Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2656: 239-260, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37249876

RESUMO

Cytotoxic exposure, predominantly during radiation and/or chemotherapy treatment for cancer, interferes with fertility in men. While moderate doses cause temporary azoospermia allowing eventual recovery of spermatogenesis, higher doses of sterilizing agents can cause permanent sterility by killing the spermatogonial stem cells (SSCs). In this chapter, the methods involved in the following aspects of cytotoxic regeneration are described: (i) designing rodent and non-human primate models for regeneration of spermatogenesis after cytotoxic treatment by radiation and chemotherapy; (ii) analysis of SSCs with respect to the impact of the cytotoxic treatment, including analysis of spermatogonial clones, scoring the testicular section to analyze the extent of spermatogenic recovery, preparation of testicular and epididymal sperm, and collection of semen in non-human primates for sperm analysis; and (iii) preparation and delivery of a GnRH antagonist and steroids for enhancement or induction of spermatogonial differentiation, leading to the regeneration of spermatogenesis, largely applicable in the rat model.


Assuntos
Antineoplásicos , Sêmen , Masculino , Ratos , Animais , Espermatogênese , Espermatogônias , Testículo , Antineoplásicos/farmacologia , Primatas
2.
Andrology ; 9(5): 1603-1616, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33960147

RESUMO

BACKGROUND: Cancer treatment of prepubertal patients impacts future fertility due to the abolition of spermatogonial stem cells (SSCs). In macaques, spermatogenesis could be regenerated by intratesticular transplantation of SSCs, but no studies have involved cytotoxic treatment before puberty and transplantation after puberty, which would be the most likely clinical scenario. OBJECTIVES: To evaluate donor-derived functional sperm production after SSC transplantation to adult monkeys that had received testicular irradiation during the prepubertal period. MATERIALS AND METHODS: We obtained prepubertal testis tissue by unilaterally castrating six prepubertal monkeys and 2 weeks later irradiated the remaining testes with 6.9 Gy. However, because spermatogenic recovery was observed, we irradiated them again 14 months later with 7 Gy. Three of the monkeys were treated with GnRH-antagonist (GnRH-ant) for 8 weeks. The cryopreserved testis cells from the castrated testes were then allogeneically transplanted into the intact testes of all monkeys. Tissues were harvested 10 months later for analyses. RESULTS: In three of the six monkeys, 61%, 38%, and 11% of the epididymal sperm DNA were of the donor genotype. The ability to recover donor-derived sperm production was not enhanced by the GnRH-ant pretreatment. However, the extent of filling seminiferous tubules during the transplantation procedure was correlated with the eventual production of donor spermatozoa. The donor epididymal spermatozoa from the recipient with 61% donor contribution were capable of fertilizing rhesus eggs and forming embryos. Although the transplantation was done into the rete testis, two GnRH-ant-treated monkeys, which did not produce donor-derived epididymal spermatozoa, displayed irregular tubular cords in the interstitium containing testicular spermatozoa derived from the transplanted donor cells. DISCUSSION AND CONCLUSION: The results further support that sperm production can be restored in non-human primates from tissues cryopreserved prior to prepubertal and post-pubertal gonadotoxic treatment by transplantation of these testicular cells after puberty into seminiferous tubules.


Assuntos
Células-Tronco Germinativas Adultas/transplante , Puberdade/efeitos da radiação , Lesões Experimentais por Radiação/terapia , Espermatogênese/efeitos da radiação , Transplante de Células-Tronco , Animais , Criopreservação , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Antagonistas de Hormônios/administração & dosagem , Macaca mulatta , Masculino , Lesões Experimentais por Radiação/fisiopatologia , Túbulos Seminíferos , Espermatozoides/efeitos da radiação , Testículo/fisiopatologia , Testículo/efeitos da radiação
3.
J Am Vet Med Assoc ; 258(2): 161-164, 2021 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-33405981

Assuntos
Animais
4.
Andrology ; 8(5): 1428-1441, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32351003

RESUMO

BACKGROUND: In male pre-pubertal cancer patients, radiation and chemotherapy impact future fertility by eradication of spermatogonial stem cells (SSCs). In macaques, spermatogenesis could be regenerated by intratesticular transplantation of SSCs, but only a small percentage of spermatozoa produced were of donor origin. Transient hormone suppression with a GnRH antagonist (GnRH-ant) enhanced spermatogenic recovery from transplanted SSCs. OBJECTIVES: To evaluate donor-derived and endogenous spermatogenic recovery after SSC transplantation into irradiated monkeys and to test whether hormone suppression around the time of transplantation facilitates spermatogenic recovery. MATERIALS AND METHODS: Testes of 15 adult rhesus monkeys were irradiated with 7 Gy and 4 months later transplanted, to one of the testes, with cryopreserved testicular cells containing SSCs from unrelated monkeys. Monkeys were either treated with GnRH-ant for 8 weeks before transplantation, GnRH-ant from 4 weeks before to 4 weeks after transplantation, or with no GnRH-ant. Tissues were harvested 10 months after transplantation. RESULTS: Two of the 15 monkeys, a control and a pre-transplantation GnRH-ant-treated, showed substantially higher levels of testicular spermatogenesis and epididymal sperm output in the transplanted side as compared to the untransplanted. Over 84% of epididymal spermatozoa on the transplanted side had the donor genotype and were capable of fertilizing eggs after intracytoplasmic sperm injection forming morulae of the donor paternal origin. Low levels of donor spermatozoa (~1%) were also identified in the epididymis of three additional monkeys. Transplantation also appeared to enhance endogenous spermatogenesis. DISCUSSION AND CONCLUSION: We confirmed that SSC transplantation can be used for restoration of fertility in male cancer survivors exposed to irradiation as a therapeutic agent. The success rate of this procedure, however, is low. The success of filling the tubules with the cell suspension, but not the GnRH-ant treatment, was related to the level of colonization by transplanted cells.


Assuntos
Células-Tronco Germinativas Adultas/transplante , Espermatogênese/fisiologia , Espermatogônias/transplante , Transplante de Células-Tronco/métodos , Testículo/efeitos da radiação , Animais , Macaca mulatta , Masculino , Lesões Experimentais por Radiação
5.
FASEB J ; 33(7): 8423-8435, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30991836

RESUMO

Cytochrome P450 family 26 subfamily B member 1 (CYP26B1) regulates the concentration of all-trans retinoic acid (RA) and plays a key role in germ cell differentiation by controlling local distribution of RA. The mechanisms regulating Cyp26b1 expression in postnatal Sertoli cells, the main components of the stem cell niche, are so far unknown. During gonad development, expression of Cyp26b1 is maintained by Steroidogenic Factor 1 (SF-1) and Sex-Determining Region Y Box-9 (SOX9), which ensure that RA is degraded and germ cell differentiation is blocked. Here, we show that the NOTCH target Hairy/Enhancer-of-Split Related with YRPW Motif 1 (HEY1), a transcriptional repressor, regulates germ cell differentiation via direct binding to the Cyp26b1 promoter and thus inhibits its expression in Sertoli cells. Further, using in vivo germ cell ablation, we demonstrate that undifferentiated type A spermatogonia are the cells that activate NOTCH signaling in Sertoli cells through their expression of the NOTCH ligand JAGGED-1 (JAG1) at stage VIII of the seminiferous epithelium cycle, therefore mediating germ cell differentiation by a ligand concentration-dependent process. These data therefore provide more insights into the mechanisms of germ cell differentiation after birth and potentially explain the spatiotemporal RA pulses driving the transition between undifferentiated to differentiating spermatogonia.-Parekh, P. A., Garcia, T. X., Waheeb, R., Jain, V., Gandhi, P., Meistrich, M. L., Shetty, G., Hofmann, M.-C. Undifferentiated spermatogonia regulate Cyp26b1 expression through NOTCH signaling and drive germ cell differentiation.


Assuntos
Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Receptores Notch/metabolismo , Ácido Retinoico 4 Hidroxilase/biossíntese , Transdução de Sinais , Espermatogônias/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Receptores Notch/genética , Ácido Retinoico 4 Hidroxilase/genética , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Espermatogônias/citologia , Fator Esteroidogênico 1/genética , Fator Esteroidogênico 1/metabolismo
6.
Science ; 363(6433): 1314-1319, 2019 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-30898927

RESUMO

Testicular tissue cryopreservation is an experimental method to preserve the fertility of prepubertal patients before they initiate gonadotoxic therapies for cancer or other conditions. Here we provide the proof of principle that cryopreserved prepubertal testicular tissues can be autologously grafted under the back skin or scrotal skin of castrated pubertal rhesus macaques and matured to produce functional sperm. During the 8- to 12-month observation period, grafts grew and produced testosterone. Complete spermatogenesis was confirmed in all grafts at the time of recovery. Graft-derived sperm were competent to fertilize rhesus oocytes, leading to preimplantation embryo development, pregnancy, and the birth of a healthy female baby. Pending the demonstration that similar results are obtained in noncastrated recipients, testicular tissue grafting may be applied in the clinic.


Assuntos
Preservação da Fertilidade/métodos , Fertilização , Espermatogênese , Espermatozoides/crescimento & desenvolvimento , Testículo/fisiologia , Testículo/transplante , Animais , Autoenxertos , Criopreservação , Macaca mulatta , Masculino , Reprodução , Maturidade Sexual , Transplante Autólogo
7.
Nat Commun ; 9(1): 5339, 2018 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-30559363

RESUMO

A major challenge in stem cell differentiation is the availability of bioassays to prove cell types generated in vitro are equivalent to cells in vivo. In the mouse, differentiation of primordial germ cell-like cells (PGCLCs) from pluripotent cells was validated by transplantation, leading to the generation of spermatogenesis and to the birth of offspring. Here we report the use of xenotransplantation (monkey to mouse) and homologous transplantation (monkey to monkey) to validate our in vitro protocol for differentiating male rhesus (r) macaque PGCLCs (rPGCLCs) from induced pluripotent stem cells (riPSCs). Specifically, transplantation of aggregates containing rPGCLCs into mouse and nonhuman primate testicles overcomes a major bottleneck in rPGCLC differentiation. These findings suggest that immature rPGCLCs once transplanted into an adult gonadal niche commit to differentiate towards late rPGCs that initiate epigenetic reprogramming but do not complete the conversion into ENO2-positive spermatogonia.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/transplante , Espermatócitos/citologia , Espermatogênese/fisiologia , Espermatogônias/citologia , Testículo/metabolismo , Animais , Células Cultivadas , Feminino , Humanos , Macaca mulatta , Masculino , Camundongos , Camundongos Nus , Proteínas de Neoplasias/metabolismo , Fosfopiruvato Hidratase/metabolismo , Transplante Heterólogo , Transplante Homólogo
8.
Hum Reprod ; 33(12): 2249-2255, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30358843

RESUMO

STUDY QUESTION: Can transplanted primate testicular cells form seminiferous tubules de novo, supporting complete spermatogenesis? SUMMARY ANSWER: Cryopreserved testicular cells from a prepubertal monkey can reorganize in an adult monkey recipient testis forming de novo seminiferous tubular cords supporting complete spermatogenesis. WHAT IS KNOWN ALREADY: De novo morphogenesis of testicular tissue using aggregated cells from non-primate species grafted either subcutaneously or in the testis can support spermatogenesis. STUDY DESIGN, SIZE, DURATION: Two postpubertal rhesus monkeys (Macaca mulatta) were given testicular irradiation. One monkey was given GnRH-antagonist treatment from 8 to 16 weeks after irradiation, while the other received sham injections. At 16 weeks, cryopreserved testicular cells from two different prepubertal monkeys [43 × 106 viable (Trypan-blue excluding) cells in 260 µl, and 80 × 106 viable cells in 400 µl] were transplanted via ultrasound-guided injections to one of the rete testis in each recipient, and immune suppression was given. The contralateral testis was sham transplanted. Testes were analyzed 9 months after transplantation. PARTICIPANTS/MATERIALS, SETTING, METHODS: Spermatogenic recovery was assessed by testicular volume, weight, histology and immunofluorescence. Microsatellite genotyping of regions of testicular sections obtained by LCM determined whether the cells were derived from the host or transplanted cells. MAIN RESULTS AND THE ROLE OF CHANCE: Transplanted testis of the GnRH-antagonist-treated recipient, but not the sham-treated recipient, contained numerous irregularly shaped seminiferous tubular cords, 89% of which had differentiating germ cells, including sperm in a few of them. The percentages of donor genotype in different regions of this testis were as follows: normal tubule, 0%; inflammatory, 0%; abnormal tubule region, 67%; whole interior of abnormal tubules, >99%; adluminal region of the abnormal tubules, 92%. Thus, these abnormal tubules, including the enclosed germ cells, were derived de novo from the donor testicular cells. LARGE SCALE DATA: Not applicable. LIMITATIONS, REASONS FOR CAUTION: The de novo tubules were observed in only one out of the two monkeys transplanted with prepubertal donor testicular cells. WIDER IMPLICATIONS OF THE FINDINGS: These findings may represent a promising strategy for restoration of fertility in male childhood cancer survivors. The approach could be particularly useful in those exposed to therapeutic agents that are detrimental to the normal development of the tubule somatic cells affecting the ability of the endogenous tubules to support spermatogenesis, even from transplanted spermatogonial stem cells. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by research grants P01 HD075795 from Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD/NIH) to K.E.O and Cancer Center Support Grant P30 CA016672 from NCI/NIH to The University of Texas MD Anderson Cancer Center. The authors declare that they have no competing interests.


Assuntos
Túbulos Seminíferos/fisiologia , Espermatogênese/fisiologia , Testículo/citologia , Testículo/transplante , Animais , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Antagonistas de Hormônios/farmacologia , Macaca mulatta , Masculino
9.
Cell Rep ; 12(7): 1069-70, 2015 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-26287751

RESUMO

In this issue of Cell Reports, DeFalco et al. (2015) characterize a novel macrophage population associated with the peritubular lamina of mouse testes. These macrophages may create a niche not for the self-renewal of stem cells but rather the induction of their differentiation.


Assuntos
Macrófagos/metabolismo , Espermatogênese , Espermatogônias/citologia , Nicho de Células-Tronco , Testículo/citologia , Animais , Masculino
10.
PLoS One ; 9(4): e93311, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24691397

RESUMO

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.


Assuntos
Antineoplásicos Alquilantes/efeitos adversos , Ciclofosfamida/efeitos adversos , Exposição Materna , Folículo Ovariano/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal , Espermatogênese/efeitos dos fármacos , Neoplasias Testiculares/etiologia , Animais , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Folículo Ovariano/efeitos da radiação , Ovário/efeitos dos fármacos , Ovário/metabolismo , Ovário/patologia , Ovário/efeitos da radiação , Gravidez , Contagem de Espermatozoides , Espermatogênese/efeitos da radiação , Testículo/efeitos dos fármacos , Testículo/metabolismo , Testículo/patologia , Testículo/efeitos da radiação
11.
Fertil Steril ; 101(1): 3-13, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24314923

RESUMO

Improved therapies for cancer and other conditions have resulted in a growing population of long-term survivors. Infertility is an unfortunate side effect of some cancer therapies that impacts the quality of life of survivors who are in their reproductive or prereproductive years. Some of these patients have the opportunity to preserve their fertility using standard technologies that include sperm, egg, or embryo banking, followed by IVF and/or ET. However, these options are not available to all patients, especially the prepubertal patients who are not yet producing mature gametes. For these patients, there are several stem cell technologies in the research pipeline that may give rise to new fertility options and allow infertile patients to have their own biological children. We will review the role of stem cells in normal spermatogenesis as well as experimental stem cell-based techniques that may have potential to generate or regenerate spermatogenesis and sperm. We will present these technologies in the context of the fertility preservation paradigm, but we anticipate that they will have broad implications for the assisted reproduction field.


Assuntos
Células-Tronco Adultas/fisiologia , Células Germinativas/fisiologia , Regeneração/fisiologia , Espermatogênese/fisiologia , Células-Tronco Adultas/transplante , Animais , Feminino , Células Germinativas/transplante , Humanos , Masculino , Técnicas de Reprodução Assistida/tendências , Espermatozoides/fisiologia , Espermatozoides/transplante
12.
Reproduction ; 146(4): 363-76, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23884860

RESUMO

Ionizing radiation has been shown to arrest spermatogenesis despite the presence of surviving stem spermatogonia, by blocking their differentiation. This block is a result of damage to the somatic environment and is reversed when gonadotropins and testosterone are suppressed, but the mechanisms are still unknown. We examined spermatogonial differentiation and Sertoli cell factors that regulate spermatogonia after irradiation, during hormone suppression, and after hormone suppression combined with Leydig cell elimination with ethane dimethane sulfonate. These results showed that the numbers and cytoplasmic structure of Sertoli cells are unaffected by irradiation, only a few type A undifferentiated (Aund) spermatogonia and even fewer type A1 spermatogonia remained, and immunohistochemical analysis showed that Sertoli cells still produced KIT ligand (KITLG) and glial cell line-derived neurotrophic factor (GDNF). Some of these cells expressed KIT receptor, demonstrating that the failure of differentiation was not a result of the absence of the KIT system. Hormone suppression resulted in an increase in Aund spermatogonia within 3 days, a gradual increase in KIT-positive spermatogonia, and differentiation mainly to A3 spermatogonia after 2 weeks. KITL (KITLG) protein expression did not change after hormone suppression, indicating that it is not a factor in the stimulation. However, GDNF increased steadily after hormone suppression, which was unexpected since GDNF is supposed to promote stem spermatogonial self-renewal and not differentiation. We conclude that the primary cause of the block in spermatogonial development is not due to Sertoli cell factors such (KITL\GDNF) or the KIT receptor. As elimination of Leydig cells in addition to hormone suppression resulted in differentiation to the A3 stage within 1 week, Leydig cell factors were not necessary for spermatogonial differentiation.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Células Intersticiais do Testículo/metabolismo , Células de Sertoli/metabolismo , Espermatogênese/fisiologia , Espermatogônias/fisiologia , Fator de Células-Tronco/metabolismo , Testosterona/farmacologia , Androgênios/farmacologia , Animais , Diferenciação Celular/efeitos da radiação , Células Cultivadas , Técnicas Imunoenzimáticas , Células Intersticiais do Testículo/efeitos dos fármacos , Células Intersticiais do Testículo/efeitos da radiação , Masculino , Ratos , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/efeitos da radiação , Espermatogênese/efeitos dos fármacos , Espermatogênese/efeitos da radiação , Espermatogônias/efeitos dos fármacos , Espermatogônias/efeitos da radiação
13.
PLoS One ; 7(7): e41146, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22911753

RESUMO

Although decades of research have established that androgen is essential for spermatogenesis, androgen's mechanism of action remains elusive. This is in part because only a few androgen-responsive genes have been definitively identified in the testis. Here, we propose that microRNAs--small, non-coding RNAs--are one class of androgen-regulated trans-acting factors in the testis. Specifically, by using androgen suppression and androgen replacement in mice, we show that androgen regulates the expression of several microRNAs in Sertoli cells. Our results reveal that several of these microRNAs are preferentially expressed in the testis and regulate genes that are highly expressed in Sertoli cells. Because androgen receptor-mediated signaling is essential for the pre- and post-meiotic germ cell development, we propose that androgen controls these events by regulating Sertoli/germ cell-specific gene expression in a microRNA-dependent manner.


Assuntos
Androgênios/farmacologia , MicroRNAs/genética , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/metabolismo , Animais , Sequência de Bases , Análise por Conglomerados , Desmocolinas , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Especificidade de Órgãos/genética , Espermatogênese/efeitos dos fármacos , Espermatogênese/genética , Testículo/metabolismo , Testosterona/farmacologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
14.
PLoS One ; 7(2): e32064, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22348147

RESUMO

The prevalence of testicular germ cell tumors (TGCT), a common solid tissue malignancy in young men, has been annually increasing at an alarming rate of 3%. Since the majority of testicular cancers are derived from germ cells at the stage of transformation of primordial germ cell (PGC) into gonocytes, the increase has been attributed to maternal/fetal exposures to environmental factors. We examined the effects of an estrogen (diethylstilbestrol, DES), an antiandrogen (flutamide), or radiation on the incidence of testicular germ cell tumors in genetically predisposed 129.MOLF-L1 (L1) congenic mice by exposing them to these agents on days 10.5 and 11.5 of pregnancy. Neither flutamide nor DES produced noticeable increases in testis cancer incidence at 4 weeks of age. In contrast, two doses of 0.8-Gy radiation increased the incidence of TGCT from 45% to 100% in the offspring. The percentage of mice with bilateral tumors, weights of testes with TGCT, and the percentage of tumors that were clearly teratomas were higher in the irradiated mice than in controls, indicating that irradiation induced more aggressive tumors and/or more foci of initiation sites in each testis. This radiation dose did not disrupt spermatogenesis, which was qualitatively normal in tumor-free testes although they were reduced in size. This is the first proof of induction of testicular cancer by an environmental agent and suggests that the male fetus of women exposed to radiation at about 5-6 weeks of pregnancy might have an increased risk of developing testicular cancer. Furthermore, it provides a novel tool for studying the molecular and cellular events of testicular cancer pathogenesis.


Assuntos
Feto/efeitos da radiação , Efeitos Tardios da Exposição Pré-Natal , Neoplasias Testiculares/etiologia , Antagonistas de Androgênios/toxicidade , Animais , Dietilestilbestrol/toxicidade , Estrogênios não Esteroides/toxicidade , Feminino , Flutamida/toxicidade , Predisposição Genética para Doença , Masculino , Exposição Materna , Camundongos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética
15.
Toxicol Sci ; 126(2): 545-53, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22273744

RESUMO

Previous studies with Lewis/Brown-Norway (BN) F1 hybrid rats indicated that spermatogenesis was much more sensitive to ionizing radiation than in the widely studied outbred Sprague Dawley stock, suggesting that there were genetically based differences; however, the relative sensitivities of various inbred strains had not been established. As a first step to defining the genes responsible for these differences, we compared the sensitivities of seven rat strains to radiation damage of spermatogenesis. Recovery of spermatogenesis was examined 10 weeks after 5-Gy irradiation of seven strains (BN, Lewis, Long-Evans, Wistar Kyoto, spontaneously hypertensive [SHR], Fischer 344, and Sprague Dawley). The percentages of tubules containing differentiated cells and testicular sperm counts showed that BN and Lewis were most sensitive to radiation (< 2% of tubules recovered, < 2 × 10(5) late spermatids per testis), Long-Evans, Wistar Kyoto, Fischer, and SHR were more resistant, and Sprague Dawley was the most resistant (98% of tubules recovered, 2 × 10(7) late spermatids per testis). Although increases in intratesticular testosterone levels and interstitial fluid volume after irradiation had been suggested as factors inhibiting recovery of spermatogenesis, neither appeared to correlate with the radiation sensitivity of spermatogenesis in these strains. In all strains, the atrophic tubules without differentiated germ cells nevertheless showed the presence of type A spermatogonia, indicating that their differentiation was blocked. Thus, we conclude that the differences in radiation sensitivity of recovery of spermatogenesis between rat strains of different genetic backgrounds can be accounted for by differences in the extent of the radiation-induced block of spermatogonial differentiation.


Assuntos
Tolerância a Radiação , Espermatogênese/efeitos da radiação , Animais , Masculino , Ratos , Ratos Endogâmicos , Especificidade da Espécie , Contagem de Espermatozoides
16.
Endocrinology ; 152(9): 3504-14, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21733828

RESUMO

Why both testosterone (T) suppression and cryptorchidism reverse the block in spermatogonial differentiation in adult mice homozygous for the juvenile spermatogonial depletion (jsd) mutation has been a conundrum. To resolve this conundrum, we analyzed interrelations between T suppression, testicular temperature, and spermatogonial differentiation and used in vitro techniques to separate the effects of the two treatments on the spermatogonial differentiation block in jsd mice. Temporal analysis revealed that surgical cryptorchidism rapidly stimulated spermatogonial differentiation whereas androgen ablation treatment produced a delayed and gradual differentiation. The androgen suppression caused scrotal shrinkage, significantly increasing the intrascrotal temperature. When serum T or intratesticular T (ITT) levels were modulated separately in GnRH antagonist-treated mice by exogenous delivery of T or LH, respectively, the inhibition of spermatogonial differentiation correlated with the serum T and not with ITT levels. Thus, the block must be caused by peripheral androgen action. When testicular explants from jsd mice were cultured in vitro at 32.5 C, spermatogonial differentiation was not observed, but at 37 C significant differentiation was evident. In contrast, addition of T to the culture medium did not block the stimulation of spermatogonial differentiation at 37 C, and androgen ablation with aminoglutethimide and hydroxyflutamide did not stimulate differentiation at 32.5 C, suggesting that T had no direct effect on spermatogonial differentiation in jsd mice. These data show that elevation of temperature directly overcomes the spermatogonial differentiation block in adult jsd mice and that T suppression acts indirectly in vivo by causing scrotal regression and thereby elevating the testicular temperature.


Assuntos
Androgênios/farmacologia , Temperatura Corporal/efeitos dos fármacos , Ribonucleoproteínas Nucleolares Pequenas/genética , Espermatogênese/efeitos dos fármacos , Testículo/efeitos dos fármacos , Testosterona/farmacologia , Animais , Temperatura Corporal/fisiologia , Criptorquidismo , Homozigoto , Hormônio Luteinizante/farmacologia , Masculino , Camundongos , Ribonucleoproteínas Nucleolares Pequenas/metabolismo , Escroto/efeitos dos fármacos , Escroto/fisiologia , Espermatogênese/fisiologia , Espermatogônias/efeitos dos fármacos , Espermatogônias/fisiologia , Testículo/fisiologia
17.
Biol Reprod ; 85(4): 823-33, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21653891

RESUMO

Despite numerous observations of the effects of estrogens on spermatogenesis, identification of estrogen-regulated genes in the testis is limited. Using rats in which irradiation had completely blocked spermatogonial differentiation, we previously showed that testosterone suppression with gonadotropin-releasing hormone-antagonist acyline and the antiandrogen flutamide stimulated spermatogenic recovery and that addition of estradiol (E2) to this regimen accelerated this recovery. We report here the global changes in testicular cell gene expression induced by the E2 treatment. By minimizing the changes in other hormones and using concurrent data on regulation of the genes by these hormones, we were able to dissect the effects of estrogen on gene expression, independent of gonadotropin or testosterone changes. Expression of 20 genes, largely in somatic cells, was up- or downregulated between 2- and 5-fold by E2. The unexpected and striking enrichment of transcripts not corresponding to known genes among the E2-downregulated probes suggested that these might represent noncoding mRNAs; indeed, we have identified several as miRNAs and their potential target genes in this system. We propose that genes for which expression levels are altered in one direction by irradiation and in the opposite direction by both testosterone suppression and E2 treatment are candidates for controlling the block in differentiation. Several genes, including insulin-like 3 (Insl3), satisfied those criteria. If they are indeed involved in the inhibition of spermatogonial differentiation, they may be candidate targets for treatments to enhance recovery of spermatogenesis following gonadotoxic exposures, such as those resulting from cancer therapy.


Assuntos
Estradiol/uso terapêutico , Estrogênios/uso terapêutico , Regulação da Expressão Gênica/efeitos dos fármacos , Espermatogênese/efeitos dos fármacos , Espermatogênese/efeitos da radiação , Testículo/efeitos dos fármacos , Testículo/metabolismo , Antagonistas de Androgênios/uso terapêutico , Animais , Cruzamentos Genéticos , Quimioterapia Combinada , Flutamida/uso terapêutico , Raios gama , Regulação da Expressão Gênica/efeitos da radiação , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Antagonistas de Hormônios/uso terapêutico , Insulina/genética , Insulina/metabolismo , Masculino , MicroRNAs/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Oligopeptídeos/uso terapêutico , Proteínas/genética , Proteínas/metabolismo , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos Lew , Testículo/patologia , Testículo/efeitos da radiação , Testosterona/antagonistas & inibidores
18.
Biol Reprod ; 82(1): 54-65, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19684331

RESUMO

Although gonadotropins and androgen are required for normal spermatogenesis and both testosterone and follicle-stimulating hormone (FSH) are responsible for the inhibition of spermatogonial differentiation that occurs in irradiated rats, it has been difficult to identify the specific genes involved. To study specific hormonally regulated changes in somatic cell gene expression in the testis that may be involved in these processes, without the complication of changing populations of germ cells, we used irradiated LBNF(1) rats, the testes of which contain almost exclusively somatic cells except for a few type A spermatogonia. Three different groups of these rats were treated with various combinations of gonadotropin-releasing hormone antagonist, an androgen receptor antagonist (flutamide), testosterone, and FSH, and we compared the gene expression levels 2 wk later to those of irradiated-only rats by microarray analysis. By dividing the gene expression patterns into three major patterns and 11 subpatterns, we successfully distinguished, in a single study, the genes that were specifically regulated by testosterone, by luteinizing hormone (LH), and by FSH from the large number of genes that were not hormonally regulated in the testis. We found that hormones produced more dramatic upregulation than downregulation of gene expression: Testosterone had the strongest upregulatory effect, LH had a modest but appreciable upregulatory effect, and FSH had a minor upregulatory effect. We also separately identified the somatic cell genes that were chronically upregulated by irradiation. Thus, the present study identified gene expression changes that may be responsible for hormonal action on somatic cells to support normal spermatogenesis and the hormone-mediated block in spermatogonial development after irradiation.


Assuntos
Hormônio Foliculoestimulante/metabolismo , Regulação da Expressão Gênica , Hormônio Luteinizante/metabolismo , Testículo/metabolismo , Testosterona/farmacologia , Animais , Flutamida/farmacologia , Raios gama , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Células Germinativas/efeitos dos fármacos , Células Germinativas/metabolismo , Células Germinativas/efeitos da radiação , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Oligopeptídeos/farmacologia , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Testículo/efeitos dos fármacos , Testículo/efeitos da radiação , Testosterona/sangue
19.
J Androl ; 30(4): 440-51, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19136390

RESUMO

Irradiation of LBNF(1) rat testes induces spermatogonial differentiation arrest, which can be reversed by gonadotropin-releasing hormone (GnRH) antagonist-induced suppression of intratesticular testosterone (ITT) and follicle-stimulating hormone (FSH). Although exogenous estrogen treatment also enhanced spermatogenic recovery, as measured by the tubule differentiation index (TDI), it was not clear whether estrogen stimulated spermatogonial differentiation only by further suppressing ITT or by an additional independent mechanism as well. To resolve this question, we performed the following experiments. At 15 weeks after irradiation, rats were treated with GnRH antagonist; some also received 17beta-estradiol (E2) and were killed 4 weeks later. GnRH antagonist treatment increased the TDI from 0% to 8%, and addition of E2 further increased the TDI to 39%. However, E2 addition further reduced ITT from 7 ng/g testis, observed with GnRH antagonist to 3 ng/g testis, so decreased ITT levels might have contributed to recovery. Next GnRH antagonist-treated rats were given exogenous testosterone and flutamide to stabilize ITT levels and block its action. This increased TDI slightly from 8% to 13%, but the further addition of E2 significantly raised the TDI to 27%, indicating it acted by a mechanism independent of ITT levels. Plots of TDI for all treatment groups compared with ITT, FSH, or a linear combination of ITT and FSH showed that treatments including E2 produced higher TDI values than did treatments without E2. These results indicate that there was an effect of E2 on spermatogonial differentiation because of an additional direct action on the testis that is unrelated to its suppression of testosterone or gonadotropins.


Assuntos
Estradiol/farmacologia , Espermatogênese/efeitos dos fármacos , Testículo/efeitos dos fármacos , Testículo/efeitos da radiação , Antagonistas de Androgênios/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Estradiol/metabolismo , Flutamida/farmacologia , Hormônio Foliculoestimulante/sangue , Masculino , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos Lew , Ratos Sprague-Dawley , Espermatogênese/efeitos da radiação , Espermatogônias/efeitos dos fármacos , Testosterona/antagonistas & inibidores , Testosterona/farmacologia
20.
Reproduction ; 137(3): 497-508, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19036951

RESUMO

Irradiation of rat testes leads to the failure to support differentiation of the surviving spermatogonia due to damage of the somatic environment. To determine the involvement of Sertoli cells in this somatic damage, we transplanted seminiferous tubule cells from normal immature GFP-transgenic rats into the testes of irradiated rats. The donor Sertoli cells colonized and developed in the host testes. In many seminiferous tubules, the donor Sertoli cells formed abnormal spherical structures in the lumen, but in some tubules they formed a normal-appearing epithelium, but with only isolated spermatogonia, on the basement membrane. When the donor cells were injected into the interstitial region of the testis, they formed tubule-like structures containing Sertoli cells and occasional isolated spermatogonia, both of donor origin. Surprisingly, in host tubules adjacent to these newly formed donor-cell tubules or adjacent to the endogenous tubules with abnormal donor Sertoli-cell structures, endogenous spermatogonia differentiated to the spermatocyte or even to spermatid stages. Around these newly donor cell-formed tubules and the host tubules with abnormal donor Sertoli-cell structures, many cells including macrophages, which perhaps represented chronic inflammation, accumulated in the interstitium. We conclude that the donor Sertoli cells that colonized the seminiferous tubules did not directly support recovery of spermatogenesis. Instead, the colonizing Sertoli cells acted indirectly on the interstitium to stimulate localized differentiation of endogenous spermatogonia.


Assuntos
Células de Sertoli/transplante , Espermatogênese/efeitos da radiação , Testículo/efeitos da radiação , Animais , Contagem de Células , Diferenciação Celular , Imuno-Histoquímica , Masculino , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos Lew , Ratos Transgênicos , Células de Sertoli/fisiologia , Espermatogênese/fisiologia , Espermatozoides/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...