Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Endocrinology ; 163(1)2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34902009

RESUMEN

Concordant transcriptional regulation can generate multiple gene products that collaborate to achieve a common goal. Here we report a case of concordant transcriptional regulation that instead drives a single protein to be produced in the same cell type from divergent promoters. This gene product-the RHOX5 homeobox transcription factor-is translated from 2 different mRNAs with different 5' untranslated regions (UTRs) transcribed from alternative promoters. Despite the fact that these 2 promoters-the proximal promoter (Pp) and the distal promoter (Pd)-exhibit different patterns of tissue-specific activity, share no obvious sequence identity, and depend on distinct transcription factors for expression, they exhibit a remarkably similar expression pattern in the testes. In particular, both depend on androgen signaling for expression in the testes, where they are specifically expressed in Sertoli cells and have a similar stage-specific expression pattern during the seminiferous epithelial cycle. We report evidence for 3 mechanisms that collaborate to drive concordant Pp/Pd expression. First, both promoters have an intrinsic ability to respond to androgen receptor and androgen. Second, the Pp acts as an enhancer to promote androgen-dependent transcription from the Pd. Third, Pd transcription is positively autoregulated by the RHOX5 protein, which is first produced developmentally from the Pp. Together, our data support a model in which the Rhox5 homeobox gene evolved multiple mechanisms to activate both of its promoters in Sertoli cells to produce Rhox5 in an androgen-dependent manner during different phases of spermatogenesis.


Asunto(s)
Andrógenos/metabolismo , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Regiones Promotoras Genéticas , Células de Sertoli/metabolismo , Factores de Transcripción/genética , Regiones no Traducidas 5' , Animales , Metilación de ADN , Genes Homeobox , Masculino , Ratones , Ratones Endogámicos C57BL , Plásmidos/metabolismo , Isoformas de Proteínas , Receptores Androgénicos/metabolismo , Túbulos Seminíferos/metabolismo , Espermatogénesis , Testículo/metabolismo , Factores de Transcripción/metabolismo
2.
Andrology ; 9(5): 1603-1616, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33960147

RESUMEN

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.


Asunto(s)
Células Madre Germinales Adultas/trasplante , Pubertad/efectos de la radiación , Traumatismos Experimentales por Radiación/terapia , Espermatogénesis/efectos de la radiación , Trasplante de Células Madre , Animales , Criopreservación , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Antagonistas de Hormonas/administración & dosificación , Macaca mulatta , Masculino , Traumatismos Experimentales por Radiación/fisiopatología , Túbulos Seminíferos , Espermatozoides/efectos de la radiación , Testículo/fisiopatología , Testículo/efectos de la radiación
3.
PLoS One ; 15(12): e0242218, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33370316

RESUMEN

Improvements in survival rates with gonad-sparing protocols for childhood and adolescence cancer have increased the optimism of survivors to become parents after treatment. Findings in rodents indicate that chromosomal aberrations can be induced in male germ cells by genotoxic exposures and transmitted to offspring and future generations with effects on development, fertility and health. Thus, there is a need for effective technologies to identify human sperm carrying chromosomal aberrations to assess the germ-line risks, especially for cancer survivors who have received genotoxic therapies. The time-dependent changes in the burden of sperm carrying structural chromosomal aberrations were assessed for the first time in a cancer setting, using the AM8 sperm FISH protocol which simultaneously detects abnormalities in chromosomal structure and number in sperm. Nine Hodgkin lymphoma (HL) patients provided 20 semen samples before, during, and after NOVP therapy (Novantrone, Oncovin, Velban and Prednisone) and radiation therapy that produced scattered gonadal doses from <0.05 to 0.6 Gy. Late meiosis was found to be the most sensitive to NOVP treatment for the production of sperm with chromosomal abnormalities, both in structure and number. Earlier stages of spermatogenesis were less sensitive and there was no evidence that therapy-exposed stem cells resulted in increased frequencies of sperm with abnormalities in chromosomal structure or number. This indicates that NOVP therapy may increase the risks for paternal transmission of chromosomal structural aberrations for sperm produced 32 to 45 days after a treatment with these drugs and implies that there are no excess risks for pregnancies conceived more than 6 months after this therapy. This clinical evaluation of the AM8 sperm FISH protocol indicates that it is a promising tool for assessing an individual's burden of sperm carrying chromosomal structural aberrations as well as aneuploidies after cancer therapy, with broad applications in other clinical and environmental situations that may pose aneugenic or clastogenic risks to human spermatogenesis.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Aberraciones Cromosómicas/efectos de los fármacos , Enfermedad de Hodgkin/terapia , Meiosis/efectos de los fármacos , Análisis de Semen/métodos , Espermatozoides/efectos de los fármacos , Adulto , Células Madre Germinales Adultas/efectos de los fármacos , Células Madre Germinales Adultas/efectos de la radiación , Supervivientes de Cáncer , Quimioradioterapia/efectos adversos , Quimioradioterapia/métodos , Aberraciones Cromosómicas/efectos de la radiación , Estudios de Cohortes , Preservación de la Fertilidad , Humanos , Hibridación Fluorescente in Situ/métodos , Masculino , Meiosis/efectos de la radiación , Mitoxantrona/efectos adversos , Mutagénesis/efectos de los fármacos , Mutagénesis/efectos de la radiación , Tratamientos Conservadores del Órgano/efectos adversos , Tratamientos Conservadores del Órgano/métodos , Órganos en Riesgo/efectos de la radiación , Prednisona/efectos adversos , Dosificación Radioterapéutica , Espermatogénesis/efectos de los fármacos , Espermatogénesis/efectos de la radiación , Espermatozoides/fisiología , Espermatozoides/efectos de la radiación , Testículo/efectos de los fármacos , Testículo/efectos de la radiación , Factores de Tiempo , Vinblastina/efectos adversos , Vincristina/efectos adversos
4.
Andrology ; 8(5): 1428-1441, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32351003

RESUMEN

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.


Asunto(s)
Células Madre Germinales Adultas/trasplante , Espermatogénesis/fisiología , Espermatogonias/trasplante , Trasplante de Células Madre/métodos , Testículo/efectos de la radiación , Animales , Macaca mulatta , Masculino , Traumatismos Experimentales por Radiación
5.
Andrology ; 8(3): 545-558, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31821745

RESUMEN

BACKGROUND: Men who have just started cytotoxic therapy for cancer are uncertain and concerned about whether spermatozoa collected or pregnancies occurring during therapy might be transmitting genetic damage to offspring. There are no comprehensive guidelines on the risks of different doses of the various cytotoxic, and usually genotoxic, antineoplastic agents. OBJECTIVES: To develop a schema showing the risks of mutagenic damage when spermatozoa, exposed to various genotoxic agents during spermatogenesis, are collected or used to produce a pregnancy. MATERIALS AND METHODS: A comprehensive literature review was performed updating the data on genetic and epigenetic effects of genotoxic agents on animal and human spermatozoa exposed during spermatogenic development. RESULTS: Relevant data on human spermatozoa and offspring are extremely limited, but there are extensive genetic studies in experimental animals that define sensitivities for specific drugs and times. The animal data were extrapolated to humans based on the stage when the cells were exposed and the relative kinetics of spermatogenesis and were consistent with the limited human data. In humans, alkylating agents and radiation should already induce a high risk of mutations in spermatozoa produced within 1 or 2 weeks after initiation of therapy. Topoisomerase II inhibitors and possibly microtubule inhibitors produce the greatest risk at weeks 5-7 of therapy. Nucleoside analogs, antimetabolites, and bleomycin exert their mutagenic effects on spermatozoa collected at 7-10 weeks of therapy. DISCUSSION AND CONCLUSIONS: A schema showing the time from initiation of therapy at which specific antineoplastic agents can cause significant levels of genetic damage in conceptuses and live offspring was developed. The estimates and methods for computing the level of such risk from an individual patient's treatment regimen will enable patients and counselors to make informed decisions on the use of spermatozoa or continuation of a pregnancy.


Asunto(s)
Antineoplásicos/efectos adversos , Radioterapia/efectos adversos , Espermatozoides/efectos de los fármacos , Espermatozoides/efectos de la radiación , Animales , Daño del ADN/efectos de la radiación , Quimioterapia , Humanos , Masculino
6.
Hum Reprod ; 34(8): 1404-1415, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31348830

RESUMEN

STUDY QUESTION: What effect does cancer treatment have on levels of spontaneous selfish fibroblast growth factor receptor 2 (FGFR2) point mutations in human sperm? SUMMARY ANSWER: Chemotherapy and radiotherapy do not increase levels of spontaneous FGFR2 mutations in sperm but, unexpectedly, highly-sterilizing treatments dramatically reduce the levels of the disease-associated c.755C > G (Apert syndrome) mutation in sperm. WHAT IS KNOWN ALREADY: Cancer treatments lead to short-term increases in gross DNA damage (chromosomal abnormalities and DNA fragmentation) but the long-term effects, particularly at the single nucleotide resolution level, are poorly understood. We have exploited an ultra-sensitive assay to directly quantify point mutation levels at the FGFR2 locus. STUDY DESIGN, SIZE, DURATION: 'Selfish' mutations are disease-associated mutations that occur spontaneously in the sperm of most men and their levels typically increase with age. Levels of mutations at c.752-755 of FGFR2 (including c.755C > G and c.755C > T associated with Apert and Crouzon syndromes, respectively) in semen post-cancer treatment from 18 men were compared to levels in pre-treatment samples from the same individuals (n = 4) or levels in previously screened population controls (n = 99). PARTICIPANTS/MATERIALS, SETTING, METHODS: Cancer patients were stratified into four different groups based on the treatments they received and the length of time for spermatogenesis recovery. DNA extracted from semen samples was analysed using a previously established highly sensitive assay to identify mutations at positions c.752-755 of FGFR2. Five to ten micrograms of semen genomic DNA was spiked with internal controls for quantification purposes, digested with MboI restriction enzyme and gel extracted. Following PCR amplification, further MboI digestion and a nested PCR with barcoding primers, samples were sequenced on Illumina MiSeq. Mutation levels were determined relative to the spiked internal control; in individuals heterozygous for a nearby common single nucleotide polymorphism (SNP), mutations were phased to their respective alleles. MAIN RESULTS AND THE ROLE OF CHANCE: Patients treated with moderately-sterilizing alkylating regimens and who recovered spermatogenesis within <3 years after therapy (Group 3, n = 4) or non - alkylating chemotherapy and/or low gonadal radiation doses (Group 1, n = 4) had mutation levels similar to untreated controls. However, patients who had highly-sterilizing alkylating treatments (i.e. >5 years to spermatogenesis recovery) (Group 2, n = 7) or pelvic radiotherapy (Group 4, n = 3) exhibited c.755C > G mutation levels at or below background. Two patients (A and B) treated with highly-sterilizing alkylating agents demonstrated a clear reduction from pre-treatment levels; however pre-treatment samples were not available for the other patients with low mutation levels. Therefore, although based on their age we would expect detectable levels of mutations, we cannot exclude the possibility that these patients also had low mutation levels pre-treatment. In three patients with low c.755C > G levels at the first timepoint post-treatment, we observed increasing mutation levels over time. For two such patients we could phase the mutation to a nearby polymorphism (SNP) and determine that the mutation counts likely originated from a single or a small number of mutational events. LIMITATIONS, REASONS FOR CAUTION: This study was limited to 18 patients with different treatment regimens; for nine of the 18 patients, samples from only one timepoint were available. Only 12 different de novo substitutions at the FGFR2 c.752-755 locus were assessed, two of which are known to be disease associated. WIDER IMPLICATIONS OF THE FINDINGS: Our data add to the body of evidence from epidemiological studies and experimental data in humans suggesting that male germline stem cells are resilient to the accumulation of spontaneous mutations. Collectively, these data should provide physicians and health-care professionals with reassuring experimental-based evidence for counselling of male cancer patients contemplating their reproductive options several years after treatment. STUDY FUNDING/COMPETING INTEREST(S): This work was primarily supported by grants from the Wellcome (grant 091182 to AG and AOMW; grant 102 731 to AOMW), the University of Oxford Medical Sciences Division Internal Fund (grant 0005128 to GJM and AG), the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre Programme (to AG) and the US National Institutes of Health (to MLM). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. None of the authors has any conflicts of interest to declare. TRIAL REGISTRATION NUMBER: NA.


Asunto(s)
Antineoplásicos/administración & dosificación , Supervivientes de Cáncer , Neoplasias/terapia , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Espermatozoides/efectos de la radiación , Adulto , Antineoplásicos/uso terapéutico , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Humanos , Masculino , Mutación/efectos de los fármacos , Mutación/efectos de la radiación , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Radioterapia , Análisis de Semen , Recuento de Espermatozoides , Espermatogénesis/efectos de los fármacos , Espermatogénesis/efectos de la radiación , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo
7.
FASEB J ; 33(7): 8423-8435, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30991836

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Receptores Notch/metabolismo , Ácido Retinoico 4-Hidroxilasa/biosíntesis , Transducción de Señal , Espermatogonias/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Masculino , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Receptores Notch/genética , Ácido Retinoico 4-Hidroxilasa/genética , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Espermatogonias/citología , Factor Esteroidogénico 1/genética , Factor Esteroidogénico 1/metabolismo
8.
Science ; 363(6433): 1314-1319, 2019 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-30898927

RESUMEN

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.


Asunto(s)
Preservación de la Fertilidad/métodos , Fertilización , Espermatogénesis , Espermatozoides/crecimiento & desarrollo , Testículo/fisiología , Testículo/trasplante , Animales , Autoinjertos , Criopreservación , Macaca mulatta , Masculino , Reproducción , Maduración Sexual , Trasplante Autólogo
9.
Nat Commun ; 9(1): 5339, 2018 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-30559363

RESUMEN

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.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/trasplante , Espermatocitos/citología , Espermatogénesis/fisiología , Espermatogonias/citología , Testículo/metabolismo , Animales , Células Cultivadas , Femenino , Humanos , Macaca mulatta , Masculino , Ratones , Ratones Desnudos , Proteínas de Neoplasias/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Trasplante Heterólogo , Trasplante Homólogo
10.
Hum Reprod ; 33(12): 2249-2255, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30358843

RESUMEN

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.


Asunto(s)
Túbulos Seminíferos/fisiología , Espermatogénesis/fisiología , Testículo/citología , Testículo/trasplante , Animales , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Antagonistas de Hormonas/farmacología , Macaca mulatta , Masculino
11.
Sci Rep ; 6: 32960, 2016 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-27608812

RESUMEN

HMGB4 is a new member in the family of HMGB proteins that has been characterized in sperm cells, but little is known about its functions in somatic cells. Here we show that HMGB4 and the highly similar rat Transition Protein 4 (HMGB4L1) are expressed in neuronal cells. Both proteins had slow mobility in nucleus of living NIH-3T3 cells. They interacted with histones and their differential expression in transformed cells of the nervous system altered the post-translational modification statuses of histones in vitro. Overexpression of HMGB4 in HEK 293T cells made cells more susceptible to cell death induced by topoisomerase inhibitors in an oncology drug screening array and altered variant composition of histone H3. HMGB4 regulated over 800 genes in HEK 293T cells with a p-value ≤0.013 (n = 3) in a microarray analysis and displayed strongest association with adhesion and histone H2A -processes. In neuronal and transformed cells HMGB4 regulated the expression of an oligodendrocyte marker gene PPP1R14a and other neuronal differentiation marker genes. In conclusion, our data suggests that HMGB4 is a factor that regulates chromatin and expression of neuronal differentiation markers.


Asunto(s)
Cromatina/metabolismo , Regulación de la Expresión Génica , Proteínas HMGB/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Neurogénesis , Neuronas/fisiología , Animales , Línea Celular , Perfilación de la Expresión Génica , Humanos , Ratones , Análisis por Micromatrices , Ratas
12.
Cell Rep ; 12(7): 1069-70, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26287751

RESUMEN

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.


Asunto(s)
Macrófagos/metabolismo , Espermatogénesis , Espermatogonias/citología , Nicho de Células Madre , Testículo/citología , Animales , Masculino
13.
Artículo en Inglés | MEDLINE | ID: mdl-25953399

RESUMEN

This workshop reviewed the current science to inform and recommend the best evidence-based approaches on the use of germ cell genotoxicity tests. The workshop questions and key outcomes were as follows. (1) Do genotoxicity and mutagenicity assays in somatic cells predict germ cell effects? Limited data suggest that somatic cell tests detect most germ cell mutagens, but there are strong concerns that dictate caution in drawing conclusions. (2) Should germ cell tests be done, and when? If there is evidence that a chemical or its metabolite(s) will not reach target germ cells or gonadal tissue, it is not necessary to conduct germ cell tests, notwithstanding somatic outcomes. However, it was recommended that negative somatic cell mutagens with clear evidence for gonadal exposure and evidence of toxicity in germ cells could be considered for germ cell mutagenicity testing. For somatic mutagens that are known to reach the gonadal compartments and expose germ cells, the chemical could be assumed to be a germ cell mutagen without further testing. Nevertheless, germ cell mutagenicity testing would be needed for quantitative risk assessment. (3) What new assays should be implemented and how? There is an immediate need for research on the application of whole genome sequencing in heritable mutation analysis in humans and animals, and integration of germ cell assays with somatic cell genotoxicity tests. Focus should be on environmental exposures that can cause de novo mutations, particularly newly recognized types of genomic changes. Mutational events, which may occur by exposure of germ cells during embryonic development, should also be investigated. Finally, where there are indications of germ cell toxicity in repeat dose or reproductive toxicology tests, consideration should be given to leveraging those studies to inform of possible germ cell genotoxicity.


Asunto(s)
Células Germinativas , Mutación de Línea Germinal , Mutágenos/toxicidad , Animales , Análisis Mutacional de ADN/métodos , Análisis Mutacional de ADN/normas , Educación , Estudio de Asociación del Genoma Completo/métodos , Estudio de Asociación del Genoma Completo/normas , Células Germinativas/metabolismo , Células Germinativas/patología , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/normas , Humanos , Pruebas de Mutagenicidad/métodos , Pruebas de Mutagenicidad/normas , Medición de Riesgo
14.
Nat Commun ; 5: 3812, 2014 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-24818823

RESUMEN

One of the most remarkable chromatin remodelling processes occurs during spermiogenesis, the post-meiotic phase of sperm development during which histones are replaced with sperm-specific protamines to repackage the genome into the highly compact chromatin structure of mature sperm. Here we identify Chromodomain helicase DNA binding protein 5 (Chd5) as a master regulator of the histone-to-protamine chromatin remodelling process. Chd5 deficiency leads to defective sperm chromatin compaction and male infertility in mice, mirroring the observation of low CHD5 expression in testes of infertile men. Chd5 orchestrates a cascade of molecular events required for histone removal and replacement, including histone 4 (H4) hyperacetylation, histone variant expression, nucleosome eviction and DNA damage repair. Chd5 deficiency also perturbs expression of transition proteins (Tnp1/Tnp2) and protamines (Prm1/2). These findings define Chd5 as a multi-faceted mediator of histone-to-protamine replacement and depict the cascade of molecular events underlying this process of extensive chromatin remodelling.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Cromatina/metabolismo , ADN Helicasas/genética , Infertilidad Masculina/genética , Espermatogénesis/genética , Espermatozoides/metabolismo , Acetilación , Animales , Proteínas Cromosómicas no Histona/metabolismo , ADN Helicasas/metabolismo , Reparación del ADN , Histonas/metabolismo , Infertilidad Masculina/metabolismo , Masculino , Ratones , Microscopía Electrónica de Transmisión , Protaminas/metabolismo , Testículo/metabolismo
15.
Microsc Microanal ; 20(4): 1304-11, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24834474

RESUMEN

High quality fixation often inactivates epitopes and gentler fixation can fail to preserve biological structure at the required resolution. For studies of male reproduction, immunofluorescence techniques using paraformaldehyde fixation associated with paraffin as an embedding medium gives good epitope preservation, although the cell becomes morphologically compromised. On the other hand, glutaraldehyde associated with a plastic resin has been used with success to recognize and distinguish each spermatogonial cell subtype, but the antigenic sites become inaccessible to antibodies. Here we describe a new method that provides excellent morphological details of testicular cells while preserving the binding capacity of epitopes. Using a combination of glutaraldehyde and paraformaldehyde as a fixative and LR White resin for embedding, we show that it is possible to clearly recognize spermatogonial subtypes (Aund, A-A4, In and B spermatogonia) on 1-µm thick-sections and to label epitopes such as bromodeoxyuridine, a marker used for cellular cycle studies in the testis. The information gained from this procedure can be critical for understanding spermatogonial process of self-renewal and differentiation.


Asunto(s)
Espermatogonias/citología , Coloración y Etiquetado/métodos , Testículo/citología , Adhesión del Tejido/métodos , Fijación del Tejido/métodos , Animales , Masculino , Ratones Endogámicos C57BL
16.
PLoS One ; 9(4): e93311, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24691397

RESUMEN

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.


Asunto(s)
Antineoplásicos Alquilantes/efectos adversos , Ciclofosfamida/efectos adversos , Exposición Materna , Folículo Ovárico/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Espermatogénesis/efectos de los fármacos , Neoplasias Testiculares/etiología , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Folículo Ovárico/efectos de la radiación , Ovario/efectos de los fármacos , Ovario/metabolismo , Ovario/patología , Ovario/efectos de la radiación , Embarazo , Recuento de Espermatozoides , Espermatogénesis/efectos de la radiación , Testículo/efectos de los fármacos , Testículo/metabolismo , Testículo/patología , Testículo/efectos de la radiación
17.
Fertil Steril ; 101(1): 3-13, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24314923

RESUMEN

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.


Asunto(s)
Células Madre Adultas/fisiología , Células Germinativas/fisiología , Regeneración/fisiología , Espermatogénesis/fisiología , Células Madre Adultas/trasplante , Animales , Femenino , Células Germinativas/trasplante , Humanos , Masculino , Técnicas Reproductivas Asistidas/tendencias , Espermatozoides/fisiología , Espermatozoides/trasplante
18.
Fertil Steril ; 100(5): 1180-6, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24012199

RESUMEN

Treatment of cancer with chemo- or radiotherapy causes reduction of sperm counts often to azoospermic levels that may persist for several years or be permanent. The time course of declines in sperm count can be predicted by the sensitivity of germ cells, with differentiating spermatogonia being most sensitive, and the known kinetics of recovery. Recovery from oligo- or azoospermia is more variable and depends on whether there is killing of stem cells and alteration of the somatic environment that normally supports differentiation of stem cells. Of the cytotoxic therapeutic agents, radiation and most alkylating drugs are the most potent at producing long-term azoospermia. Most of the newer biologic targeted therapies, except those used to target radioisotopes or toxins to cells, seem to have only modest effects, mostly on the endocrine aspects of the male reproductive system; however, their effects when used in combination with cytotoxic agents have not been well studied.


Asunto(s)
Antineoplásicos/efectos adversos , Azoospermia/etiología , Neoplasias/terapia , Oligospermia/etiología , Traumatismos por Radiación/etiología , Espermatogénesis , Espermatozoides , Azoospermia/inducido químicamente , Azoospermia/fisiopatología , Azoospermia/prevención & control , Humanos , Masculino , Terapia Molecular Dirigida , Oligospermia/inducido químicamente , Oligospermia/fisiopatología , Oligospermia/prevención & control , Traumatismos por Radiación/fisiopatología , Radioterapia/efectos adversos , Recuperación de la Función , Medición de Riesgo , Factores de Riesgo , Recuento de Espermatozoides , Espermatogénesis/efectos de los fármacos , Espermatogénesis/efectos de la radiación , Espermatozoides/efectos de los fármacos , Espermatozoides/patología , Espermatozoides/efectos de la radiación , Factores de Tiempo
19.
Reproduction ; 146(4): 363-76, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23884860

RESUMEN

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.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Células Intersticiales del Testículo/metabolismo , Células de Sertoli/metabolismo , Espermatogénesis/fisiología , Espermatogonias/fisiología , Factor de Células Madre/metabolismo , Testosterona/farmacología , Andrógenos/farmacología , Animales , Diferenciación Celular/efectos de la radiación , Células Cultivadas , Técnicas para Inmunoenzimas , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/efectos de la radiación , Masculino , Ratas , Células de Sertoli/efectos de los fármacos , Células de Sertoli/efectos de la radiación , Espermatogénesis/efectos de los fármacos , Espermatogénesis/efectos de la radiación , Espermatogonias/efectos de los fármacos , Espermatogonias/efectos de la radiación
20.
Methods Mol Biol ; 927: 299-307, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22992924

RESUMEN

Male germ cells in all mammals are arranged within the seminiferous epithelium of the testicular tubules in a set of well-defined cell associations called stages. The cellular associations found in these stages and characteristics of the cells used to identify the stages have been well described. Here we present a binary decision key roadmap for identifying stages and present several examples of how staging tubules can be used to better assess the developmental profile of gene expression during spermatogenesis and defects in spermatogenesis arising in pathological conditions resulting from genetic mutations in mice. In particular, when one or more cells of a cellular association cannot be clearly identified or are missing, the cell types that should be present can be precisely identified by knowledge of the approximate or exact stage of the tubule cross section.


Asunto(s)
Túbulos Seminíferos/citología , Espermatogénesis/fisiología , Acrosoma/metabolismo , Acrosoma/patología , Animales , Ciclo Celular/fisiología , Diferenciación Celular/genética , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/genética , Ácido Graso Desaturasas/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Noqueados , Epitelio Seminífero/citología , Epitelio Seminífero/patología , Túbulos Seminíferos/patología , Espermátides/metabolismo , Espermátides/patología , Espermatogénesis/genética , Coloración y Etiquetado/métodos , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...