Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Hum Reprod ; 25(1): 158-67, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19815622

ABSTRACT

BACKGROUND: Given the significant drawbacks of using human embryonic stem (hES) cells for regenerative medicine, the search for alternative sources of multipotent cells is ongoing. Studies in mice have shown that multipotent ES-like cells can be derived from neonatal and adult testis. Here we report the derivation of ES-like cells from adult human testis. METHODS: Testis material was donated for research by four men undergoing bilateral castration as part of prostate cancer treatment. Testicular cells were cultured using StemPro medium. Colonies that appeared sharp edged and compact were collected and subcultured under hES-specific conditions. Molecular characterization of these colonies was performed using RT-PCR and immunohistochemistry. (Epi)genetic stability was tested using bisulphite sequencing and karyotype analysis. Directed differentiation protocols in vitro were performed to investigate the potency of these cells and the cells were injected into immunocompromised mice to investigate their tumorigenicity. RESULTS: In testicular cell cultures from all four men, sharp-edged and compact colonies appeared between 3 and 8 weeks. Subcultured cells from these colonies showed alkaline phosphatase activity and expressed hES cell-specific genes (Pou5f1, Sox2, Cripto1, Dnmt3b), proteins and carbohydrate antigens (POU5F1, NANOG, SOX2 and TRA-1-60, TRA-1-81, SSEA4). These ES-like cells were able to differentiate in vitro into derivatives of all three germ layers including neural, epithelial, osteogenic, myogenic, adipocyte and pancreatic lineages. The pancreatic beta cells were able to produce insulin in response to glucose and osteogenic-differentiated cells showed deposition of phosphate and calcium, demonstrating their functional capacity. Although we observed small areas with differentiated cell types of human origin, we never observed extensive teratomas upon injection of testis-derived ES-like cells into immunocompromised mice. CONCLUSIONS: Multipotent cells can be established from adult human testis. Their easy accessibility and ethical acceptability as well as their non-tumorigenic and autogenic nature make these cells an attractive alternative to human ES cells for future stem cell therapies.


Subject(s)
Cell Culture Techniques , Embryonic Stem Cells/cytology , Multipotent Stem Cells/cytology , Testis/cytology , Aged , Aged, 80 and over , Animals , Cell Differentiation , Cell Proliferation , Embryonic Stem Cells/metabolism , Humans , Karyotyping , Male , Mice , Middle Aged , Multipotent Stem Cells/metabolism
2.
Biol Reprod ; 63(6): 1893-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11090462

ABSTRACT

Using immunohistochemistry, the expression of the D-type cyclin proteins was studied in the developing and adult mouse testis. Both during testicular development and in adult testis, cyclin D(1) is expressed only in proliferating gonocytes and spermatogonia, indicating a role for cyclin D(1) in spermatogonial proliferation, in particular during the G(1)/S phase transition. Cyclin D(2) is first expressed at the start of spermatogenesis when gonocytes produce A(1) spermatogonia. In the adult testis, cyclin D(2) is expressed in spermatogonia around stage VIII of the seminiferous epithelium when A(al) spermatogonia differentiate into A(1) spermatogonia and also in spermatocytes and spermatids. To further elucidate the role of cyclin D(2) during spermatogenesis, cyclin D(2) expression was studied in vitamin A-deficient testis. Cyclin D(2) was not expressed in the undifferentiated A spermatogonia in vitamin A-deficient testis but was strongly induced in these cells after the induction of differentiation of most of these cells into A(1) spermatogonia by administration of retinoic acid. Overall, cyclin D(2) seems to play a role at the crucial differentiation step of undifferentiated spermatogonia into A(1) spermatogonia. Cyclin D(3) is expressed in both proliferating and quiescent gonocytes during testis development. Cyclin D(3) expression was found in terminally differentiated Sertoli cells, in Leydig cells, and in spermatogonia in adult testis. Hence, although cyclin D(3) may control G(1)/S transition in spermatogonia, it probably has a different role in Sertoli and Leydig cells. In conclusion, the three D-type cyclins are differentially expressed during spermatogenesis. In spermatogonia, cyclins D(1) and D(3) seem to be involved in cell cycle regulation, whereas cyclin D(2) likely has a role in spermatogonial differentiation.


Subject(s)
Cyclin D1/physiology , Cyclins/physiology , Germ Cells/physiology , Testis/cytology , Animals , Cell Differentiation/physiology , Cell Division/physiology , Cyclin D2 , Cyclin D3 , Female , Immunohistochemistry , Male , Mice , Mice, Inbred Strains , Pregnancy , Testis/growth & development , Testis/metabolism , Vitamin A Deficiency/metabolism , Vitamin A Deficiency/pathology
3.
Mol Reprod Dev ; 56(3): 353-9, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10862001

ABSTRACT

Using immunohistochemical techniques and Western blot analysis, the possible role of Bcl-2 family members Bax, Bcl-2, Bcl-x(s), and Bcl-x(l) in male germ cell density-related apoptosis and DNA damage induced apoptosis was studied. The apoptosis inducer Bax was localized in all mouse and human testicular cell types, but despite the fact that irradiation induces its transcriptional activator, p53 in the human, Bax expression did not change after irradiation. The apoptosis inhibitor Bcl-2 appeared to be present in late spermatocytes and spermatids and was up-regulated in these cells after a dose of 4 Gy of X-rays. Finally, Bcl-x was expressed in both the mouse and human testis. The apoptosis inhibiting long transcripts of Bcl-x, Bcl-x(l), were expressed in spermatogonia and spermatocytes and were up-regulated after X-irradiation. The apoptosis inducing shorter form of Bcl-x, Bcl-x(s), was found to be expressed only in somatic cells, like peritubular and Leydig cells. While Bax is important in germ cell density regulation, Bax expression did not change after DNA damage inflicted by X-radiation. Hence, spermatogonial apoptosis after X-irradiation may not be induced via the apoptosis inducer Bax. Furthermore, as Bcl-x(l), but not Bcl-2, is present in spermatogonia and spermatocytes, Bcl-x(l) may regulate germ cell density, possibly in cooperation with Bax. As Bcl-x(l) expression is enhanced after irradiation, this protein may also have a role in the response of spermatogonia and spermatocytes to irradiation.


Subject(s)
Apoptosis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Testis/metabolism , Animals , Humans , Male , Mice , Mice, Knockout , Testis/pathology , Testis/radiation effects , bcl-2-Associated X Protein , bcl-X Protein
4.
Endocrinology ; 140(4): 1834-40, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10098522

ABSTRACT

p27kip1 is a cyclin-dependent kinase inhibitor that regulates the G1/S transition of the cell cycle. Immunohistochemical analysis showed that during mouse testicular development p27kip1 is induced when the fetal germ cells, gonocytes, become quiescent on day 16 postcoitum, suggesting that p27kip1 is an important factor for the G1/G0 arrest in gonocytes. In the adult mouse and human testis, in general, spermatogonia are proliferating actively, except for undifferentiated spermatogonia that also go through a long G1/G0 arrest. However, none of the different types of germ cells immunohistochemically stained for p27kip1. During development, Sertoli cells are proliferating actively and only occasionally were lightly p27kip1 stained Sertoli cells observed. In contrast, in the adult testis the terminally differentiated Sertoli cells heavily stain for p27kip1. Twenty to 30% of both fetal and adult type Leydig cells lightly stained for p27kip1, possibly indicating the proportion of terminally differentiated cells in the Leydig cell population. In p27kip1 knockout mice, aberrations in the spermatogenic process were observed. First, an increase in the numbers ofA spermatogonia was found, and second, abnormal (pre)leptotene spermatocytes were observed, some of which seemingly tried to enter a mitotic division instead of entering the meiotic prophase. These observations indicate that p27kip1 has a role in the regulation of spermatogonial proliferation, or apoptosis, and the onset of the meiotic prophase in preleptotene spermatocytes. However, as p27kip1 is only expressed in Sertoli cells, the role of p27kip1 in both spermatogonia and preleptotene spermatocytes must be indirect. Hence, part of the supportive and/or regulatory role of Sertoli cells in the spermatogenic process depends on the expression of p27kip1 in these cells. Finally, we show that the expression of p27kip1 transiently increases by a factor of 3 after x-irradiation in whole testicular lysates. Hence, p27kip1 seems to be involved in the cellular response after DNA damage.


Subject(s)
Cell Cycle Proteins , Enzyme Inhibitors/analysis , Microtubule-Associated Proteins/physiology , Testis/growth & development , Tumor Suppressor Proteins , Adult , Animals , Animals, Newborn , Cell Differentiation , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/antagonists & inhibitors , Gestational Age , Humans , Immunohistochemistry , Leydig Cells/chemistry , Leydig Cells/cytology , Male , Mice , Mice, Knockout , Microtubule-Associated Proteins/analysis , Sertoli Cells/chemistry , Sertoli Cells/cytology , Spermatogenesis , Spermatozoa/chemistry , Spermatozoa/cytology , Testis/cytology , Testis/embryology , X-Rays
5.
Cell Death Differ ; 5(8): 669-77, 1998 Aug.
Article in English | MEDLINE | ID: mdl-10200522

ABSTRACT

The p53 protein appeared to be involved in both spermatogonial cell proliferation and radiation response. During normal spermatogenesis in the mouse, spermatogonia do not express p53, as analyzed by immunohistochemistry. However, after a dose of 4 Gy of X-rays, a distinct p53 staining was present in spermatogonia, suggesting that, in contrast to other reports, p53 does have a role in spermatogonia. To determine the possible role of p53 in spermatogonia, histological analysis was performed in testes of both p53 knock out C57BL/6 and FvB mice. The results indicate that p53 is an important factor in normal spermatogonial cell production as well as in the regulation of apoptosis after DNA damage. First, p53 knock out mouse testes contained about 50% higher numbers of A1 spermatogonia, indicating that the production of differentiating type spermatogonia by the undifferentiated spermatogonia is enhanced in these mice. Second, 10 days after a dose of 5 Gy of X-rays, in the p53 knock out testes, increased numbers of giant sized spermatogonial stem cells were found, indicating disturbance of the apoptotic process in these cells. Third, in the p53 knock out testis, the differentiating A2-B spermatogonia are more radioresistant compared to their wild-type controls, indicating that p53 is partly indispensable in the removal of lethally irradiated differentiating type spermatogonia. In accordance with our immunohistochemical data, Western analysis showed that levels of p53 are increased in total adult testis lysates after irradiation. These data show that p53 is important in the regulation of cell production during normal spermatogenesis either by regulation of cell proliferation or, more likely, by regulating the apoptotic process in spermatogonia. Furthermore, after irradiation, p53 is important in the removal of lethally damaged spermatogonia.


Subject(s)
Spermatogenesis/physiology , Tumor Suppressor Protein p53/physiology , Animals , Cell Count , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Testis/radiation effects , Tumor Suppressor Protein p53/genetics , X-Rays
6.
Mol Reprod Dev ; 47(3): 240-7, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9170103

ABSTRACT

During spermatogenesis, the radiosensitivity of testicular cells changes considerably. To investigate the molecular mechanisms underlying these radiosensitivity differences, p21(Cip1/WAF1) expression was studied before and after irradiation in the adult mouse testis. P21(Cip1/WAF1) is a cyclin-dependent kinase inhibitor (CDI) and has a role in the G1/S checkpoint and differentiation. P21(Cip1/WAF1) expression was observed in the normal testis, using Western blotting analysis. After a dose of 4 Gy, but not after 0.3 Gy, an increase in p21(Cip1/WAF1) expression could be determined in whole testis lysates. To investigate which germ cells are involved in p21(Cip1/WAF1) protein expression, immunohistochemical analysis was performed on irradiated testis. In the normal testis a weak staining for p21(Cip1/WAF1) was found in pachytene spermatocytes in epithelial stage V up to step 5 spermatids. A dose of 4 Gy of X-irradiation resulted in a transient increase of p21(Cip1/WAF1) staining in these cells with a maximum at 6 h post irradiation, despite the fact that the irradiation did not induce an increase in the number of apoptotic spermatocytes. When a dose of 0.3 Gy was given, no increase in p21(Cip1/WAF1) staining was observed. Using the TUNEL technique, a 10-fold increase in apoptotic spermatogonia was found after a dose of 4 Gy. However, no staining for p21(Cip1/WAF1) was observed in spermatogonia, suggesting that these cells do not undergo a p21(Cip1/WAF1)-induced G1 arrest prior to DNA repair or apoptosis. These data imply that p21(Cip1/WAF1) is a factor which could be important during the meiotic prophase in spermatocytes and repair mechanisms in these cells, but not in spermatogonial cell cycle delay or apoptosis induction.


Subject(s)
Cyclins/biosynthesis , Testis/radiation effects , Animals , Apoptosis , Blotting, Western , Cyclin-Dependent Kinase Inhibitor p21 , Male , Mice , RNA, Messenger/analysis , Testis/metabolism
7.
Radiat Res ; 141(1): 66-73, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7997516

ABSTRACT

The effects of prenatal X irradiation on postnatal development of the CBA/P mouse testis was studied. At days 14, 15 and 18 post coitus pregnant female mice were exposed to single doses of X rays ranging from 0.25-1.5 Gy. Higher doses resulted in extensive loss of fetal mice. In the male offspring, at days 3 and 31 post partum, the numbers of gonocytes, type A spermatogonia and Sertoli cells per testis were determined using the disector method. Furthermore, after irradiation at day 15 post coitus, the numbers of Leydig cells, mesenchymal cells, macrophages, myoid cells, lymphatic endothelial cells, endothelial cells and perivascular cells per testis were also determined at days 3 and 31 post partum. At day 3 post partum, the number of germ cells was decreased after irradiation at days 14 and 15 post coitus. A D0 value of 0.7 Gy was determined for the radiosensitivity of the gonocytes at day 14 post coitus. A D0 value of 0.8 Gy was determined for the gonocytes at day 15 post coitus which, however, seems to be less accurate. No accurate D0 value could be determined for the gonocytes at day 18 post coitus. At day 31 post partum, the repopulation of the seminiferous epithelium as well as testis weights and tubular diameters were more affected by irradiation with increasing age of the mice at the time of irradiation. The percentage of tubular cross sections showing spermatids decreased with increasing dose after irradiation at days 15 and 18 post coitus, but not after irradiation at day 14 post coitus. Furthermore, in tubular cross sections showing spermatids, exposure of testes to 1.25 and 1.5 Gy at day 18 post coitus resulted in significantly lower numbers of spermatids per cross section when compared to those testes exposed to the same doses at day 15 post coitus. This indicates that the radiosensitivity of the gonocytes increases with fetal age. Prenatal irradiation did not cause significant changes in the numbers per testis of the Sertoli cells or the interstitial cell types. The present results indicate that, in the fetal mouse testis, the spermatogonial stem cells are more sensitive to X irradiation than in the adult testis, while Sertoli cells and interstitial cells are relatively resistant.


Subject(s)
Aging/radiation effects , Prenatal Exposure Delayed Effects , Spermatogenesis/radiation effects , Testis/radiation effects , Animals , Body Weight/radiation effects , Dose-Response Relationship, Radiation , Female , Fetal Death , Fetus , Gestational Age , Male , Mice , Mice, Inbred CBA , Organ Size/radiation effects , Pregnancy , Sertoli Cells/pathology , Sertoli Cells/radiation effects , Testis/embryology , Testis/pathology , X-Rays
8.
Radiat Res ; 139(3): 316-26, 1994 Sep.
Article in English | MEDLINE | ID: mdl-8073114

ABSTRACT

The effects of total-body X irradiation on the prepubertal testis of the CBA/P mouse have been studied. At either day 14 or day 29 post partum male mice were exposed to single doses of X rays ranging from 1.5-6.0 Gy. At 1 week after irradiation the repopulation index method was used to study the radiosensitivity of the spermatogonial stem cells. A D0 value of 1.8 Gy was determined for the stem cells at day 14 post partum as well as for the stem cells at day 29 post partum, indicating that the radiosensitivity of the spermatogonial stem cells in the prepubertal mouse testis is already comparable to that observed in the adult mouse. One, 2 or 3 weeks after irradiation total cell numbers per testis of Sertoli cells, Leydig cells, mesenchymal cells, macrophages, myoid cells, lymphatic endothelial cells, endothelium and perivascular cells were determined using the disector method. The Sertoli cells and interstitial cell types appeared to be relatively radioresistant during the prepubertal period. No significant changes in plasma testosterone levels were found, indicating that there is no Leydig cell dysfunction after exposure to doses up to 6 Gy during the prepubertal period. Taken together, the radioresponse of the prepubertal mouse testis is comparable to that of the adult mouse testis.


Subject(s)
Testis/radiation effects , Age Factors , Animals , Body Weight/radiation effects , Leydig Cells/radiation effects , Male , Mice , Mice, Inbred CBA , Organ Size/radiation effects , Sertoli Cells/radiation effects , Spermatogonia/radiation effects , Testis/cytology , X-Rays
9.
J Reprod Fertil ; 99(2): 479-85, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8107030

ABSTRACT

The postnatal development of body and testis weight and the size of the testicular cell populations were studied in CBA mice up to day 52 post partum. The body weight increased from 1.3 g at day 1 to 22.5 g at day 52. Over the same interval the testis weight showed a faster increase from about 1 mg to almost 60 mg. Spermatogenesis was found to be complete by day 35. The numbers of A spermatogonia, Sertoli cells, Leydig cells, mesenchymal cells, macrophages, myoid cells, lymphatic endothelial cells, endothelial cells and perivascular cells per testis were studied from day 3 to day 50, using the dissector method. The number of A spermatogonia increased from 0.2 x 10(5) at day 3 to 6.5 x 10(5) at day 21 and remained more or less constant thereafter. The Sertoli cell population increased during the first three weeks after birth to reach the adult level of approximately 18 x 10(5) cells per testis. In the interstitium the Leydig cells showed a sharp increase between days 11 and 31 followed by a small decrease to ultimately 9 x 10(5) cells per testis. The Leydig cells formed 8% of the total number of interstitial cells per testis at day 11, increasing to 30% at day 50. The number of mesenchymal cells did not change until day 36, decreasing thereafter from about 2.5 x 10(5) to 1 x 10(5) cells per testis at day 50. However, the percentage of the total number of interstitial cells that were mesenchymal cells decreased from 59% to 4%.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Testis/growth & development , Animals , Body Weight/physiology , Cell Count , Cell Division/physiology , Endothelium/cytology , Endothelium, Lymphatic/cytology , Leydig Cells/cytology , Macrophages/cytology , Male , Mesoderm/cytology , Mice , Mice, Inbred CBA , Organ Size/physiology , Sertoli Cells/cytology , Spermatogenesis/physiology , Spermatogonia/cytology , Testis/anatomy & histology , Testis/cytology
10.
Article in English | MEDLINE | ID: mdl-1348894

ABSTRACT

The sensitivity of resting and proliferating cells of the seminal vesicle to X-irradiation and adriamycin has been investigated. Stimulation with testosterone propionate (250 micrograms/day) was started 11 days after castration in BALB/c mice. X-rays (2.5-7.5 Gy total body irradiation) and intraperitoneal injections of adriamycin (4-16 mg/kg body weight) were administered at various times before or after induction of proliferation by testosterone injection. The DNA contents and the weights of the seminal vesicles were determined at 4 days after the start of stimulation. A Do for X-rays of about 10 Gy was found for the seminal vesicle epithelium. For both X-irradiation and adriamycin no significant differences in sensitivity were observed between quiescent (Go) and proliferating (G1; S) seminal vesicle cells.


Subject(s)
Doxorubicin/pharmacology , Seminal Vesicles/radiation effects , Animals , Castration , Cell Cycle/radiation effects , DNA/analysis , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Epithelium/drug effects , Epithelium/radiation effects , Male , Mice , Mice, Inbred BALB C , Mitosis/drug effects , Mitosis/radiation effects , Organ Size/drug effects , Organ Size/radiation effects , Seminal Vesicles/drug effects , Seminal Vesicles/pathology , Testosterone/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...