results of the present study provide a rationale for the possibility of the application of a low-frequency magnetic field and especially UHF electromagnetic radiation for the further development of the promising therapeutic and preventive technologies with a view to their introduction into routine clinical practice dealing with radiation-induced pathology.


Subject(s)
Electromagnetic Radiation , Sertoli Cells/radiation effects , Spermatogonia/radiation effects , Animals , Male , Rats , Sertoli Cells/ultrastructure , Spermatogonia/ultrastructure
11.
Anim Reprod Sci ; 193: 58-67, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29636209

ABSTRACT

Although germ cells from donor rams transplanted into irradiated recipient testes have produced donor derived offspring, efficiency is low. Further optimization of recipient irradiation protocols will add precision to the depletion of recipient spermatogonia prior to germ cell transplant. Three irradiation doses (9,12,15 Gy) were administered to ram lambs aged 14 weeks (Group 1) and 20 weeks (Group 2), then testicular biopsies were collected 1, 2 and 3 months after irradiation. At 1 month after irradiation of Group 1, only the largest dose (15 Gy) reduced spermatogonia numbers below 10% of non-irradiated controls, whereas in Group 2 lambs, each irradiation dose reduced spermatogonia below 10% of controls. In both Groups, fewer differentiated germ cells were present in seminiferous tubules compared to controls. At 2 months after irradiation, spermatogonia numbers in both Groups increased more than sixfold to be similar to controls, whereas fewer differentiated germ cells were present in the tubules of both Groups. At 3 months in Group 1, each irradiation dose reduced spermatogonia numbers to <30% of controls and fewer tubules contained differentiated germ cells. Lesser expression of spermatogonial genes, VASA and UCHL-1, was observed in the 15 Gy group. In Group 2, only 12 Gy treated tubules contained fewer spermatogonia. Knowledge of these subtle differences between age groups in the effect of irradiation doses on spermatogonia or differentiated germ cell numbers and the duration of recovery of spermatogonia numbers after irradiation will aid the timing of germ cell transplants into prepubertal recipient lambs.


Subject(s)
Aging/physiology , Radiation Tolerance/physiology , Sexual Maturation/physiology , Sheep , Spermatogonia/radiation effects , Age Factors , Animals , Gamma Rays , Gene Expression Regulation/radiation effects , Male , Radiation Dosage , Sexual Maturation/radiation effects , Spermatogenesis/radiation effects , Spermatogonia/physiology , Spermatogonia/transplantation , Testis/cytology , Testis/physiology , Testis/radiation effects , Transplantation Conditioning/methods , Transplantation Conditioning/veterinary
12.
Anim Reprod Sci ; 186: 31-36, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28919176

ABSTRACT

Several methods have been developed to suppress spermatogenesis in recipient males before spermatogonial stem cells (SSCs) transplantation. The aim of this study was to compare two different methods of depleting endogenous spermatogenesis in recipient ROSS 308 strain adult roosters. Gamma-radiation and alkylating agent busulfan were utilized to infertilize adult roosters (ROSS 308 strain). Two radiation therapy regimes (based on 60co isotope) were conducted locally to testes using 40Gy (5×8Gy with three-day intervals) and 30Gy (3×10Gy with three-day intervals). And two different levels of busulfan 60mg(40+20) and 50mg(30+20) with 10-day intervals were injected intraperitoneally. The results showed that both radiation therapy regimes and both busulfan levels reduced sperm motility and sperm concentration significantly compared with control group. Moreover, there were no significant differences between gamma radiation and busulfan treatments in progressive and total motility of sperm reduction. Sperm concentration reached to zero at the end of the 4th week of experiment in all treatment groups. Also histological examinations revealed that both treatments could significantly reduce the diameter of seminiferous tubules and thickness of epithelium. None of the treatments had significant effect on body weight in comparison with control group and the health status of experimental roosters remained good throughout the study. Given that, the risk probability of high doses of radiation exposure and busulfan, it can be concluded that the 30Gy (3×10Gy) and 50mg (30+20) are appropriate for suppression of endogenous spermatogenesis in mature roosters.


Subject(s)
Adult Germline Stem Cells/transplantation , Busulfan/pharmacology , Chickens , Gamma Rays , Infertility/veterinary , Stem Cell Transplantation/veterinary , Animals , Infertility/chemically induced , Male , Spermatogenesis , Spermatogonia/drug effects , Spermatogonia/radiation effects , Stem Cell Transplantation/methods , Transplant Recipients
13.
Radiat Res ; 187(6): 722-731, 2017 06.
Article in English | MEDLINE | ID: mdl-28418813

ABSTRACT

Ionizing radiation can induce mutations, and the majority of radiation-induced mutations in mammalian cells are deletions. The most critical types of radiation-induced DNA damage are DNA double-strand breaks, and these breaks are repaired by either the homologous recombination (HR) pathway or the non-homologous end joining (NHEJ) pathway. The HR pathway is not as mutagenic as the NHEJ pathway, and it is expected that radiation-induced deletions would usually have little sequence similarity around the deletion junction points. Here we report sequence data from the regions around the rejoined junctions of 33 de novo copy-number mutations (27 deletions and 6 duplications) obtained from offspring sired by male mice that were irradiated at the spermatogonia stage and from nonirradiated controls. The results indicate that deletions can be classified into three major groups. In group 1, nine deletions were found to share long blocks of similar sequences (200-6,000 bp) at the junctions and the deletion size varied extensively (1 kb to 2 Mb) (e.g., illegitimate recombination). In group 2, five deletions shared short identical sequences (0-7 bp) at the junctions, and the deletion sizes were shorter than 200 kb (e.g., micro-homology-mediated repair). Additional three-deletion candidates of this group were also found but turned out to be inherited from mosaic parents. They are therefore not included in germline mutations. In group 3, twelve deletions shared little sequence similarity (only 0-2 bp) at the junctions (likely due to NHEJ repair) and deletion sizes were longer than 200 kb. Group 1 consisted of deletions found in both spontaneous and irradiated genomes and thus, were probably caused by spontaneous events during meiosis or DNA replication. Group 2 consisted mainly of deletions found in nonexposed genomes. Group 3 consisted primarily of deletions that occurred in the irradiated genomes. Among the duplications, we found no indication of any association with radiation exposures. These results indicate that large size (>200 kb) and little sequence similarity around the rejoined sites are likely to be a hallmark of radiation-induced deletions in mice.


Subject(s)
Conserved Sequence/genetics , Conserved Sequence/radiation effects , DNA Breaks/radiation effects , Gene Deletion , Spermatogonia/physiology , Spermatogonia/radiation effects , Animals , Base Sequence , Dose-Response Relationship, Radiation , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Radiation Dosage , Radiation, Ionizing
14.
Mol Hum Reprod ; 23(3): 141-154, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28130393

ABSTRACT

Study question: Does irradiation evoke adverse effects in germ and somatic cells in testis xenografts from prepubertal monkeys? Summary answer: In addition to the expected depletion of germ cells, a dose-dependent effect of irradiation was observed at the mRNA and protein level in Sertoli and peritubular myoid cells. What is known already: Testicular irradiation studies in monkeys have focused on the dose-dependent effects on germ cells. Previous studies using intact animals or xenografts reported that germ cells are highly sensitive to irradiation. Their depletion was demonstrated by morphometric and histological analyses. The effect of irradiation on expression of Sertoli and peritubular myoid cell markers, however, has not yet been described. Study design, size, duration: The testes of two prepubertal macaques (Macaca fascicularis) were dissected into testicular fragments. Fragments were randomly exposed in vitro to one of the following three doses of irradiation: 0 Gy, n = 60; 1 Gy, n = 54; 4 Gy, n = 72. Non-irradiated control fragments (0 Gy) were placed into the Faxitron for 6.6 min without irradiation. For 1 Gy and 4 Gy irradiation was applied for 1.7 and 6.6 min, respectively. Grafts were then either immediately analyzed or subcutaneously implanted under the back skin of 39 nude mice and analyzed after 6.5 months. Participants/materials setting methods: Post grafting, 133 testicular xenografts were retrieved. The body weight, serum testosterone level and seminal vesical weight of the host mice as well as the number and weight of retrieved grafts were determined. Larger grafts were used to evaluate both mRNA expression profiles and protein expression patterns. In total, 71 testicular fragments were used for morphometric and histological analysis while 68 fragments were analyzed for gene expression. For PCR arrays, M. fascicularis-specific primer sequences were employed. Irradiation-induced changes in the transcript levels of 34 marker genes were determined for each testicular graft. The effects of irradiation on peritubular myoid cells and Sertoli cells were confirmed by immunohistochemical analysis of chemokine (C-X-C motif) ligand type 11 (CXCL11), alpha smooth muscle actin (SMA) and chemokine (C-X-C motif) ligand type 12 (CXCL12). Main results and the role of chance: The four testes gave rise to 106 xenografts, which were individually analyzed, limiting the role of chance despite using only two monkeys in the study. Prior to grafting, the two donors displayed spermatogonia as the most advanced germ cell type in 95% and 70% of seminiferous tubules, respectively, while remaining tubules contained SCO. No spermatocytes were encountered prior to grafting in either monkey. After 6.5 months, non-irradiated grafts displayed spermatocytes in 15.4% and 1.8% of seminiferous tubules indicating an induction of meiosis. Irradiation resulted in a complete absence of spermatocytes. The percentage of seminiferous tubules containing spermatogonia declined in a dose-dependent manner. In non-irradiated xenografts, ~40% of tubules contained spermatogonia. This proportion was reduced to 3.4% and 4.3% in the 1 Gy treated group and to 1.3% and 0.2% in 4 Gy irradiated grafts. A dose-dependent decline in mRNA levels of selected germ cell marker genes supported the morphologically detected loss of germ cells. Irradiation had no effect on CXCL12 transcript levels. At the protein level, CXCL12-positive Sertoli cells were most abundant in the 1 Gy group compared to the 4 Gy group (P < 0.05), indicating a potential role of CXCL12 during recovery of primate spermatogenesis. The most prominent radiation-evoked changes were for CXCL11, which was localized to smooth muscle cells of blood vessels and seminiferous tubules. Transcript levels declined in a dose-dependent manner in grafts from both monkeys (MM687: P < 0.01 (0 Gy versus 4 Gy), MM627: P < 0.05 (0 Gy versus 4 Gy), P < 0.001 (1 Gy versus 4 Gy)). CXCL11 patterns of protein expression revealed irradiation-dependent changes as well. That peritubular cells are affected by X-irradiation was substantiated by changes at the transcript level between 1 and 4 Gy exposed groups (P < 0.01) and at the protein level of SMA (P < 0.05, 0 Gy versus 4 Gy). Large scale data: n/a. Limitations, reasons for caution: The spermatogonial stem cell system in primates is remarkably different from rodents. Therefore, data from a non-human primate may be more relevant to man. However, species-specific differences amongst primates cannot be fully excluded and the use of only two donors may raise concerns toward the generalization of the findings. There may also be important differences across the prepubertal period (e.g. infancy, early childhood) that are not represented by the ages included in the present study. Wider implications of the findings: This study is the first to indicate relevant testicular somatic cell responses following irradiation of prepubertal primate tissue. In addition to the well-known depletion of germ cells, the changes in Sertoli, and in particular peritubular myoid, cells may have important consequences for spermatogenic recovery. These novel findings should be taken into consideration when irradiation effects are assessed in tumor survivors. Study funding and competing interest(s): Interdisciplinary Center for Clinical Research (IZKF) Münster (Schl2/001/13) and the Excellence Cluster 'Cells in Motion' at the University Münster. There are no conflicts of interest to declare.


Subject(s)
Choristoma , Heterografts/radiation effects , Seminiferous Tubules/radiation effects , Sertoli Cells/radiation effects , Spermatogenesis/radiation effects , Spermatogonia/radiation effects , Actins/genetics , Actins/metabolism , Animals , Chemokine CXCL11/genetics , Chemokine CXCL11/metabolism , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Gamma Rays , Gene Expression Regulation , Heterografts/cytology , Heterografts/metabolism , Macaca fascicularis , Male , Mice , Mice, Nude , Seminiferous Tubules/cytology , Seminiferous Tubules/metabolism , Sertoli Cells/cytology , Sertoli Cells/metabolism , Sexual Maturation/physiology , Signal Transduction , Skin , Spermatogonia/cytology , Spermatogonia/metabolism , Transplantation, Heterologous
15.
Toxicol Ind Health ; 33(4): 373-384, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27738269

ABSTRACT

Even though there are contradictory reports regarding the cellular and molecular changes induced by mobile phone emitted radiofrequency radiation (RFR), the possibility of any biological effect cannot be ruled out. In view of a widespread and extensive use of mobile phones, this study evaluates alterations in male germ cell transformation kinetics following RFR exposure and after recovery. Swiss albino mice were exposed to RFR (900 MHz) for 4 h and 8 h duration per day for 35 days. One group of animals was terminated after the exposure period, while others were kept for an additional 35 days post-exposure. RFR exposure caused depolarization of mitochondrial membranes resulting in destabilized cellular redox homeostasis. Statistically significant increases in the damage index in germ cells and sperm head defects were noted in RFR-exposed animals. Flow cytometric estimation of germ cell subtypes in mice testis revealed 2.5-fold increases in spermatogonial populations with significant decreases in spermatids. Almost fourfold reduction in spermatogonia to spermatid turnover (1C:2C) and three times reduction in primary spermatocyte to spermatid turnover (1C:4C) was found indicating arrest in the premeiotic stage of spermatogenesis, which resulted in loss of post-meiotic germ cells apparent from testis histology and low sperm count in RFR-exposed animals. Histological alterations such as sloughing of immature germ cells into the seminiferous tubule lumen, epithelium depletion and maturation arrest were also observed. However, all these changes showed recovery to varied degrees following the post-exposure period indicating that the adverse effects of RFR on mice germ cells are detrimental but reversible. To conclude, RFR exposure-induced oxidative stress causes DNA damage in germ cells, which alters cell cycle progression leading to low sperm count in mice.


Subject(s)
Cell Phone , DNA Damage/radiation effects , Oligospermia/etiology , Radiation Injuries, Experimental/etiology , Radio Waves/adverse effects , Spermatogenesis/radiation effects , Spermatozoa/radiation effects , Animals , Comet Assay , Dose-Response Relationship, Radiation , Kinetics , Male , Meiotic Prophase I/radiation effects , Membrane Potential, Mitochondrial/radiation effects , Mice , Oligospermia/pathology , Oxidative Stress/radiation effects , Radiation Injuries, Experimental/pathology , Seminiferous Tubules/pathology , Seminiferous Tubules/radiation effects , Sperm Head/pathology , Sperm Head/radiation effects , Spermatogonia/pathology , Spermatogonia/radiation effects , Spermatozoa/pathology , Toxicity Tests, Subchronic
16.
Radiat Res ; 186(6): 568-576, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27869554

ABSTRACT

Until the end of the 20th century, mouse germ cell data on induced mutation rates, which were collected using classical genetic methods at preselected specific loci, provided the principal basis for estimates of genetic risks from radiation in humans. The work reported on here is an extension of earlier efforts in this area using molecular methods. It focuses on validating the use of array comparative genomic hybridization (array CGH) methods for identifying radiation-induced copy number variants (CNVs) and specifically for DNA deletions. The emphasis on deletions stems from the view that it constitutes the predominant type of radiation-induced genetic damage, which is relevant for estimating genetic risks in humans. In the current study, deletion mutations were screened in the genomes of F1 mice born to unirradiated or 4 Gy irradiated sires at the spermatogonia stage (100 offspring each). The array CGH analysis was performed using a "2M array" with over 2 million probes with a mean interprobe distance of approximately 1 kb. The results provide evidence of five molecularly-confirmed paternally-derived deletions in the irradiated group (5/100) and one in the controls (1/100). These data support a calculation, which estimates that the mutation rate is 1 × 10-2/Gy per genome for induced deletions; this is much lower than would be expected if one assumes that the specific locus rate of 1 × 10-5/locus per Gy (at 34 loci) is applicable to other genes in the genome. The low observed rate of induced deletions suggests that the effective number of genes/genomic regions at which recoverable deletions could be induced would be only approximately 1,000. This estimate is far lower than expected from the size of the mouse genome (>20,000 genes). Such a discrepancy between observation and expectation can occur if the genome contains numerous genes that are far less sensitive to radiation-induced deletions, if many deletion-bearing offspring are not viable or if the current method is substandard for detecting small deletions.


Subject(s)
Comparative Genomic Hybridization , Genomics , Mutagenesis/radiation effects , Oligonucleotide Array Sequence Analysis , Sequence Deletion/immunology , Spermatogonia/metabolism , Spermatogonia/radiation effects , Animals , Female , Male , Mice , Sequence Deletion/radiation effects
17.
Andrology ; 4(3): 412-24, 2016 05.
Article in English | MEDLINE | ID: mdl-26991593

ABSTRACT

Irradiation with 6 Gy produces a complete block of spermatogonial differentiation in LBNF1 rats that would be permanent without treatment. Subsequent suppression of gonadotropins and testosterone (T) restores differentiation to the spermatocyte stage; however, this process requires 6 weeks. We evaluated the role of Leydig cells (LCs) in maintenance of the block in spermatogonial differentiation after exposure to radiation by specifically eliminating functional LCs with ethane dimethane sulfonate (EDS). EDS (but not another alkylating agent), given at 10 weeks after irradiation, induced spermatogonial differentiation in 24% of seminiferous tubules 2 weeks later. However, differentiation became blocked again at 4 weeks as LCs recovered. When EDS was followed by treatment with GnRH antagonist and flutamide, sustained spermatogonial differentiation was induced in >70% of tubules within 2 weeks. When EDS was followed by GnRH antagonist plus exogenous T, which also inhibits LC recovery but restores follicle stimulating hormone (FSH) levels, the spermatogonial differentiation was again rapid but transient. These results confirm that the factors that block spermatogonial differentiation are indirectly regulated by T, and probably FSH, and that adult and possibly immature LCs contribute to the production of such inhibitory factors. We tested whether insulin-like 3 (INSL3), a LC-produced protein whose expression correlated with the block in spermatogonial differentiation, was indeed responsible for the block by injecting synthetic INSL3 into the testes and knocking down its expression in vivo with siRNA. Neither treatment had any effect on spermatogonial differentiation. The Leydig cell products that contribute to the inhibition of spermatogonial differentiation in irradiated rats remain to be elucidated.


Subject(s)
Leydig Cells/radiation effects , Spermatogenesis/radiation effects , Spermatogonia/radiation effects , Androgen Antagonists/pharmacology , Animals , Flutamide/pharmacology , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Leydig Cells/cytology , Leydig Cells/drug effects , Male , Mesylates/pharmacology , Oligopeptides/pharmacology , Radiation Dosage , Rats , Seminiferous Tubules/cytology , Seminiferous Tubules/drug effects , Seminiferous Tubules/radiation effects , Spermatogenesis/drug effects , Spermatogenesis/physiology , Spermatogonia/cytology , Spermatogonia/drug effects , Testosterone/pharmacology
18.
Zhonghua Nan Ke Xue ; 22(6): 491-495, 2016 Jun.
Article in Chinese | MEDLINE | ID: mdl-28963835

ABSTRACT

OBJECTIVE: To investigate the influence of cellphone electromagnetic radiation (CER) on the testicular ultrastructure and the apoptosis of spermatogenic cells in male rats.atability, feasibility, applicability, and controllability in the construction of experimental animal models, we compared the major anatomic features of the penis of 20 adult beagle dogs with those of 10 adult men. Using microsurgical techniques, we performed cross-transplantation of the penis in the 20 (10 pairs) beagle dogs and observed the survival rate of the transplanted penises by FK506+MMF+MP immune induction. We compared the relevant indexes with those of the 10 cases of microsurgical replantation of the amputated penis. METHODS: Thirty adult male SD rats were equally randomized into a 2 h CER, a 4 h CER, and a normal control group, the former two groups exposed to 30 days of 900 MHz CER for 2 and 4 hours a day, respectively, while the latter left untreated. Then the changes in the ultrastructure of the testis tissue were observed under the transmission electron microscope and the apoptosis of the spermatogenic cells was determined by TUNEL. RESULTS: Compared with the normal controls, the rats of the 2 h CER group showed swollen basement membrane of seminiferous tubules, separated tight junction of Sertoli cells, increased cell intervals, apparent vacuoles and medullization in some mitochondria, and increased apoptosis of spermatogenic cells, mainly the apoptosis of primary spermatocytes (P<0.05 ). In comparison with the 2 h CER group, the animals of the 4 h CER group exhibited swollen basement membrane of seminiferous tubules, more separated tight junction of Sertoli cells, wider cell intervals, incomplete membrane of spermatogonial cells, fragments of cytoplasm, nuclear pyknosis and notch, slight dilation of perinuclear space, abnormalities of intracellular mitochondria with vacuoles, fuzzy structure, and fusion or disappearance of some cristae, and increased damage of mitochondria and apoptosis of spermatogenic cells, including the apoptosis of spermatogonial cells, primary spermatocytes, and secondary spermatocytes (P<0.05 ). CONCLUSIONS: CER can damage the testicular ultrastructure and increase the apoptosis of spermatogenic cells of the male rat in a time-dependent manner, and the apoptosis of spermatogenic cells may be associated with the damage to mitochondria.


Subject(s)
Cell Phone , Electromagnetic Radiation , Testis/radiation effects , Animals , Apoptosis , Male , Mitochondria/radiation effects , Random Allocation , Rats , Rats, Sprague-Dawley , Seminiferous Tubules/radiation effects , Sertoli Cells/radiation effects , Spermatocytes/radiation effects , Spermatogonia/radiation effects , Testis/ultrastructure
19.
Int J Radiat Oncol Biol Phys ; 92(5): 1123-1131, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26059351

ABSTRACT

PURPOSE: Testicular spermatogenesis is extremely sensitive to radiation-induced damage, and even low scattered doses to testis from radiation therapy may pose reproductive risks with potential treatment-related infertility. Radiation-induced DNA double-strand breaks (DSBs) represent the greatest threat to the genomic integrity of spermatogonial stem cells (SSCs), which are essential to maintain spermatogenesis and prevent reproduction failure. METHODS AND MATERIALS: During daily low-dose radiation with 100 mGy or 10 mGy, radiation-induced DSBs were monitored in mouse testis by quantifying 53 binding protein 1 (53BP-1) foci in SSCs within their stem cell niche. The accumulation of DSBs was correlated with proliferation, differentiation, and apoptosis of testicular germ cell populations. RESULTS: Even very low doses of ionizing radiation arrested spermatogenesis, primarily by inducing apoptosis in spermatogonia. Eventual recovery of spermatogenesis depended on the survival of SSCs and their functional ability to proliferate and differentiate to provide adequate numbers of differentiating spermatogonia. Importantly, apoptosis-resistant SSCs resulted in increased 53BP-1 foci levels during, and even several months after, fractionated low-dose radiation, suggesting that surviving SSCs have accumulated an increased load of DNA damage. CONCLUSIONS: SSCs revealed elevated levels of DSBs for weeks after radiation, and if these DSBs persist through differentiation to spermatozoa, this may have severe consequences for the genomic integrity of the fertilizing sperm.


Subject(s)
DNA Breaks, Double-Stranded , Spermatogenesis/radiation effects , Spermatogonia/cytology , Spermatogonia/radiation effects , Stem Cells/radiation effects , Animals , Apoptosis/radiation effects , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Cell Survival/genetics , Cell Survival/physiology , Cell Survival/radiation effects , DNA Damage , DNA Repair , Male , Mice , Radiation Dosage , Seminiferous Tubules/cytology , Testis , Time Factors
20.
J Pak Med Assoc ; 65(3): 300-5, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25933566

ABSTRACT

OBJECTIVE: To determine the effects of two different radiation doses on sperm parameters and the role of testosterone treatment on rat spermatogenesis. METHODS: The experimental animal study was conducted at Marmara University, Istanbul, Turkey, from September 2012 to January 2013. Male Sprague Dawley 4-6 months old rats weighing 300-350g were randomely divided into 5 equal groups as control, low dose irradiation, testosterone administration following low dose irradiation, high dose irradiation, and testosterone administration following high dose irradiation. The animals were kept at a constant temperature in a room with 12h light and dark cycles. After the group-wise intervention, sperm concentration, testicular size, and histopathological examination of seminiferous tubules were noted. SPSS 10 was used for statistical analysis. RESULTS: The 40 rats in the study were divided in 5 groups of 8(20%) each. In low dose radiation, adverse effects were only temporarily observed with the return of almost normal testicular function at the end of two months with or without testosterone supplementation. In contrast, in high dose radiation, hormonal treatment effect was controversial. CONCLUSIONS: Testosterone treatment had no significant effect upon recovery after irradiation. In order to prevent the untoward effects of radiation, shielding of the remaining testis in a proper manner is crucial to avoid the harmful effects of the scattered radiation.


Subject(s)
Androgens/pharmacology , Spermatogenesis/drug effects , Spermatogenesis/radiation effects , Testis/drug effects , Testis/radiation effects , Testosterone/pharmacology , Animals , Dose-Response Relationship, Radiation , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Seminiferous Tubules/drug effects , Seminiferous Tubules/radiation effects , Sertoli Cells/drug effects , Sertoli Cells/radiation effects , Sperm Count , Spermatids/drug effects , Spermatids/radiation effects , Spermatocytes/drug effects , Spermatocytes/radiation effects , Spermatogonia/drug effects , Spermatogonia/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...