Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
PLoS Biol ; 20(5): e3001618, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35536782

RESUMO

Treatment of cancer in children is increasingly successful but leaves many prepubertal boys suffering from infertility or subfertility later in life. A current strategy to preserve fertility in these boys is to cryopreserve a testicular biopsy prior to treatment with the expectation of future technologies allowing for the reintroduction of stem cells and restoration of spermatogenesis. Spermatogonial stem cells (SSCs) form the basis of male reproduction, differentiating into all germ cell types, including mature spermatozoa and can regenerate spermatogenesis following transplantation into an infertile testis. Here, we demonstrate that rat SSCs frozen for more than 20 years can be transplanted into recipient mice and produce all differentiating germ cell types. However, compared with freshly isolated cells or those frozen for a short period of time, long-frozen cells do not colonize efficiently and showed reduced production of spermatids. Single-cell RNA sequencing revealed similar profiles of gene expression changes between short- and long-frozen cells as compared with fresh immediately after thawing. Conversely, following transplantation, long-frozen samples showed enhanced stem cell signaling in the undifferentiated spermatogonia compartment, consistent with self-renewal and a lack of differentiation. In addition, long-frozen samples showed fewer round spermatids with detectable protamine expression, suggesting a partial block of spermatogenesis after meiosis resulting in a lack of elongating spermatids. These findings strongly suggest that prolonged cryopreservation can impact the success of transplantation to produce spermatogenesis, which may not be revealed by analysis of the cells immediately after thawing. Our analysis uncovered persistent effects of long-term freezing not found in other cryopreservation studies that lacked functional regeneration of the tissue and this phenomenon must be accounted for any future therapeutic application.


Assuntos
Células-Tronco Germinativas Adultas , Espermatogênese , Animais , Criopreservação/métodos , Humanos , Masculino , Camundongos , Ratos , Espermatogênese/genética , Espermatogônias/metabolismo , Células-Tronco , Testículo
2.
Biol Reprod ; 105(2): 503-518, 2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-33959758

RESUMO

Retinoic acid (RA) induces spermatogonial differentiation, but the mechanism by which it operates remains largely unknown. We developed a germ cell culture assay system to study genes involved in spermatogonial differentiation triggered by RA. Stimulated by RA 8 (Stra8), a RA-inducible gene, is indispensable for meiosis initiation, and its deletion results in a complete block of spermatogenesis at the pre-leptotene/zygotene stage. To interrogate the role of Stra8 in RA mediated differentiation of spermatogonia, we derived germ cell cultures from the neonatal testis of both wild type and Stra8 knock-out mice. We provide the first evidence that Stra8 plays a crucial role in modulating the responsiveness of undifferentiated spermatogonia to RA and facilitates transition to a differentiated state. Stra8-mediated differentiation is achieved through the downregulation of a large portfolio of genes and pathways, most notably including genes involved in the spermatogonial stem cell self-renewal process. We also report here for the first time the role of transcription elongation regulator-1 like (Tcerg1l) as a downstream effector of RA-induced spermatogonial differentiation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Embrião de Mamíferos/embriologia , Camundongos/genética , Espermatogônias , Fatores de Elongação da Transcrição/genética , Tretinoína/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Masculino , Camundongos/embriologia , Fatores de Elongação da Transcrição/metabolismo
3.
Physiol Rev ; 92(2): 577-95, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22535892

RESUMO

This review addresses current understanding of the germline stem cell niche unit in mammalian testes. Spermatogenesis is a classic model of tissue-specific stem cell function relying on self-renewal and differentiation of spermatogonial stem cells (SSCs). These fate decisions are influenced by a niche microenvironment composed of a growth factor milieu that is provided by several testis somatic support cell populations. Investigations over the last two decades have identified key determinants of the SSC niche including cytokines that regulate SSC functions and support cells providing these factors, adhesion molecules that influence SSC homing, and developmental heterogeneity of the niche during postnatal aging. Emerging evidence suggests that Sertoli cells are a key support cell population influencing the formation and function of niches by secreting soluble factors and possibly orchestrating contributions of other support cells. Investigations with mice have shown that niche influence on SSC proliferation differs during early postnatal development and adulthood. Moreover, there is mounting evidence of an age-related decline in niche function, which is likely influenced by systemic factors. Defining the attributes of stem cell niches is key to developing methods to utilize these cells for regenerative medicine. The SSC population and associated niche comprise a valuable model system for study that provides fundamental knowledge about the biology of tissue-specific stem cells and their capacity to sustain homeostasis of regenerating tissue lineages. While the stem cell is essential for maintenance of all self-renewing tissues and has received considerable attention, the role of niche cells is at least as important and may prove to be more receptive to modification in regenerative medicine.


Assuntos
Espermatogênese , Espermatozoides/crescimento & desenvolvimento , Nicho de Células-Tronco , Células-Tronco/fisiologia , Testículo/crescimento & desenvolvimento , Envelhecimento , Animais , Proliferação de Células , Citocinas/fisiologia , Humanos , Masculino , Camundongos , Ratos , Células de Sertoli , Espermatozoides/citologia , Testículo/citologia
4.
Biol Reprod ; 99(1): 52-74, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29617903

RESUMO

Spermatogonial stem cells (SSCs) are the most primitive spermatogonia in the testis and have an essential role to maintain highly productive spermatogenesis by self-renewal and continuous generation of daughter spermatogonia that differentiate into spermatozoa, transmitting genetic information to the next generation. Since the 1950s, many experimental methods, including histology, immunostaining, whole-mount analyses, and pulse-chase labeling, had been used in attempts to identify SSCs, but without success. In 1994, a spermatogonial transplantation method was reported that established a quantitative functional assay to identify SSCs by evaluating their ability to both self-renew and differentiate to spermatozoa. The system was originally developed using mice and subsequently extended to nonrodents, including domestic animals and humans. Availability of the functional assay for SSCs has made it possible to develop culture systems for their ex vivo expansion, which dramatically advanced germ cell biology and allowed medical and agricultural applications. In coming years, SSCs will be increasingly used to understand their regulation, as well as in germline modification, including gene correction, enhancement of male fertility, and conversion of somatic cells to biologically competent male germline cells.


Assuntos
Células-Tronco Germinativas Adultas/citologia , Espermatogênese/fisiologia , Espermatogônias/citologia , Testículo/citologia , Animais , Humanos , Masculino
5.
J Urol ; 194(5): 1420-4, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26032139

RESUMO

PURPOSE: Cryopreservation of testicular tissue with subsequent reimplantation after therapy has the potential to preserve fertility for prepubertal boys with cancer. We present the histology and feasibility of testicular tissue procurement for this novel approach. MATERIALS AND METHODS: We performed a prospective cohort study of boys at significant risk for treatment associated gonadotoxicity who were eligible for an experimental research protocol between 2008 and 2011. Open testicular biopsy was performed while the patients were anesthetized for another treatment related procedure. Half of the specimen was reserved for cryopreservation, while the other half was used for research purposes. Semithin sections of the biopsy specimens were evaluated for histological features and compared to age adjusted reference values. RESULTS: A total of 34 boys underwent biopsy between March 2008 and October 2011. Of the patients 29 had solid tumors and 5 underwent hematopoietic stem cell transplantation for benign disease. A total of 27 patients had adequate tissue for histological analysis. Median patient age was 8.7 years (IQR 2.2 to 11.5). All children had either normal (81.5% of patients) or increased (18.5%) numbers of germ cells per tubule for their age. However, 5 of 26 patients (19%) older than 6 months had no evidence of adult dark spermatogonia and 9 of 16 (56%) older than 6 years had no evidence of primary spermatocytes on biopsy, which would be expected based on age norms. These findings are suggestive of abnormal germ cell maturation. CONCLUSIONS: The preliminary histological findings of abnormal spermatogenesis maturation in the testes of prepubertal boys with cancer warrants further investigation.


Assuntos
Transplante de Células/métodos , Preservação da Fertilidade/métodos , Fertilidade , Infertilidade Masculina/prevenção & controle , Neoplasias/diagnóstico , Testículo/patologia , Adolescente , Biópsia , Criança , Pré-Escolar , Criopreservação , Seguimentos , Humanos , Infertilidade Masculina/diagnóstico , Infertilidade Masculina/patologia , Masculino , Neoplasias/metabolismo , Neoplasias/terapia , Estudos Prospectivos , Contagem de Espermatozoides , Espermatogênese , Fatores de Tempo
6.
Pediatr Blood Cancer ; 61(9): 1673-8, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24777742

RESUMO

BACKGROUND: Infertility is an unfortunate treatment-related consequence for some pediatric malignancies as well as some non-malignant conditions treated with stem cell transplant. Unlike pubertal males, prepubertal males cannot produce semen for cryopreservation. This manuscript reports on the acceptability and safety of a multi-institutional protocol for offering testicular tissue cryopreservation to families of prepubertal male children at highest risk for infertility. Data on decision influences, decision-making control, and emotional state when considering this option are described. PROCEDURE: Prepubertal males facing gonadotoxic therapy were offered testicular cryopreservation. Post-biopsy, patients were followed for acute side effects. In addition, parents and patients were asked to complete questionnaires, whether or not they chose to cryopreserve tissue. RESULTS: Seventy-four prepubertal male children were approached. Fifty-seven families (77%) consented to the testicular biopsy; 48 of 57 underwent the procedure. There was one post-operative side effect. Parents who agreed to testicular cryopreservation and those that did not felt in control of this decision. Parents who consented to the biopsy and refusers were not deterred by the experimental nature of the protocol. An important decision-making influence was the risk of the biopsy. CONCLUSION: Biopsy and cryopreservation of testicular tissue from prepubertal male children was performed successfully and safely at three institutions. Parents faced with this option at diagnosis can make an informed decision and weigh carefully the risks and benefits. Although asked to make a decision soon after they were given a difficult diagnosis, parents uniformly felt in control of this decision.


Assuntos
Criopreservação , Tomada de Decisões , Infertilidade Masculina/prevenção & controle , Pais/psicologia , Preservação do Sêmen/psicologia , Testículo/citologia , Preservação de Tecido , Adolescente , Criança , Pré-Escolar , Seguimentos , Humanos , Lactente , Infertilidade Masculina/etiologia , Infertilidade Masculina/psicologia , Masculino , Neoplasias/complicações , Neoplasias/terapia
7.
Proc Natl Acad Sci U S A ; 108(31): 12740-5, 2011 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-21768389

RESUMO

MicroRNAs (miRs) play a key role in the control of gene expression in a wide array of tissue systems, where their functions include the regulation of self-renewal, cellular differentiation, proliferation, and apoptosis. However, the functional importance of individual miRs in controlling spermatogonial stem cell (SSC) homeostasis has not been investigated. Using high-throughput sequencing, we profiled the expression of miRs in the Thy1(+) testis cell population, which is highly enriched for SSCs, and the Thy1(-) cell population, composed primarily of testis somatic cells. In addition, we profiled the global expression of miRs in cultured germ cells, also enriched for SSCs. Our results demonstrate that miR-21, along with miR-34c, -182, -183, and -146a, are preferentially expressed in the Thy1(+) SSC-enriched population, compared with Thy1(-) somatic cells. Importantly, we demonstrate that transient inhibition of miR-21 in SSC-enriched germ cell cultures increased the number of germ cells undergoing apoptosis and significantly reduced the number of donor-derived colonies of spermatogenesis formed from transplanted treated cells in recipient mouse testes, indicating that miR-21 is important in maintaining the SSC population. Moreover, we show that in SSC-enriched germ cell cultures, miR-21 is regulated by the transcription factor ETV5, known to be critical for SSC self-renewal.


Assuntos
Proliferação de Células , MicroRNAs/genética , Espermatogônias/citologia , Células-Tronco/metabolismo , Animais , Apoptose/genética , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Perfilação da Expressão Gênica , Biblioteca Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Espermatogênese/genética , Espermatogônias/metabolismo , Transplante de Células-Tronco/métodos , Testículo/citologia , Testículo/metabolismo , Antígenos Thy-1/genética , Antígenos Thy-1/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
8.
Hum Reprod ; 27(5): 1249-59, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22416011

RESUMO

BACKGROUND: Approximately 80% of childhood cancers can now be cured but a side effect of treatment results in about one-third of the surviving boys being infertile or severely subfertile when they reach reproductive age. Currently, more than 1 in 5000 men of reproductive age who are childhood cancer survivors suffer from this serious quality of life problem. It is possible to obtain a testicular biopsy before treatment to preserve the spermatogonial stem cells (SSCs) of the male by cryopreservation, but the results of long-term storage of SSCs on their subsequent functional ability to generate normal offspring has not been examined in any mammalian species. Moreover, it will be necessary to increase the number of these cryopreserved SSCs to remove any contaminating malignant cells and assure regeneration of spermatogenesis. METHODS AND RESULTS: In this report, we demonstrate that long-term cryopreservation (>14 years) of testis cells from mouse, rat, rabbit and baboon safeguards SSC viability, and that these cells can colonize the seminiferous tubules of recipient testes. Moreover, mouse and rat SSCs can be cultured and re-establish complete spermatogenesis, and fertile mouse progeny without apparent genetic or epigenetic errors were generated by the sperm produced. CONCLUSIONS: These findings provide a platform for fertility preservation in prepubertal boys undergoing gonadotoxic treatments.


Assuntos
Criopreservação , Preservação da Fertilidade/métodos , Espermatogônias/citologia , Células-Tronco , Animais , Feminino , Fertilidade , Masculino , Camundongos , Papio/fisiologia , Coelhos , Ratos , Injeções de Esperma Intracitoplásmicas , Espermatogônias/transplante , Testículo/citologia , Fatores de Tempo
9.
FASEB J ; 25(8): 2604-14, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21525489

RESUMO

Previous studies suggest that exogenous factors crucial for spermatogonial stem cell (SSC) self-renewal are conserved among several mammalian species. Since glial cell line-derived neurotrophic factor (GDNF) and fibroblast growth factor 2 (FGF2) are critical for rodent SSC self-renewal, we hypothesized that they might promote self-renewal of nonrodent SSCs. Therefore, we cultured testicular germ cells from prepubertal rabbits in the presence of GDNF and FGF2 and found they proliferated indefinitely as cellular clumps that displayed characteristics previously identified for rodent SSCs. The rabbit germ cells could not be maintained on mouse embryonic fibroblast (STO) feeders that support rodent SSC self-renewal in vitro but were rather supported on mouse yolk sac-derived endothelial cell (C166) feeder layers. Proliferation of rabbit germ cells was dependent on GDNF. Of critical importance was that clump-forming rabbit germ cells colonized seminiferous tubules of immunodeficient mice, proliferated for at least 6 mo, while retaining an SSC phenotype in the testes of recipient mice, indicating that they were rabbit SSCs. This study demonstrates that GDNF is a mitogenic factor promoting self-renewal that is conserved between rodent and rabbit SSCs; with an evolutionary separation of ∼ 60 million years. These findings provide a foundation to study the mechanisms governing SSC self-renewal in nonrodent species.


Assuntos
Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Espermatogônias/citologia , Espermatogônias/efeitos dos fármacos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/citologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Masculino , Camundongos , Coelhos , Especificidade da Espécie , Espermatogênese/efeitos dos fármacos , Espermatogônias/transplante , Transplante de Células-Tronco
10.
Proc Natl Acad Sci U S A ; 106(51): 21672-7, 2009 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-20018717

RESUMO

In the human testis, beginning at approximately 2 months of age, gonocytes are replaced by adult dark (Ad) and pale (Ap) spermatogonia that make up the spermatogonial stem cell (SSC) pool. In mice, the SSC pool arises from gonocytes approximately 6 days after birth. During puberty in both species, complete spermatogenesis is established by cells that differentiate from SSCs. Essentially pure populations of prepubertal human spermatogonia and mouse gonocytes were selected from testis biopsies and validated by confirming the presence of specific marker proteins in cells. Stem cell potential of germ cells was demonstrated by transplantation to mouse testes, following which the cells migrated to the basement membrane of the seminiferous tubule and were maintained similar to SSCs. Differential gene expression profiles generated between germ cells and testis somatic cells demonstrated that expression of genes previously identified as SSC and spermatogonial-specific markers (e.g., zinc-finger and BTB-domain containing 16, ZBTB16) was greatly elevated in both human spermatogonia and mouse gonocytes compared to somatic cells. Several genes were expressed at significantly higher levels in germ cells of both species. Most importantly, genes known to be essential for mouse SSC self-renewal (e.g., Ret proto-oncogene, Ret; GDNF-family receptor alpha1, Gfr alpha1; and B-cell CLL/lymphoma 6, member B, Bcl6b) were more highly expressed in both prepubertal human spermatogonia and mouse gonocytes than in somatic cells. The results indicate remarkable conservation of gene expression, notably for self-renewal genes, in these prepubertal germline cells between two species that diverged phylogenetically approximately 75 million years ago.


Assuntos
Perfilação da Expressão Gênica , Células Germinativas , Maturidade Sexual , Espermatogônias/metabolismo , Células-Tronco/citologia , Animais , Transplante de Células , Humanos , Masculino , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Proto-Oncogene Mas , Células-Tronco/metabolismo
11.
Biol Reprod ; 85(6): 1114-23, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21816850

RESUMO

Insight regarding mechanisms controlling gene expression in the spermatogonial stem cell (SSC) will improve our understanding of the processes regulating spermatogenesis and aid in treating problems associated with male infertility. In the present study, we explored the global gene expression profiles of the glial cell line-derived neurotrophic factor (GDNF)-regulated transcription factors Ets (E-twenty-six) variant gene 5 (Etv5); B-cell chronic lymphocytic leukemia (CLL)/lymphoma 6, member B (Bcl6b); and POU domain, class-3 transcription factor 1 (Pou3f1). We reasoned that these three factors may function as a core set of transcription factors, regulating genes responsible for maintaining the SSC population. Using transient siRNA oligonucleotides to individually target Etv5, Bcl6b, and Pou3f1 within mouse SSC cultures, we examined changes to the global gene expression profiles associated with these transcription factors. Only modest overlaps in the target genes regulated by the three factors were noted, but ETV5 was found to be a critical downstream regulator of GDNF signaling that mediated the expression of several known SSC self-renewal related genes, including Bcl6b and LIM homeobox 1 (Lhx1). Notably, ETV5 was identified as a regulator of Brachyury (T) and CXC chemokine receptor, type 4 (Cxcr4), and we showed that ETV5 binding to the Brachyury (T) gene promoter region is associated with an active state of transcription. Moreover, in vivo transplantation of SSCs following silencing of Brachyury (T) significantly reduced the number of donor cell-derived colonies formed within recipient mouse testes. These results suggest Brachyury is of biological importance and functions as part of GDNF/ETV5 signaling to promote self-renewal of mouse SSCs cultured in vitro.


Assuntos
Células-Tronco Adultas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Fetais/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Espermatogônias/metabolismo , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator 6 de Transcrição de Octâmero/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Proteína com Dedos de Zinco da Leucemia Promielocítica , Interferência de RNA , Proteínas Repressoras/metabolismo
12.
Biol Reprod ; 84(4): 698-706, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21191109

RESUMO

The development of techniques to maintain the spermatogonial stem cell (SSC) in vivo and in vitro for extended periods essentially allows for the indefinite continuation of an individual germline. Recent evidence indicates that the aging of male reproductive function is due to failure of the SSC niche. SSCs are routinely cultured for 6 mo, and no apparent effect of culture over this period has been observed. To determine the effects of SSC aging, we utilized an in vitro culture system, followed by quantitative transplantation experiments. After culture for 6 mo, SSCs that had been aged in vivo for 1500 days had a slower proliferation rate than SSCs that were aged in vivo to 8 or 300 days. Examination of methylation patterns revealed no apparent difference in DNA methylation between SSCs that were aged 8, 300, or 1500 days before culture. Long-term culture periods resulted in a loss of stem cell potential without an obvious change in the visual appearance of the culture. DNA microarray analysis of in vivo- and in vitro-aged SSCs identified the differential expression of several genes important for SSC function, including B-cell CLL/lymphoma 6, member B (Bcl6b), Lim homeobox protein 1 (Lhx1), and thymus cell antigen 1, theta (Thy1). Collectively, these data indicate that, although both in vitro and in vivo aging are detrimental to SSC function, in vitro aging results in greater loss of function, potentially due to a decrease in core SSC self-renewal gene expression and an increase in germ cell differentiation gene expression.


Assuntos
Células-Tronco Adultas/patologia , Células-Tronco Adultas/fisiologia , Envelhecimento/patologia , Envelhecimento/fisiologia , Espermatogônias/patologia , Espermatogônias/fisiologia , Envelhecimento/genética , Animais , Sequência de Bases , Senescência Celular/genética , Senescência Celular/fisiologia , Metilação de DNA , Primers do DNA/genética , Feminino , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Técnicas In Vitro , Masculino , Camundongos , Camundongos Transgênicos , Gravidez , Proteínas Recombinantes/genética , Injeções de Esperma Intracitoplásmicas
13.
Cell Prolif ; 54(1): e12933, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33107118

RESUMO

OBJECTIVES: Fibroblast growth factor 9 (FGF9) is expressed by somatic cells in the seminiferous tubules, yet little information exists about its role in regulating spermatogonial stem cells (SSCs). MATERIALS AND METHODS: Fgf9 overexpression lentivirus was injected into mouse testes, and PLZF immunostaining was performed to investigate the effect of FGF9 on spermatogonia in vivo. Effect of FGF9 on SSCs was detected by transplanting cultured germ cells into tubules of testes. RNA-seq of bulk RNA and single cell was performed to explore FGF9 working mechanisms. SB203580 was used to disrupt p38 MAPK pathway. p38 MAPK protein expression was detected by Western blot and qPCR was performed to determine different gene expression. Small interfering RNA (siRNA) was used to knock down Etv5 gene expression in germ cells. RESULTS: Overexpression of Fgf9 in vivo resulted in arrested spermatogenesis and accumulation of undifferentiated spermatogonia. Exposure of germ cell cultures to FGF9 resulted in larger numbers of SSCs over time. Inhibition of p38 MAPK phosphorylation negated the SSC growth advantage provided by FGF9. Etv5 and Bcl6b gene expressions were enhanced by FGF9 treatment. Gene knockdown of Etv5 disrupted the growth effect of FGF9 in cultured SSCs along with downstream expression of Bcl6b. CONCLUSIONS: Taken together, these data indicate that FGF9 is an important regulator of SSC proliferation, operating through p38 MAPK phosphorylation and upregulating Etv5 and Bcl6b in turn.


Assuntos
Fator 9 de Crescimento de Fibroblastos/metabolismo , Espermatogônias/metabolismo , Células-Tronco/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/metabolismo , Espermatogônias/citologia , Células-Tronco/citologia
14.
Biol Reprod ; 83(3): 427-33, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20505165

RESUMO

Homeostasis of many tissues is maintained by self-renewal and differentiation of stem cells. Spermatogenesis is one such system relying on the activity of spermatogonial stem cells (SSCs). Several key regulators of SSC self-renewal have been identified, yet knowledge of molecules that control SSC differentiation is undefined. In this study, we found that transient impairment of STAT3 signaling enhances SSC self-renewal in vitro without affecting general spermatogonial proliferation, indicating an alteration in the balance of SSC fate decisions that inhibited differentiation. Confirming this observation, short hairpin RNA-mediated stable reduction of STAT3 expression in cultured SSCs abolished their ability to differentiate beyond the undifferentiated spermatogonial stage following transplantation into recipient testes. Collectively, these results demonstrate that STAT3 promotes the differentiation of SSCs. In contrast, STAT3 plays a central role in maintaining self-renewal of mouse embryonic stem cells, and STAT signaling is essential for self-renewal of male germline stem cells in Drosophila.


Assuntos
Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/fisiologia , Espermatogênese/fisiologia , Espermatogônias/metabolismo , Análise de Variância , Animais , Western Blotting , Proliferação de Células , Células Cultivadas , Citometria de Fluxo , Imuno-Histoquímica , Masculino , Camundongos , Fosforilação/fisiologia , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espermatogônias/transplante , Transfecção
15.
Biol Reprod ; 82(6): 1103-11, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20181621

RESUMO

Continual spermatogenesis relies on a pool of spermatogonial stem cells (SSCs) that possess the capacity for self-renewal and differentiation. Maintenance of this pool depends on survival of SSCs throughout the lifetime of a male. Response to extrinsic stimulation from glial cell line-derived neurotrophic factor (GDNF), mediated by the PIK3/AKT signaling cascade, is a key pathway of SSC survival. In this study, we found that expression of the POU domain transcription factor POU3F1 in cultured SSCs is up-regulated via this mechanism. Reduction of Pou3f1 gene expression by short interfering RNA (siRNA) treatment induced apoptosis in cultured germ cell populations, and transplantation analyses revealed impaired SSC maintenance in vitro. POU3F1 expression was localized to spermatogonia in cross-sections of prepubertal and adult testes, implying a similar role in vivo. Through comparative analyses, we found that expression of POU5F1, another POU transcription factor implicated as essential for SSC self-renewal, is not regulated by GDNF in cultured SSCs. Transplantation analyses following siRNA treatment showed that POU5F1 expression is not essential for SSC maintenance in vitro. Additionally, expression of NODAL, a putative autocrine regulator of POU5F1 expression in mouse germ cells, could not be detected in SSCs isolated from testes or cultured SSCs. Collectively, these results indicate that POU3F1, but not POU5F1, is an intrinsic regulator of GDNF-induced survival and self-renewal of mouse SSCs.


Assuntos
Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Fator 6 de Transcrição de Octâmero/metabolismo , Espermatogênese , Espermatogônias/metabolismo , Células-Tronco/metabolismo , Animais , Apoptose , Comunicação Autócrina , Divisão Celular , Sobrevivência Celular , Masculino , Camundongos , Proteína Nodal/análise , Testículo/citologia , Testículo/metabolismo
16.
Biol Reprod ; 81(2): 293-301, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19369648

RESUMO

Loss-of-function mutation of the Kit gene causes a severe defect in spermatogenesis that results in infertility due to the inability of its cognate ligand, KIT ligand (KITL), to stimulate spermatogonial proliferation and differentiation. Although self-renewal of mouse spermatogonial stem cells (SSCs) depends on glial cell line-derived neurotrophic factor (GDNF), there is no unequivocal evidence that SSCs with a KIT deficiency can self-renew in vivo or in vitro. In the testis of W(v)/W(v) mice, in which the KIT tyrosine kinase activity is impaired, spermatogonia with SSC phenotype were identified. When W(v)/W(v) spermatogonia were cultured in an SSC culture system supplemented with GDNF in a 10% O(2) atmosphere, they formed clumps and proliferated continuously. An atmosphere of 10% O(2) was better than 21% O(2) to support SSC self-renewal. When W(v)/W(v) clump-forming germ cells were transplanted into testes of infertile wild-type busulfan-treated mice, they colonized the seminiferous tubules but did not differentiate. However, when transplanted into the testes of infertile W/W(v) pups, they restored spermatogenesis and produced spermatozoa, and progeny were generated using microinsemination. These results clearly show that SSCs exist in W(v)/W(v) testes and that they proliferate in vitro similar to wild-type SSCs, indicating that a functional KIT protein is not required for SSC self-renewal. Furthermore, the results indicate that a defect of KIT/KITL signaling of W(v)/W(v) SSCs does not prevent spermatogonial differentiation and spermatogenesis in some recipient strains.


Assuntos
Proteínas Proto-Oncogênicas c-kit/fisiologia , Espermatogênese , Espermatogônias/fisiologia , Células-Tronco/fisiologia , Análise de Variância , Animais , Proliferação de Células , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Proteína Ligante Fas/deficiência , Feminino , Citometria de Fluxo , Infertilidade/genética , Infertilidade/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Oxigênio , Proteínas Proto-Oncogênicas c-kit/genética , Túbulos Seminíferos/citologia , Espermatogênese/genética , Espermatogônias/citologia , Espermatogônias/crescimento & desenvolvimento , Transplante de Células-Tronco , Células-Tronco/citologia , Testículo/citologia , beta-Galactosidase/genética
17.
Stem Cells ; 26(4): 927-38, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18203673

RESUMO

Gene expression and consequent biological activity of adult tissue stem cells are regulated by signals emanating from the local microenvironment (niche). To gain insights into the molecular regulation of spermatogonial stem cells (SSCs), gene expression was characterized from SSCs isolated from their cognate niches of cryptorchid (stem cell-enriched), wild-type, and busulfan-treated (stem cell-depleted) mouse testes. Quantitative assessment of stem cell activity in each testis model was determined using an in vivo functional assay and correlated with gene expression using Affymetrix MGU74Av2 microarrays and the ChipStat algorithm optimized to detect gene expression from rare cells in complex tissues. We identified 389 stem/progenitor spermatogonia candidate genes, which exhibited significant overlap with genes expressed by embryonic, hematopoietic, and neural stem cells; enriched spermatogonia; and cultured SSCs identified in previous studies. Candidate cell surface markers identified by the microarray may facilitate the isolation and enrichment of stem and/or progenitor spermatogonia. Flow cytometric analyses confirmed the expression of chemokine receptor 2 (Ccr2) and Cd14 on a subpopulation cryptorchid testis cells (alpha6-integrin+, side scatter(lo)) enriched for SSCs. These cell surface molecules may mark progenitor spermatogonia but not SSCs because Ccr2+ and Cd14+ fractions failed to produce spermatogenesis upon transplantation to recipient testes. Functional annotation of candidate genes and subsequent immunohistochemistry revealed that proteins involved in post-transcriptional regulation are overrepresented in cryptorchid testes that are enriched for SSCs. Comparative analyses indicated that this is a recurrent biological theme among stem cells.


Assuntos
Criptorquidismo/genética , Processamento Pós-Transcricional do RNA/genética , Espermatogônias/fisiologia , Células-Tronco/fisiologia , Testículo/fisiologia , Animais , Criptorquidismo/metabolismo , Criptorquidismo/cirurgia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA/biossíntese , RNA/genética , Espermatogônias/transplante , Transplante de Células-Tronco/métodos
18.
Methods Mol Biol ; 2005: 205-220, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31175655

RESUMO

Spermatogonial stem cell (SSC) culture and transplantation pave the way for clinical restoration of fertility in male prepubertal cancer survivors. In this chapter we detail the steps for isolating and freezing testicular tissue along with protocols for the subsequent recovery from cryopreservation and transplantation of cells into a recipient testis. Transplantation of cultured or thawed SSCs provides not only a functional assay for identification of stem cells, a critical tool for the study of the germline stem cell niche in model organisms, but also a framework for reconstitution of spermatogenesis in humans. As proof of concept, the outlined methods have been performed successfully in the murine model and have the potential to be translated to clinical environments.


Assuntos
Células-Tronco Germinativas Adultas/transplante , Separação Celular/métodos , Criopreservação/métodos , Modelos Biológicos , Espermatogênese , Nicho de Células-Tronco , Transplante de Células-Tronco/métodos , Animais , Humanos , Masculino , Camundongos , Camundongos Transgênicos
19.
J Androl ; 28(2): 353-60, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17108363

RESUMO

Spermatozoa produced from spermatogonial stem cells (SSCs) are the vehicle by which genes of a male are passed to the next generation. A single SSC has the ability to self-renew and produce thousands of spermatozoa; therefore, it is an ideal target for genetic modification to efficiently generate transgenic animals in mammalian species. Rats are an important model organism for biological research; however, gene function studies have been difficult because of a limited ability to generate transgenic animals. Transgenic rat production through SSCs offers a means to overcome this obstacle. Because SSCs divide slowly both in vivo and in vitro, lentiviral vectors may be an ideal method for introducing stable genetic modification. Using a lentiviral vector, an enhanced green fluorescent protein (eGFP) transgene was introduced into the genome of cultured rat SSCs, which were microinjected into testes of immunodeficient mice to assess transduction efficiency. Approximately 40% of rat SSCs exposed to the lentiviral vector overnight carried the eGFP transgene and generated colonies of spermatogenesis. When transduced SSCs were transplanted into recipient rat testes, in which endogenous germ cells had been decreased but not eliminated by busulfan treatment, approximately 6% of offspring were transgenic. The transgene was stably integrated into the donor SSC genome and transmitted to and expressed by progeny in subsequent generations. Thus, lentiviral transduction of SSCs followed by transplantation is an effective means for generating transgenic rats through the male germline, and this approach may be applicable to other species in which existing methods are inadequate or not applicable.


Assuntos
Animais Geneticamente Modificados , Espermatogônias/transplante , Transplante de Células-Tronco/métodos , Animais , Proteínas de Fluorescência Verde/genética , Lentivirus/genética , Masculino , Linhagem , Ratos , Ratos Sprague-Dawley , Transdução Genética , Transfecção
20.
Methods Enzymol ; 419: 259-82, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17141059

RESUMO

The biological activities of spermatogonial stem cells (SSCs) are the foundation for spermatogenesis and thus sustained male fertility. Therefore, understanding the mechanisms governing their ability to both self-renew and differentiate is essential. Moreover, because SSCs are the only adult stem cell to contribute genetic information to the next generation, they are an excellent target for genetic modification. In this chapter, we discuss two important approaches to investigate SSCs and their cognate niche microenvironment in the mouse, the SSC transplantation assay and the long-term serum-free SSC culture method. These techniques can be used to enhance our understanding of SSC biology as well as to produce genetically modified animals.


Assuntos
Células-Tronco Adultas , Espermatogônias , Animais , Técnicas de Cultura de Células/métodos , Humanos , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA