Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 196
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Reproduction ; 160(2): 205-215, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32438343

RESUMO

The existence of cytoplasmic passages between germ cells and their potential function in the control of the spermatogenic process has long been an intriguing question. Evidence of the important role of such structures, known as intercellular bridges (ICB), in spermatogenesis has been implicated by the failure of spermatogenesis in testis-expressed gene 14 (Tex14) mutant mice, which lack the ICBs, to progress past the pachytene spermatocyte stage. Using these Tex14 mutants, the present study evaluated, for the first time, the behavior and synchrony of the spermatogonial lineage in the absence of ICBs. Our data suggest that the absence of these cytoplasmic connections between cells affects the expansion of the undifferentiated type A (Aundiff) spermatogonia compartment and their transition to A1, resulting in a significant numerical reduction of differentiating A1 spermatogonia, but did not interfere with cell amplification during subsequent mitotic steps of differentiating spermatogonia from A1 through intermediate (In). However, beginning at the type B spermatogonia, the synchrony of differentiation was impaired as some cells showed delayed differentiation compared to their behavior in a normal seminiferous epithelium cycle. Thus although spermatogonial development is able to proceed, in the absence of ICBs in Tex14-/- mutants, the yield of cells, specific steps of differentiation, the synchrony of the cell kinetics, and the subsequent progression in meiosis are quantitatively lower than normal.


Assuntos
Comunicação Celular , Diferenciação Celular , Meiose , Epitélio Seminífero/patologia , Espermatogênese , Espermatogônias/patologia , Fatores de Transcrição/fisiologia , Animais , Proliferação de Células , Citoplasma , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Epitélio Seminífero/metabolismo , Espermatogônias/metabolismo
2.
Nat Genet ; 16(1): 74-8, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9140398

RESUMO

Each year more than 20,000 children and young persons of reproductive age are exposed to known mutagens in the form of chemo- and/or radiotherapy for cancer in the States. As more of these treatments are effective there is growing concern that genetic defects are introduced in the germ cells of these young patients. It is well documented for male rodents that treatment with chemo- and radio-therapeutic agents before mating can cause genetic damage in the germ line, and the magnitude of heritable effects depends on the spermatogenic cell stage treated. Similar germinal effects are suspected to occur in humans but remain unproven. Hodgkin's disease (HD) is an example of a malignancy which is typically diagnosed during a patient's reproductive years. In our study we observed eight male HD patients who were treated with NOVP (Novanthrone, Oncovin, Vinblastine, Prednisone) chemotherapy. We evaluated sperm aneuploidy using multi-colour fluorescence in situ hybridization (FISH), and found approximately 5-fold increases in sperm with disomies, diploidies and complex genotypes involving chromosome X, Y and 8. Increases in sex chromosome aneuploidies arose from segregation errors at meiosis I as well as meiosis II. The aneuploidy effects were transient, however, declining to pretreatment levels within approximately 100 days after the end of the therapy. When compared with normal men, some HD patients showed higher proportions of certain sperm aneuploidy types even before their first therapy.


Assuntos
Aneuploidia , Antineoplásicos/efeitos adversos , Doença de Hodgkin/tratamento farmacológico , Doença de Hodgkin/genética , Espermatozoides/efeitos dos fármacos , Adulto , Antineoplásicos/uso terapêutico , Diploide , Doença de Hodgkin/radioterapia , Humanos , Hibridização in Situ Fluorescente , Masculino , Pessoa de Meia-Idade , Prednisona/efeitos adversos , Prednisona/uso terapêutico , Valores de Referência , Espermatozoides/fisiologia , Fatores de Tempo , Vimblastina/efeitos adversos , Vimblastina/uso terapêutico , Vincristina/efeitos adversos , Vincristina/uso terapêutico
3.
J Cell Biol ; 64(1): 211-22, 1975 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-1109231

RESUMO

The incorporation of radioactivity into various cells in the sequence of spermatogenesis was measured by preparing highly purified spermatozoan nuclei from the cauda epididymidis of mice at daily intervals after injection of (3H)thymidine. The stages of differentiation of these sperm at the time of thymidine administration were calculated from the kinetics of spermatogenesis. The procedure for purification of sperm nuclei included sonication, mechanical shearing, and treatment with trypsin, DNase, Triton X-100, 2M NaC1, and sodium dodecyl sulfate. DNA was isolated from these nuclei by treatment with dithiothreitol and pronase, followed by phenol extraction and ethanol precipitation. The levels of radioactivity in the epididymal sperm head preparations were low (less than 13 dpm/mouse) for 27 days after injection, and then rose dramatically to over 4 times 104 dpm/mouse. Further experiments demonstrated that the 11 dpm of 3H radioactivity contained in sperm heads at 21 or 26 days after injection of (3H)TdR was significantly above background and contamination levels from other cells or other sources. Most of the radioactivity was in the sperm DNA and represented incorporation of tritium from (3H)TdR into the nuclear DNA of meiotic cells at 0.002 percent of the rate of incorporation into S-phase cells. Little, if any, (3H)TdR was incorporation into the DNA of spermatids. The levels of DNA synthesis during the meiotic prophase in the mouse appear to be much lower than those reported for other organisms.


Assuntos
DNA/biossíntese , Meiose , Espermatogênese , Animais , Arginina/metabolismo , Diferenciação Celular , Núcleo Celular/metabolismo , Desoxirribonucleases , Ditiotreitol , Epididimo , Cinética , Masculino , Camundongos , Camundongos Endogâmicos , Microscopia Eletrônica , Nucleoproteínas/biossíntese , Polietilenoglicóis , Pronase , Cloreto de Sódio , Dodecilsulfato de Sódio , Espermatozoides/metabolismo , Espermatozoides/ultraestrutura , Timidina/metabolismo , Fatores de Tempo , Trítio , Tripsina
4.
Science ; 169(3943): 370-1, 1970 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-5450367

RESUMO

The phototropic response and distribution of photopigment in the sporangiophore of Phycomyces was investigated with a microillumination pattern. The data support the hypothesis that phototropism results from greater stimulation of growth in regions of more intense illumination and indicate that the photoreceptor extends to the outer wall of the sporangiophore.


Assuntos
Fungos/efeitos da radiação , Lasers , Efeitos da Radiação , Fungos/crescimento & desenvolvimento , Pigmentos Biológicos/metabolismo
5.
Andrology ; 7(2): 257-265, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30471208

RESUMO

BACKGROUND: Cytotoxic cancer treatments, such as irradiation, can cause permanent sterility in male mammals owing to the loss of spermatogonial stem cells. In animal models, spermatogenesis could be restored from transplanted spermatogonial stem cells. Previously, we showed that transient suppression of FSH, LH, and testosterone in the recipient with a gonadotropin-releasing hormone antagonist (GnRH-ant), given immediately after irradiation, enhanced spermatogenesis from transplanted spermatogonial stem cells in mice and monkeys. OBJECTIVES: To explore improvements in the preparation of the recipient for efficient and reliable spermatogenic recovery from spermatogonial stem cell transplantation, so that it can be used effectively in clinical practice. MATERIALS AND METHODS: In mouse recipients, we evaluated the effects of hormone suppression given after germ cell depletion was complete, which is a more clinically relevant model, and also the importance of total androgen ablation and maintenance of FSH levels. Three regimens, GnRH-ant, GnRH-ant plus flutamide (androgen receptor antagonist), and GnRH-ant plus FSH, were administered prior to and around the time of transplantation of testis cells from immature mice or from prepubertal monkeys. RESULTS: Treatment with GnRH-ant resulted in a fourfold increase in spermatogenic recovery from GFP-marked transplanted mouse cells. Total androgen ablation with the addition of flutamide, started two weeks before transplantation, did not further enhance recovery. Surprisingly, FSH supplementation, started around the time of transplantation, actually reduced spermatogenic recovery from transplanted spermatogonial stem cells in GnRH-ant-treated mice. When prepubertal monkey testicular cells were transplanted into nude mice that were given the same hormone treatments, the numbers of donor-derived colonies were independent of hormone treatment. DISCUSSION AND CONCLUSION: The enhancements in spermatogenic recovery may only occur when syngeneic or closely related donor-recipient pairs are used. These results are useful in further investigations in choosing a hormone suppression regimen in combination with spermatogonial transplantation as a treatment to restore fertility in primates after cytotoxic therapy.


Assuntos
Células-Tronco Germinativas Adultas/transplante , Antagonistas de Hormônios/farmacologia , Espermatogênese/efeitos dos fármacos , Transplante de Células-Tronco/métodos , Animais , Infertilidade Masculina , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Espermatogênese/fisiologia , Transplante Heterólogo , Transplante Isogênico
7.
Mol Cell Biol ; 19(7): 5001-13, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10373550

RESUMO

Ubiquitin-mediated proteolysis controls diverse physiological processes in eukaryotes. However, few in vivo targets of the mammalian Cdc34 and Rad6 ubiquitin-conjugating enzymes are known. A yeast-based genetic assay to identify proteins that interact with human Cdc34 resulted in three cDNAs encoding bZIP DNA binding motifs. Two of these interactants are repressors of cyclic AMP (cAMP)-induced transcription: hICERIIgamma, a product of the CREM gene, and hATF5, a novel ATF homolog. Transfection assays with mammalian cells demonstrate both hCdc34- and hRad6B-dependent ubiquitin-mediated proteolysis of hICERIIgamma and hATF5. This degradation requires an active ubiquitin-conjugating enzyme and results in abrogation of ICERIIgamma- and ATF5-mediated repression of cAMP-induced transcription. Consistent with these results, the endogenous ICER protein is elevated in cells which are null for murine Rad6B (mHR6B-/-) or transfected with dominant negative and antisense constructs of human CDC34. Based on the requirement for CREM/ICER and Rad6B proteins in spermatogenesis, we determined expression of Cdc34, Rad6B, CREM/ICER isoforms, and the Skp1-Cullin-F-box ubiquitin protein ligase subunits Cul-1 and Cul-2, which are associated with Cdc34 activity during murine testicular development. Cdc34, Rad6B, and the Cullin proteins are expressed in a developmentally regulated manner, with distinctly different patterns for Cdc34 and the Cullin proteins in germ cells. The Cdc34 and Rad6B proteins are significantly elevated in meiotic and postmeiotic haploid germ cells when chromatin modifications occur. Thus, the stability of specific mammalian transcription factors is the result of complex targeting by multiple ubiquitin-conjugating enzymes and may have an impact on cAMP-inducible gene regulation during both meiotic and mitotic cell cycles.


Assuntos
Proteínas Sanguíneas/metabolismo , AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Ligases/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Complexos Ubiquitina-Proteína Ligase , Fatores Ativadores da Transcrição , Ciclossomo-Complexo Promotor de Anáfase , Animais , Sequência de Bases , Proteínas Sanguíneas/genética , Linhagem Celular , Clonagem Molecular , Modulador de Elemento de Resposta do AMP Cíclico , Cisteína Endopeptidases/metabolismo , DNA Complementar , Proteínas de Ligação a DNA/genética , Endopeptidases/metabolismo , Expressão Gênica , Humanos , Ligases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Complexos Multienzimáticos/metabolismo , Complexo de Endopeptidases do Proteassoma , Proteínas Repressoras/genética , Espermatogênese , Fatores de Transcrição/genética , Enzimas de Conjugação de Ubiquitina , Ubiquitina-Proteína Ligases , Ubiquitinas/metabolismo
8.
Mol Cell Biol ; 16(7): 3720-9, 1996 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8668189

RESUMO

Mammalian spermiogenesis is characterized by a striking restructuring of the spermatid chromatin caused by the replacement of nucleohistones with transition proteins and their subsequent replacement with nucleoprotamines. The onset of nuclear elongation and chromatin condensation in spermatids is accompanied by a general decrease in the transcriptional activity of the DNA. A recently identified testis-specific high-mobility-group (tsHMG) protein, similar to the human mitochondrial transcription factor I and to the linker-associated protein delta of Tetrahymena thermophila micronuclei, is thought to play a structural role in this process. We confirm by immunoblot analysis of fractionated germ cells that the presence of tsHMG is restricted to transcriptionally quiescent elongating and condensing spermatids. Purified recombinant tsHMG protein displays preferential binding to supercoiled plasmid DNA, which reversibly protects the DNA against the DNA-relaxing activity of eukaryotic topoisomerase I and also impairs the transcriptional activity of this template when assayed in vitro. The tsHMG protein can also introduce negative supercoils into a relaxed plasmid substrate in a topoisomerase I-dependent manner. We also show that the tsHMG protein is the substrate of a Ca2+-phospholipid-dependent protein kinase (protein kinase C) present in testis extracts of adult mice and demonstrate that phosphorylation by protein kinase C is required for both the DNA-binding and the topoisomerase I-dependent supercoiling activities of tsHMG. Our results support the hypothesis that the spermatid tsHMG protein is a topological factor (transition protein) that can modulate the activity of topoisomerase I. This activity could contribute to the important transition in chromatin structure which leads to the decrease in DNA metabolism observed at the early stages of spermatid elongation.


Assuntos
Proteínas de Grupo de Alta Mobilidade/metabolismo , Proteínas Mitocondriais , Proteínas Nucleares , Espermátides/fisiologia , Animais , DNA Super-Helicoidal/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Proteínas de Grupo de Alta Mobilidade/biossíntese , Proteínas de Grupo de Alta Mobilidade/isolamento & purificação , Humanos , Masculino , Camundongos , Fosforilação , Plasmídeos/metabolismo , Proteínas de Protozoários/metabolismo , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Espermatócitos/metabolismo , Espermatogênese , Espermatogônias/metabolismo , Tetrahymena thermophila/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica
9.
Mol Cell Biol ; 21(21): 7243-55, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11585907

RESUMO

During mammalian spermiogenesis, major restructuring of chromatin takes place. In the mouse, the histones are replaced by the transition proteins, TP1 and TP2, which are in turn replaced by the protamines, P1 and P2. To investigate the role of TP2, we generated mice with a targeted deletion of its gene, Tnp2. Spermatogenesis in Tnp2 null mice was almost normal, with testis weights and epididymal sperm counts being unaffected. The only abnormality in testicular histology was a slight increase of sperm retention in stage IX to XI tubules. Epididymal sperm from Tnp2-null mice showed an increase in abnormal tail, but not head, morphology. The mice were fertile but produced small litters. In step 12 to 16 spermatid nuclei from Tnp2-null mice, there was normal displacement of histones, a compensatory translationally regulated increase in TP1 levels, and elevated levels of precursor and partially processed forms of P2. Electron microscopy revealed abnormal focal condensations of chromatin in step 11 to 13 spermatids and progressive chromatin condensation in later spermatids, but condensation was still incomplete in epididymal sperm. Compared to that of the wild type, the sperm chromatin of these mutants was more accessible to intercalating dyes and more susceptible to acid denaturation, which is believed to indicate DNA strand breaks. We conclude that TP2 is not a critical factor for shaping of the sperm nucleus, histone displacement, initiation of chromatin condensation, binding of protamines to DNA, or fertility but that it is necessary for maintaining the normal processing of P2 and, consequently, the completion of chromatin condensation.


Assuntos
Cromatina/ultraestrutura , Fertilidade/genética , Proteínas Nucleares/genética , Espermatozoides/fisiologia , Espermatozoides/ultraestrutura , Animais , Northern Blotting , Núcleo Celular/metabolismo , Cromatina/metabolismo , Proteínas de Ligação a DNA , Citometria de Fluxo , Deleção de Genes , Genótipo , Immunoblotting , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Modelos Genéticos , Mutagênese Sítio-Dirigida , Mutação , Espermatogênese/fisiologia , Espermatozoides/metabolismo , Testículo/ultraestrutura
10.
J Natl Cancer Inst ; 64(5): 1215-9, 1980 May.
Artigo em Inglês | MEDLINE | ID: mdl-6154171

RESUMO

The cycle-dependent cytotoxicity of seven chemotherapy agents was compared with the use of subpopulations of Chinese hamster ovary cells separated into the various phases of the cell cycle by centrifugal elutriation. The proportion of cells killed by either beta-cytosine arabinoside or hydroxyurea agreed well with the proportion of S-phase cells in the treated subpopulations. Cell-cycle survival patterns were also determined for bleomycin (BLM), adriamycin (ADR), cis-diamminedichloroplatinum (II) (cis-DDP), Asaley, and Yoshi 864. Three of the agents (cis-DDP, Asaley, and Yoshi 864) had similar cell-cycle survival patterns in that they all preferentially killed cells in the G1 phase. In contrast, BLM preferentially killed cells in G2+M phase, whereas ADR was most cytotoxic to cells in middle to late S-phase. This investigation demonstrated that separation of cultured mammalian cells into the various cell-cycle phases by centrifugal elutriation allows the rapid determination of the phase-dependent cytotoxic effects exerted by chemotherapy agents.


Assuntos
Antineoplásicos/farmacologia , Ciclo Celular , Melfalan/análogos & derivados , Mesilatos/farmacologia , Animais , Bleomicina/farmacologia , Separação Celular/métodos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Cisplatino/farmacologia , Cricetinae , Cricetulus , Citarabina/farmacologia , Doxorrubicina/farmacologia , Feminino , Hidroxiureia/farmacologia , Interfase/efeitos dos fármacos , Melfalan/farmacologia , Metáfase/efeitos dos fármacos , Ovário
11.
Cancer Res ; 39(9): 3575-82, 1979 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-476683

RESUMO

Studies of testicular cell killing in mice by several chemotherapeutic drugs have been performed to EVALUATE THE HARMFUL EFFECTS OF ONCOLYTIC AGENTS ON REPRODUCTION. Seven drugs, Adriamycin, 1-beta-D-arabinofuranosylcytosine, bleomycin, cyclophosphamide, hydroxyurea, vinblastine, and vincristine, given as single injections, were cytotoxic to differentiated spermatogonia. Adriamycin was also highly effective in killing stem cells. The other drugs produced little or no stem cell loss even at doses toxic to the animals. Negligible killing of spermatocytes and spermatids was noted at any dose level. The results demonstrated that oncolytic agents preferntially killed cells at specific stages of the spermatogenic pathway in mice at doses within the clinical range for humans.


Assuntos
Antineoplásicos/efeitos adversos , Testículo/efeitos dos fármacos , Animais , Diferenciação Celular , Injeções , Dose Letal Mediana , Masculino , Camundongos , Camundongos Endogâmicos , Espermatogônias/efeitos dos fármacos , Testículo/citologia , Fatores de Tempo
12.
Cancer Res ; 47(4): 1093-7, 1987 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-3542201

RESUMO

Protection of testicular integrity against damage from cyclophosphamide (CY) by simultaneous treatment with a gonadotropin-releasing hormone (GnRH) analogue was reported in BALB/c mice (L.M. Glode et al., Lancet, 1: 1132-1134, 1981). This approach has been used as the basis for clinical trials in various treatment centers (D. H. Johnson et al., Blood, 65:832-836, 1985) in an attempt to prevent iatrogenic sterility in males. This study aims at duplicating the original findings and obtaining quantitative data on spermatogonial killing by CY, and possible protection by GnRH, of differentiating and stem cell spermatogonia. Mice were treated with 23 daily injections of 0.4 micrograms D-leucine-6 GnRH, and with 200 mg/kg CY on Days 8, 15, and 22. Three additional groups of mice received phosphate-buffered saline and bovine serum albumin only, GnRH only, and CY only. Animals were killed at 29 days after the last injection to determine the number of late spermatids in testicular homogenates, and at 56 days for histological measurement of the ratio of elongated spermatids to Sertoli cells in the tubules. The twenty-ninth day assay was a measure of damage to differentiating spermatogonia, whose killing results in temporary sterility. The fifty-sixth day point assay assessed damage to stem spermatogonia, whose killing results in long-term or permanent sterility. Sperm counts at 29 days were identical in saline-treated control mice and GnRH-treated mice; no sperm were present in the CY-treated mice, both with and without GnRH. Thus, killing of differentiating spermatogonia by CY is not prevented by GnRH treatment. Similarly, counts of spermatids at 56 days showed no difference between saline- and GnRH-treated groups; a reduction to approximately 40% of control counts was observed equally with CY and CY plus GnRH treatments. Since GnRH treatment did not alter spermatogonial kinetics in BALB/c mice, it is not surprising that it did not protect against CY-induced damage. Thus, the mouse is not a suitable model for analyzing such effects of GnRH on spermatogenesis, and further studies in other experimental animals are needed if they are to be used as a rationale for clinical administration of GnRH to cancer patients.


Assuntos
Ciclofosfamida/toxicidade , Hormônio Liberador de Gonadotropina/farmacologia , Testículo/efeitos dos fármacos , Animais , Sobrevivência Celular/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/análogos & derivados , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Tamanho do Órgão/efeitos dos fármacos , Espermatogênese/efeitos dos fármacos
13.
Cancer Res ; 59(15): 3557-60, 1999 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10446960

RESUMO

Previous studies have shown that treatment of rats with gonadotropin-releasing hormone (GnRH) analogues or steroids either before exposure to procarbazine or radiation or after irradiation enhances subsequent levels of spermatogenesis. We demonstrate here that giving a GnRH agonist after procarbazine injection also enhances spermatogenesis and fertility. We also demonstrate that GnRH agonist stimulated recovery of spermatogenesis and fertility not only when the hormone was administered immediately after irradiation, but also at 20 weeks after irradiation, after the decline in spermatogenesis had occurred. These results suggest that GnRH agonist treatment given to azoospermic men after cytotoxic therapy for cancer may stimulate the recovery of spermatogenesis and fertility.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Antineoplásicos/toxicidade , Hormônio Liberador de Gonadotropina/agonistas , Infertilidade Masculina/tratamento farmacológico , Leuprolida/uso terapêutico , Procarbazina/toxicidade , Espermatogênese/efeitos dos fármacos , Animais , Antineoplásicos Hormonais/farmacologia , Diferenciação Celular/efeitos dos fármacos , Feminino , Hormônio Foliculoestimulante/sangue , Raios gama/efeitos adversos , Infertilidade Masculina/induzido quimicamente , Leuprolida/farmacologia , Hormônio Luteinizante/sangue , Masculino , Ratos , Ratos Sprague-Dawley , Espermatogênese/efeitos da radiação , Testosterona/sangue
14.
Cancer Res ; 37(12): 4291-6, 1977 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-922721

RESUMO

Centrifugal elutriation was used to separate cells dissociated from two hypotetraploid mouse solid tumors, a fibrosarcoma and a sarcoma derived from L-P59 cells, based on their sedimentation rates. The separation was rapid, requiring less than 1 hr; yielded about 80 percent cell recovery; and resulted in little loss of cell viability. Aanlysis of DNA content by flow cytometry demonstrated the synchrony obtained with these tumor cells. The fractions with the lowest sedimentation rates contained predominantly normal cells, those with intermediate sedimentation rates contained predominantly tumor cells in the G1 phase of the cell cycle, and those with the highest sedimentation rates contained mostly tumor cells in S or G2. The clonogenicity of L-P59 cells, assayed in culture, markedly increased with increasing sedimentation rates. In contrast, the clonogencity of fibrosarcoma cells, assayed in vivo by a lung colony assay, was lower for the smaller cells, but was essentially constant among the larger cells. Autoradiography of cells labeled in vivo with tritiated thymidine demonstrated no differences in the proportions of cycling cells in various fractions. These results demonstrate that subpopulations differing in cell type, phase of the cell cycle, and clonogencity can be rapidly separated from solid tumors by centrifugal elutriation.


Assuntos
Separação Celular/métodos , Sarcoma Experimental , Animais , Contagem de Células , Ciclo Celular , Sobrevivência Celular , Centrifugação , Células Clonais/patologia , Feminino , Fibrossarcoma/patologia , Camundongos , Camundongos Endogâmicos C3H , Sarcoma Experimental/patologia
15.
Cancer Res ; 57(6): 1091-7, 1997 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-9067277

RESUMO

Hormone treatments that suppress sperm production enhance the recovery of spermatogenesis after gonadal exposure to various cytotoxic agents. It has generally been assumed that the mechanism of protection involved an arrest of spermatogonial kinetics. To test this hypothesis critically, we examined spermatogonial kinetics and numbers in rats in which the completion of spermatogenesis was suppressed with a 6-week testosterone plus 17beta-estradiol treatment that protected the testis from procarbazine-induced damage. Histological examination showed that the numbers of A-aligned, intermediate, and B spermatogonia and preleptotene spermatocytes and their mitoses were unaffected by testosterone plus 17beta-estradiol treatment. Flow cytometric analysis of bromodeoxyuridine-labeled cells showed that the percentage of diploid cells undergoing DNA synthesis, the progression of B spermatogonia and preleptotene spermatocytes through S-phase, the division of intermediate and B spermatogonia, the entry of intermediate spermatogonia into their next S-phase as type B cells, and the progression of cells through meiotic prophase were either unchanged or very slightly increased. Thus, changes in spermatogonial numbers or suppression of their proliferation cannot account for protection of spermatogenesis from exposure to cytotoxic agents.


Assuntos
Estradiol/uso terapêutico , Inibidores do Crescimento/toxicidade , Infertilidade Masculina/induzido quimicamente , Procarbazina/toxicidade , Espermatogênese/efeitos dos fármacos , Espermatogônias/efeitos dos fármacos , Testosterona/uso terapêutico , Animais , Divisão Celular/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Esquema de Medicação , Implantes de Medicamento , Estradiol/administração & dosagem , Infertilidade Masculina/prevenção & controle , Masculino , Ratos , Espermatogônias/patologia , Testosterona/administração & dosagem
16.
Cancer Res ; 42(1): 122-31, 1982 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-7198505

RESUMO

The harmful effects of 14 chemotherapeutic drugs on spermatogenesis in the mouse have been evaluated by studies of testicular cell killing and morphological and genetic damage produced. Male mice were given drugs as single injections at various doses up to the toxic levels. Prednisone and 6-mercaptopurine produced little or no cytotoxicity. All other drugs tested killed differentiated spermatogonia. Of these, methotrexate, cyclohexylchlorethylnitrosourea, cis-platinum, and mechlorethamine did not show significant stem cell killing. Bischlorethylnitrosourea, chlorambucil, 5-fluorouracil, mitomycin C, antinomycin D, and procarbazine showed some stem cell killing. Triethylenethiophosphoramide (thio-TEPA) was the only drug in this group which killed large numbers of stem cells. Only 5-fluorouracil and cis-platinum killed spermatocytes, and only cis-platinum killed spermatids. Several drugs induced chromosome breaks in treated spermatocytes. Thio-TEPA was effective in inducing chromosome translocations in treated spermatocytes and probably also in spermatocytes which originated from surviving treated stem cells. It had been our hypothesis that the cytotoxic effects of these drugs on mouse testicular stem cells would correlate with the duration of azoospermia observed in patients. This was shown not to be the case. Thus, the cytotoxic effects of single injections of single chemotherapeutic agents on the mouse testis did not appear to be predictive of which drugs will cause long-term azoospermia in humans.


Assuntos
Antineoplásicos/farmacologia , Testículo/efeitos dos fármacos , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos , Masculino , Camundongos , Espermatogênese/efeitos dos fármacos , Espermatogônias/efeitos dos fármacos , Testículo/patologia
17.
Cancer Res ; 50(2): 370-4, 1990 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-2295076

RESUMO

The mutagenic effects of doxorubicin (Adriamycin, ADR) on mouse spermatogonial stem cells were examined by analysis of spermatocyte chromosomes and of dominant lethality transmitted through the spermatozoa. The effects of ADR on mutations, cytotoxicity, and sperm head abnormalities were compared with those of radiation. The cytotoxic effect of 6 Gy of gamma-radiation on stem spermatogonia was equivalent to about 4-5 mg ADR/kg. Chromosomal translocations were observed in 0.6% of the spermatocytes of mice treated with ADR (2-6 mg/kg). In contrast, 6 Gy of radiation induced translocations in 11.1% of spermatocytes. No increase in dominant lethality was observed after treatment with ADR at doses up to 6 mg/kg, while the frequency after 6 Gy of radiation was 3.6%. Based on these results, ADR would be expected to be only a weak inducer of balanced chromosomal rearrangements. Because ADR at 4.5 mg/kg was much weaker than 6 Gy of gamma-radiation at inducing chromosomal translocations, but just as effective at inducing sperm head abnormalities, the level of sperm head abnormalities is not indicative of balanced chromosomal rearrangements induced in stem spermatogonia by cytotoxic agents.


Assuntos
Aberrações Cromossômicas , Doxorrubicina/toxicidade , Espermatogônias/efeitos dos fármacos , Espermatozoides/efeitos dos fármacos , Animais , Masculino , Camundongos , Camundongos Endogâmicos C3H , Mutação , Cabeça do Espermatozoide/efeitos dos fármacos , Espermatogônias/ultraestrutura
18.
Cancer Res ; 37(10): 3690-3, 1977 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-908016

RESUMO

Exponentially growing FSA 1233 cells, one of the clones isolated from a mouse fibrosarcoma, were synchronized by fractionation according to cell size by centrifugal elutriation. Cells from each fraction were analyzed by flow microfluorometry to determine the stages of the cell cycle and were injected i.v. to determine lung colony-forming efficiency. In vitro plating efficiency of these cells was similar throughout the cell cycle except for a slight reduction at G2 + M. On the other hand, lung colony-forming efficiency showed marked cell cycle and cell size dependencies, being lowest at G1, highest at S, and declining slightly at G2.


Assuntos
Neoplasias Pulmonares/patologia , Metástase Neoplásica/patologia , Animais , Contagem de Células , Ciclo Celular , Separação Celular , Células Clonais/patologia , DNA de Neoplasias/análise , Masculino , Camundongos , Camundongos Endogâmicos C3H , Transplante de Neoplasias , Neoplasias Experimentais/análise , Neoplasias Experimentais/patologia , Transplante Isogênico
19.
Cancer Res ; 54(4): 1027-34, 1994 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-8313358

RESUMO

Procarbazine produces long-term sterility in the male by killing stem spermatogonia. The degree and selectivity of protection of stem spermatogonia in rats from procarbazine by pretreatment with steroid hormones were investigated. Male LBNF1 rats were treated for 6 weeks with Silastic implants containing testosterone plus 17 beta-estradiol. The hormone-treated rats and sham-treated controls were given a single injection of graded doses of procarbazine and the hormone implants were removed the next day. Spermatogonial stem cell survival and function, assessed by the repopulation indices and sperm head counts 10 weeks later, showed that stem spermatogonia were protected by testosterone plus 17 beta-estradiol treatment from the toxic effects of procarbazine with a dose-modifying protection factor of about 2.5. In contrast, there was no hormonal protection from the procarbazine-induced killing of differentiating spermatogonia, preleptotene spermatocytes, and spermatocytes in meiotic prophase or from the delay in maturation of round spermatids, assessed 9 days after procarbazine injection by histological or flow cytometric methods. In addition, there was no hormonal protection from the procarbazine-induced decline in body weights and lymphocyte counts, indicating that the gastrointestinal, neurological, and hematological systems were not protected. The specificity of protection indicates that the hormonal protection of the stem spermatogonia is not the result of a systemic or overall testicular decrease in drug delivery, decrease in bioactivation, nor increase in drug detoxification, except possibly within the stem cells themselves. We conclude that the degree of hormonal protection and its specificity would be appropriate for clinical application provided that the mechanism of protection is elucidated and appears applicable to humans.


Assuntos
Hormônios Esteroides Gonadais/farmacologia , Procarbazina/toxicidade , Espermatogônias/efeitos dos fármacos , Testículo/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , DNA/análise , Relação Dose-Resposta a Droga , Citometria de Fluxo , Contagem de Leucócitos/efeitos dos fármacos , Masculino , Procarbazina/metabolismo , Ratos , Ratos Nus
20.
Cancer Res ; 54(3): 811-7, 1994 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-8306344

RESUMO

Solid tumors contain populations of proliferating (P) and quiescent (Q) cells. Shifting between these populations occurs continuously and cells are recruited from quiescence to proliferate (Q-->P) as a result of exogenously applied or endogenous cell depleting stimuli. Direct measurements of the proliferation kinetics of these Q-->P cells in solid tumors are difficult to make because of the much larger percentage of P-cells. In order to specifically analyze the kinetics of the Q-->P cells, double thymidine analogue labeling was used. This was accomplished by first labeling in vivo all of the P-cells in MCaK tumors using continuous exposure to chlorodeoxyuridine (CldUrd) administered by a minipump over 21 h. About 75% of the aneuploid cells are P-cells based on CldUrd labeling. At different times after the pumps were removed, the tumors were pulse-labeled with iododeoxyuridine (IdUrd) and harvested 6 h later. A 3-color flow cytometry assay was used to simultaneously and independently analyze CldUrd and IdUrd incorporation, as well as DNA content. The Q-->P cells were identified as having only been labeled with IdUrd. The length of their S-phase was calculated from the movement of the Q-->P cells during the 6 h after IdUrd labeling. The results showed the length of S-phase for the recruited cells to be slightly, but significantly, longer than the length of S-phase for the total cells (11 h versus 9 h, respectively). Thus, the recruited cells appear to have slightly slower kinetics than the proliferating cells in the absence of a perturbing stimulus such as radiotherapy or chemotherapy.


Assuntos
Neoplasias Mamárias Experimentais/patologia , Animais , Ciclo Celular/fisiologia , Divisão Celular/fisiologia , DNA de Neoplasias/análise , Desoxiuridina/análogos & derivados , Desoxiuridina/metabolismo , Modelos Animais de Doenças , Citometria de Fluxo , Idoxuridina/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos C3H
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA