Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Bioessays ; 45(1): e2200112, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36300921

RESUMO

Cancers often express hundreds of genes otherwise specific to germ cells, the germline/cancer (GC) genes. Here, we present and discuss the hypothesis that activation of a "germline program" promotes cancer cell malignancy. We do so by proposing four hallmark processes of the germline: meiosis, epigenetic plasticity, migration, and metabolic plasticity. Together, these hallmarks enable replicative immortality of germ cells as well as cancer cells. Especially meiotic genes are frequently expressed in cancer, implying that genes unique to meiosis may play a role in oncogenesis. Because GC genes are not expressed in healthy somatic tissues, they form an appealing source of specific treatment targets with limited side effects besides infertility. Although it is still unclear why germ cell specific genes are so abundantly expressed in cancer, from our hypothesis it follows that the germline's reproductive program is intrinsic to cancer development.


Assuntos
Neoplasias , Humanos , Neoplasias/genética , Células Germinativas , Carcinogênese/metabolismo , Meiose , Reprodução
2.
PLoS Genet ; 17(9): e1009773, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34499650

RESUMO

During gametogenesis in mammals, meiosis ensures the production of haploid gametes. The timing and length of meiosis to produce female and male gametes differ considerably. In contrast to males, meiotic prophase I in females initiates during development. Hence, the knowledge regarding progression through meiotic prophase I is mainly focused on human male spermatogenesis and female oocyte maturation during adulthood. Therefore, it remains unclear how the different stages of meiotic prophase I between human oogenesis and spermatogenesis compare. Analysis of single-cell transcriptomics data from human fetal germ cells (FGC) allowed us to identify the molecular signatures of female meiotic prophase I stages leptotene, zygotene, pachytene and diplotene. We have compared those between male and female germ cells in similar stages of meiotic prophase I and revealed conserved and specific features between sexes. We identified not only key players involved in the process of meiosis, but also highlighted the molecular components that could be responsible for changes in cellular morphology that occur during this developmental period, when the female FGC acquire their typical (sex-specific) oocyte shape as well as sex-differences in the regulation of DNA methylation. Analysis of X-linked expression between sexes during meiotic prophase I suggested a transient X-linked enrichment during female pachytene, that contrasts with the meiotic sex chromosome inactivation in males. Our study of the events that take place during meiotic prophase I provide a better understanding not only of female meiosis during development, but also highlights biomarkers that can be used to study infertility and offers insights in germline sex dimorphism in humans.


Assuntos
Cromossomos Humanos X , Células Germinativas , Prófase Meiótica I , Fatores Sexuais , Transcrição Gênica , Citoesqueleto/metabolismo , Metilação de DNA , Feminino , Expressão Gênica , Genitália Feminina/patologia , Humanos , Masculino , Oócitos/metabolismo
3.
Hum Reprod ; 38(11): 2208-2220, 2023 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-37671592

RESUMO

STUDY QUESTION: Are human ovarian aging and the age-related female fertility decline caused by oxidative stress and mitochondrial dysfunction in oocytes? SUMMARY ANSWER: We found oxidative damage in oocytes of advanced maternal age, even at the primordial follicle stage, and confirmed mitochondrial dysfunction in such oocytes, which likely resulted in the use of alternative energy sources. WHAT IS KNOWN ALREADY: Signs of reactive oxygen species-induced damage and mitochondrial dysfunction have been observed in maturing follicles, and even in early stages of embryogenesis. However, although recent evidence indicates that also primordial follicles have metabolically active mitochondria, it is still often assumed that these follicles avoid oxidative phosphorylation to prevent oxidative damage in dictyate arrested oocytes. Data on the influence of ovarian aging on oocyte metabolism and mitochondrial function are still limited. STUDY DESIGN, SIZE, DURATION: A set of 39 formalin-fixed and paraffin-embedded ovarian tissue biopsies were divided into different age groups and used for immunofluorescence analysis of oxidative phosphorylation activity and oxidative damage to proteins, lipids, and DNA. Additionally, 150 immature oocytes (90 germinal vesicle oocytes and 60 metaphase I oocytes) and 15 cumulus cell samples were divided into different age groups and used for targeted metabolomics and lipidomics analysis. PARTICIPANTS/MATERIALS, SETTING, METHODS: Ovarian tissues used for immunofluorescence microscopy were collected through PALGA, the nationwide network, and registry of histo- and cytopathology in The Netherlands. Comprehensive metabolomics and lipidomics were performed by liquid-liquid extraction and full-scan mass spectrometry, using oocytes and cumulus cells of women undergoing ICSI treatment based on male or tubal factor infertility, or fertility preservation for non-medical reasons. MAIN RESULTS AND THE ROLE OF CHANCE: Immunofluorescence imaging on human ovarian tissue indicated oxidative damage by protein and lipid (per)oxidation already at the primordial follicle stage. Metabolomics and lipidomics analysis of oocytes and cumulus cells in advanced maternal-age groups demonstrated a shift in the glutathione-to-oxiglutathione ratio and depletion of phospholipids. Age-related changes in polar metabolites suggested a decrease in mitochondrial function, as demonstrated by NAD+, purine, and pyrimidine depletion, while glycolysis substrates and glutamine accumulated, with age. Oocytes from women of advanced maternal age appeared to use alternative energy sources like glycolysis and the adenosine salvage pathway, and possibly ATP which showed increased production in cumulus cells. LIMITATIONS, REASONS FOR CAUTION: The immature oocytes used in this study were all subjected to ovarian stimulation with high doses of follicle-stimulating hormones, which might have concealed some age-related differences. WIDER IMPLICATIONS OF THE FINDINGS: Further studies on how to improve mitochondrial function, or lower oxidative damage, in oocytes from women of advanced maternal age, for instance by supplementation of NAD+ precursors to promote mitochondrial biogenesis, are warranted. In addition, supplementing the embryo medium of advanced maternal-age embryos with such compounds could be a treatment option worth exploring. STUDY FUNDING/COMPETING INTEREST(S): The study was funded by the Amsterdam UMC. The authors declare to have no competing interests. TRIAL REGISTRATION NUMBER: N/A.


Assuntos
NAD , Oócitos , Humanos , Feminino , Masculino , NAD/metabolismo , Oócitos/metabolismo , Estresse Oxidativo , Mitocôndrias/metabolismo , Envelhecimento
4.
J Biol Chem ; 295(32): 10885-10900, 2020 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-32487749

RESUMO

tRNA-derived small RNAs (tsRNAs) from spermatozoa could act as acquired epigenetic factors and contribute to offspring phenotypes. However, the roles of specific tsRNAs in early embryo development remain to be elucidated. Here, using pigs as a research model, we probed the tsRNA dynamics during spermatogenesis and sperm maturation and demonstrated the delivery of tsRNAs from semen-derived exosomes to spermatozoa. By microinjection of antisense sequences into in vitro fertilized oocytes and subsequent single-cell RNA-seq of embryos, we identified a specific functional tsRNA group (termed here Gln-TTGs) that participate in the early cleavage of porcine preimplantation embryos, probably by regulating cell cycle-associated genes and retrotransposons. We conclude that specific tsRNAs present in mature spermatozoa play significant roles in preimplantation embryo development.


Assuntos
Blastocisto , Divisão Celular , RNA de Transferência de Glutamina/fisiologia , RNA/metabolismo , Espermatozoides/metabolismo , Animais , Desenvolvimento Embrionário , Feminino , Masculino , Microinjeções , Gravidez , Maturação do Esperma , Espermatogênese , Suínos
5.
Development ; 145(16)2018 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-29540502

RESUMO

To prevent chromosomal aberrations being transmitted to the offspring, strict meiotic checkpoints are in place to remove aberrant spermatocytes. However, in about 1% of males these checkpoints cause complete meiotic arrest leading to azoospermia and subsequent infertility. Here, we unravel two clearly distinct meiotic arrest mechanisms that occur during prophase of human male meiosis. Type I arrested spermatocytes display severe asynapsis of the homologous chromosomes, disturbed XY-body formation and increased expression of the Y chromosome-encoded gene ZFY and seem to activate a DNA damage pathway leading to induction of p63, possibly causing spermatocyte apoptosis. Type II arrested spermatocytes display normal chromosome synapsis, normal XY-body morphology and meiotic crossover formation but have a lowered expression of several cell cycle regulating genes and fail to silence the X chromosome-encoded gene ZFX Discovery and understanding of these meiotic arrest mechanisms increases our knowledge of how genomic stability is guarded during human germ cell development.


Assuntos
Pontos de Checagem do Ciclo Celular/genética , Meiose/genética , Prófase/genética , Espermatócitos/metabolismo , Espermatogênese/fisiologia , Apoptose/fisiologia , Azoospermia/genética , Dano ao DNA/genética , Reparo do DNA/genética , Perfilação da Expressão Gênica , Humanos , Fatores de Transcrição Kruppel-Like/biossíntese , Masculino , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo
6.
Development ; 144(20): 3659-3673, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28935708

RESUMO

Spermatogenesis is a dynamic developmental process that includes stem cell proliferation and differentiation, meiotic cell divisions and extreme chromatin condensation. Although studied in mice, the molecular control of human spermatogenesis is largely unknown. Here, we developed a protocol that enables next-generation sequencing of RNA obtained from pools of 500 individually laser-capture microdissected cells of specific germ cell subtypes from fixed human testis samples. Transcriptomic analyses of these successive germ cell subtypes reveals dynamic transcription of over 4000 genes during human spermatogenesis. At the same time, many of the genes encoding for well-established meiotic and post-meiotic proteins are already present in the pre-meiotic phase. Furthermore, we found significant cell type-specific expression of post-transcriptional regulators, including expression of 110 RNA-binding proteins and 137 long non-coding RNAs, most of them previously not linked to spermatogenesis. Together, these data suggest that the transcriptome of precursor cells already contains the genes necessary for cellular differentiation and that timely translation controlled by post-transcriptional regulators is crucial for normal development. These established transcriptomes provide a reference catalog for further detailed studies on human spermatogenesis and spermatogenic failure.


Assuntos
Espermatogênese , Espermatozoides/citologia , Transcriptoma , Adulto , Animais , Biópsia , Diferenciação Celular , Cromatina/química , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Microdissecção e Captura a Laser , Masculino , Meiose , Camundongos , Pessoa de Meia-Idade , Família Multigênica , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Espermatogônias/citologia , Testículo/citologia
7.
Hum Reprod ; 35(8): 1797-1807, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32644109

RESUMO

STUDY QUESTION: How do high-quality human preimplantation embryos influence the endometrium to promote their own implantation? SUMMARY ANSWER: High-quality human preimplantation embryos secrete a specific microRNA (miRNA), hsa-miR-320a, which promotes migration of human endometrial stromal cells (hESCs). WHAT IS KNOWN ALREADY: We have previously shown that high-quality human preimplantation embryos excrete unknown factors that influence migration of hESCs. STUDY DESIGN, SIZE, DURATION: Embryo excreted miRNAs, specifically those excreted by high-quality embryos, were identified and their effect on hESCs was determined by measuring the migration capacity and gene expression patterns of primary isolated hESCs. PARTICIPANTS/MATERIALS, SETTING, METHODS: Embryo conditioned medium (ECM) from routine ICSI procedures was used to identify embryo excreted miRNAs. miRNome analyses were performed on ECM from individually cultured embryos with high morphological quality, with low morphological quality or empty control medium. MiRNA mimics and inhibitors were then used to further study the effect of miRNAs of interest on migration and gene expression of hESCs. Migration assays were performed using hESCs that were obtained from endometrial biopsies performed on hysterectomy specimens from women that received surgery for spotting due to a niche in a cesarean section scar. MAIN RESULTS AND THE ROLE OF CHANCE: By using miRNA mimics and inhibitors, we showed that hsa-miR-320a alone can stimulate migration of decidualized hESCs, accurately resembling the response typically triggered only by high-quality embryos. Transcriptome analysis further demonstrated that this effect is very likely mediated via altered expression of genes involved in cell adhesion and cytoskeleton organization. LIMITATIONS, REASONS FOR CAUTION: The effect of hsa-miR-320a on hESCs was measured in vitro. Further studies on the in vivo effect of hsa-miR-320a are warranted. WIDER IMPLICATIONS OF THE FINDINGS: Implantation failure is one of the major success limiting factors in human reproduction. By secreting hsa-miR-320a, high-quality human preimplantation embryos directly influence hESCs, most likely to prime the endometrium at the implantation site for successful implantation. Together, our results indicate that hsa-miR-320a may be a promising target to further increase success rates in assisted reproduction. STUDY FUNDING/COMPETING INTEREST(S): The study was funded by the Amsterdam University Medical Centers and the Amsterdam Reproduction & Development Research Institute. R.P.B., G.H. and S.M. have a patent on the use of hsa-miR-320a in assisted reproduction treatments pending. TRIAL REGISTRATION NUMBER: N/A.


Assuntos
Cesárea , MicroRNAs , Blastocisto , Movimento Celular , Endométrio , Feminino , Humanos , MicroRNAs/genética , Gravidez , Células Estromais
8.
Reprod Biomed Online ; 40(6): 867-879, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32417200

RESUMO

Many clinics offer routine genetic testing of pregnancy loss tissue. This review presents a comprehensive literature search and meta-analysis on chromosomal abnormality rates of pregnancy loss tissue from women with a single or recurrent pregnancy loss. A total of 55 studies published since 2000 were included, analysed on the prevalence of test failure rates, abnormality detection rates and percentages of trisomy, monosomy X, structural abnormalities and other clinically (ir)relevant abnormalities detected by conventional karyotyping, array-comparative genomic hybridization (aCGH), single nucleotide polymorphism (SNP) array, fluorescence in-situ hybridization (FISH) and multiplex ligation-dependent probe amplification (MLPA). The detected prevalence of chromosomal abnormalities was 48% (95% confidence interval [CI] 39-57) using aCGH, 38% (95% CI 28-49) with FISH, 25% (95% CI 12-42) using MLPA, 60% (95% CI 58-63) using SNP array and 47% (95% CI 43-51) with conventional karyotyping. The percentage of detected abnormalities did not differ between women that suffered sporadic (46%; 95% CI 39-53) or recurrent (46%; 95% CI 39-52) pregnancy loss. In view of the high prevalence of chromosomal abnormalities in pregnancy loss tissue, and the low chance of recurrence of the same chromosomal aberration, it was concluded that detection of specific chromosomal abnormalities in pregnancy loss tissue has no clinical benefit. Therefore, routine testing of pregnancy loss tissue for chromosomal abnormalities is not recommended.


Assuntos
Aberrações Cromossômicas , Análise Citogenética , Aborto Espontâneo/genética , Feminino , Humanos , Hibridização in Situ Fluorescente , Cariótipo , Gravidez
9.
Gynecol Endocrinol ; 36(4): 360-364, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31389284

RESUMO

Repeated implantation failure (RIF) is a poorly understood reproductive pathology defined by the inability to achieve a clinical pregnancy in at least three consecutive IVF cycles. In this study, we investigated whether the onset of decidualization, marked by prolactin (PRL) expression, is associated with RIF. We performed a retrospective cohort study using endometrial biopsies from women with idiopathic subfertility, that conceived naturally during the same cycle in which the biopsy was taken (group 1; n = 15) conceived naturally within three months after the biopsy was taken (group 2; n = 20), or unsuccessfully underwent six IUI cycles and three IVF cycles with transfer of at least one high-quality embryo (group 3, RIF; n = 20). Our results demonstrated that immunohistochemical PRL-staining was present in 8/15 women from group 1 (53.3%), in 1/20 women from group 2 (5.0%), and in 11/20 women from group 3 (55.0%). Increased proliferation, analyzed by Ki67 expression, was seen in women that were pregnant during the biopsy, compared to all women combined that were not pregnant (p≤.01). In conclusion, our study demonstrates that premature expression of the decidualization marker PRL during the luteal phase is associated with RIF.


Assuntos
Aborto Habitual/diagnóstico , Endométrio/metabolismo , Prolactina/metabolismo , Aborto Habitual/metabolismo , Adulto , Biomarcadores/metabolismo , Estudos de Coortes , Decídua/metabolismo , Implantação do Embrião , Endométrio/patologia , Feminino , Fertilização in vitro , Humanos , Infertilidade Feminina/diagnóstico , Infertilidade Feminina/metabolismo , Infertilidade Feminina/terapia , Gravidez , Prognóstico , Estudos Retrospectivos , Fatores de Tempo , Falha de Tratamento
10.
Biol Reprod ; 99(1): 75-86, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29590307

RESUMO

This review focuses on those mouse mutations that cause an effect on the morphology, viability, and/or behavior of primordial germ cells (PGCs) and gonocytes at specific steps of their fetal development up to the start of spermatogenesis, a few days after birth. To restrict the area covered, mice with mutations that cause abnormal hormone levels or mutations of genes not expressed in germ cells that secondarily cause spermatogenic problems are not discussed. To make our literature search as comprehensive as possible, Pubmed was searched for "(primordial germ cells OR prospermatogonia OR prespermatogonia OR gonocytes OR spermatogonia or meiosis or spermiogenesis or spermatogenesis) AND mouse AND (knockout or mutant or transgenic)." This search started at 2003 as mutants created earlier were already retrieved for a previous review. The resulting citations were then further selected for complete or partial arrests at the level of PGCs and/or gonocytes. Fifty-nine protein coding genes and two miRNA coding genes were found that arrest the development of PGCs and gonocytes at specific steps providing a better insight into the regulation of the development of these cells. As to be expected, often problems in fetal germ cell development have an effect on the fertility of the mice at adulthood.


Assuntos
Pontos de Checagem do Ciclo Celular/genética , Mutação , Oogênese/genética , Oogônios/citologia , Espermatogênese/genética , Espermatogônias/citologia , Animais , Masculino , Camundongos
11.
Biol Reprod ; 99(4): 853-863, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29767707

RESUMO

There is a general agreement that granulosa cell apoptosis is the cause of antral follicle attrition. Less clear is whether this pathway is also activated in case of preantral follicle degeneration, as several reports mention that the incidence of granulosa cell apoptosis in preantral follicles is negligible. Our objective is therefore to determine which cell-death pathways are involved in preantral and antral follicular degeneration.Atretic preantal and antral follicles were investigated using immunohistochemistry and laser-capture microdissection followed by quantitative real-time reverse transcription polymerase chain reaction. Microtubule-associated light-chain protein 3 (LC3), sequestosome 1 (SQSTM1/P62), Beclin1, autophagy-related protein 7 (ATG7), and cleaved caspase 3 (cCASP3) were used as markers for autophagy and apoptosis, respectively. P62 immunostaining was far less intense in granulosa cells of atretic compared to healthy preantral follicles, while no difference in LC3 and BECLIN1 immunostaining intensity was observed. This difference in P62 immunostaining was not observed in atretic antral follicles. mRNA levels of LC3 and P62 were not different between healthy and atretic (pre)antral follicles. ATG7 immunostaining was observed in granulosa cells of preantral atretic follicles, not in granulosa cells of degenerating antral follicles. The number of cCASP3-positive cells was negligible in preantral atretic follicles, while numerous in atretic antral follicles. Taken together, we conclude that preantral and antral follicular atresia is the result of activation of different cell-death pathways as antral follicular degeneration is initiated by massive granulosa cell apoptosis, while preantral follicular atresia occurs mainly via enhanced granulosa cell autophagy.


Assuntos
Atresia Folicular/metabolismo , Folículo Ovariano/citologia , Folículo Ovariano/metabolismo , Animais , Apoptose/genética , Apoptose/fisiologia , Autofagia/genética , Autofagia/fisiologia , Biomarcadores/metabolismo , Caspase 3/metabolismo , Feminino , Atresia Folicular/genética , Expressão Gênica , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Imuno-Histoquímica , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Proteína Sequestossoma-1/genética , Proteína Sequestossoma-1/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo
12.
Reprod Biomed Online ; 37(4): 409-414, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30228072

RESUMO

RESEARCH QUESTION: How stable is the pH of human preimplantation embryo culture media during IVF culture and is there variation in pH between batches of culture media? DESIGN: To evaluate pH stability, three batches of three culture media were incubated in triplicate without embryos (sham culture) at CO2 levels recommended by the manufacturers (5% or 6%) for 4 days. To evaluate differences in pH between batches, the pH of three batches of five culture media was measured in triplicate during 1 day of sham culture. Linear mixed models were used for the analysis. RESULTS: An increase in pH during 4 days of culture was found in all three culture media, but the observed increased values were within the generally accepted range for clinical practice (pH 7.2-7.4). One medium was pH 7.1 in the first 2 days, but this was within the range provided by the manufacturer for that medium. Three out of five analysed media showed batch variation in pH that exceeded the generally accepted range for clinical practice. CONCLUSIONS: A relevant difference in pH was found between batches of human preimplantation embryo culture media. This suggests that the CO2 level of incubators may need to be adjusted for new batches of culture medium based on measured pH, to anticipate batch variability and safely accommodate limited pH increase over time. This study was unable to identify the cause of the differences in pH between batches, and further investigation on a larger number of batches and other media seems warranted.


Assuntos
Meios de Cultura/normas , Técnicas de Cultura Embrionária , Humanos , Concentração de Íons de Hidrogênio
13.
Chromosoma ; 125(1): 15-27, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25947290

RESUMO

The Smc5/6 complex, along with cohesin and condensin, is a member of the structural maintenance of chromosome (SMC) family, large ring-like protein complexes that are essential for chromatin structure and function. Thanks to numerous studies of the mitotic cell cycle, Smc5/6 has been implicated to have roles in homologous recombination, restart of stalled replication forks, maintenance of ribosomal DNA (rDNA) and heterochromatin, telomerase-independent telomere elongation, and regulation of chromosome topology. The nature of these functions implies that the Smc5/6 complex also contributes to the profound chromatin changes, including meiotic recombination, that characterize meiosis. Only recently, studies in diverse model organisms have focused on the potential meiotic roles of the Smc5/6 complex. Indeed, Smc5/6 appears to be essential for meiotic recombination. However, due to both the complexity of the process of meiosis and the versatility of the Smc5/6 complex, many additional meiotic functions have been described. In this review, we provide a clear overview of the multiple functions found so far for the Smc5/6 complex in meiosis. Additionally, we compare these meiotic functions with the known mitotic functions in an attempt to find a common denominator and thereby create clarity in the field of Smc5/6 research.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Segregação de Cromossomos , Recombinação Homóloga , Meiose/genética , Animais , Cromatina/metabolismo , Proteínas Cromossômicas não Histona , DNA/metabolismo , Reparo do DNA , Replicação do DNA , Humanos , Proteínas de Saccharomyces cerevisiae/fisiologia , Telômero/metabolismo , Leveduras/metabolismo
14.
Reproduction ; 154(3): 181-195, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28576919

RESUMO

Spermatogenesis, starting with spermatogonial differentiation, is characterized by ongoing and dramatic alterations in composition and function of chromatin. Failure to maintain proper chromatin dynamics during spermatogenesis may lead to mutations, chromosomal aberrations or aneuploidies. When transmitted to the offspring, these can cause infertility or congenital malformations. The structural maintenance of chromosomes (SMC) 5/6 protein complex has recently been described to function in chromatin modeling and genomic integrity maintenance during spermatogonial differentiation and meiosis. Among the subunits of the SMC5/6 complex, non-SMC element 2 (NSMCE2) is an important small ubiquitin-related modifier (SUMO) ligase. NSMCE2 has been reported to be essential for mouse development, prevention of cancer and aging in adult mice and topological stress relief in human somatic cells. By using in vitro cultured primary mouse spermatogonial stem cells (SSCs), referred to as male germline stem (GS) cells, we investigated the function of NSMCE2 during spermatogonial proliferation and differentiation. We first optimized a protocol to generate genetically modified GS cell lines using CRISPR-Cas9 and generated an Nsmce2-/- GS cell line. Using this Nsmce2-/- GS cell line, we found that NSMCE2 was dispensable for proliferation, differentiation and topological stress relief in mouse GS cells. Moreover, RNA sequencing analysis demonstrated that the transcriptome was only minimally affected by the absence of NSMCE2. Only differential expression of Sgsm1 appeared highly significant, but with SGSM1 protein levels being unaffected without NSMCE2. Hence, despite the essential roles of NSMCE2 in somatic cells, chromatin integrity maintenance seems differentially regulated in the germline.


Assuntos
Diferenciação Celular , Proliferação de Células , Ligases/fisiologia , Meiose/fisiologia , Espermatogênese/fisiologia , Espermatogônias/citologia , Células-Tronco/citologia , Animais , Regulação da Expressão Gênica , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos DBA , Espermatogônias/metabolismo , Células-Tronco/metabolismo
15.
Int J Mol Sci ; 17(11)2016 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-27792189

RESUMO

The structural maintenance of chromosomes (SMC) protein complexes shape and regulate the structure and dynamics of chromatin, thereby controlling many chromosome-based processes such as cell cycle progression, differentiation, gene transcription and DNA repair. The SMC5/6 complex is previously described to promote DNA double-strand breaks (DSBs) repair by sister chromatid recombination, and found to be essential for resolving recombination intermediates during meiotic recombination. Moreover, in budding yeast, SMC5/6 provides structural organization and topological stress relief during replication in mitotically dividing cells. Despite the essential nature of the SMC5/6 complex, the versatile mechanisms by which SMC5/6 functions and its molecular regulation in mammalian cells remain poorly understood. By using a human osteosarcoma cell line (U2OS), we show that after the CRISPR-Cas9-mediated removal of the SMC5/6 subunit NSMCE2, treatment with the topoisomerase II inhibitor etoposide triggered an increased sensitivity in cells lacking NSMCE2. In contrast, NSMCE2 appeared not essential for a proper DNA damage response or cell survival after DSB induction by ionizing irradiation (IR). Interestingly, by way of immunoprecipitations (IPs) and mass spectrometry, we found that the SMC5/6 complex physically interacts with the DNA topoisomerase II α (TOP2A). We therefore propose that the SMC5/6 complex functions in resolving TOP2A-mediated DSB-repair intermediates generated during replication.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Ligases/metabolismo , Antígenos de Neoplasias/metabolismo , Sistemas CRISPR-Cas , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos da radiação , Proteínas Cromossômicas não Histona , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/metabolismo , Etoposídeo/efeitos adversos , Humanos , Ligases/genética , Proteínas de Ligação a Poli-ADP-Ribose , Mapas de Interação de Proteínas , Inibidores da Topoisomerase II/efeitos adversos
16.
Biol Reprod ; 91(1): 22, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24855106

RESUMO

Genome integrity is crucial for safe reproduction. Therefore, chromatin structure and dynamics should be tightly regulated during germ cell development. Chromatin structure and function are in large part determined by the structural maintenance of chromosomes (SMC) protein complexes, of which SMC5/6 recently has been shown to be involved in both spermatogonial differentiation and meiosis during mouse spermatogenesis. We therefore investigated the role of this complex in human spermatogenesis. We found SMC6 to be expressed in the human testis and present in a subset of type Adark and type Apale spermatogonia, all spermatocytes, and round spermatids. During human meiosis, SMC5/6 is located at the synaptonemal complex (SC), the XY body, and at the centromeres during meiotic metaphases. However, in contrast to mouse spermatogenesis, SMC6 is not located at pericentromeric heterochromatin in human spermatogenic cells, indicating subtle but perhaps important differences in not only SMC5/6 function but maybe also in maintenance of genomic integrity at the repetitive pericentromeric regions. Nonetheless, our data clearly indicate that the SMC5/6 complex, as shown in mice, is involved in numerous crucial processes during human spermatogenesis, such as in spermatogonial development, on the SC between synapsed chromosomes, and in DNA double-strand break repair on unsynapsed chromosomes during pachynema.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Espermatogênese/fisiologia , Testículo/metabolismo , Proteínas Cromossômicas não Histona , Humanos , Masculino , Meiose/fisiologia , Espermátides/metabolismo , Espermatócitos/metabolismo , Espermatogônias/metabolismo
17.
Cell Death Dis ; 15(1): 38, 2024 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-38216586

RESUMO

In principle, germline cells possess the capability to transmit a nearly unaltered set of genetic material to infinite future generations, whereas somatic cells are limited by strict growth constraints necessary to assure an organism's physical structure and eventual mortality. As the potential to replicate indefinitely is a key feature of cancer, we hypothesized that the activation of a "germline program" in somatic cells can contribute to oncogenesis. Our group recently described over one thousand germline specific genes that can be ectopically expressed in cancer, yet how germline specific processes contribute to the malignant properties of cancer is poorly understood. We here show that the expression of germ cell/cancer (GC) genes correlates with malignancy in lung adenocarcinoma (LUAD). We found that LUAD cells expressing more GC genes can repair DNA double strand breaks more rapidly, show higher rates of proliferation and are more resistant to ionizing radiation, compared to LUAD cells that express fewer GC genes. In particular, we identified the HORMA domain protein regulator TRIP13 to be predominantly responsible for this malignant phenotype, and that TRIP13 inhibition or expression levels affect the response to ionizing radiation and subsequent DNA repair. Our results demonstrate that GC genes are viable targets in oncology, as they induce increased radiation resistance and increased propagation in cancer cells. Because their expression is normally restricted to germline cells, we anticipate that GC gene directed therapeutic options will effectively target cancer, with limited side effects besides (temporary) infertility.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Humanos , Reparo do DNA/genética , Adenocarcinoma de Pulmão/genética , DNA , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/radioterapia , Neoplasias Pulmonares/metabolismo , Células Germinativas/metabolismo , ATPases Associadas a Diversas Atividades Celulares/metabolismo , Proteínas de Ciclo Celular/metabolismo
18.
Biochim Biophys Acta ; 1822(12): 1838-50, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22366765

RESUMO

Spermatogenesis is a complex developmental process that ultimately generates mature spermatozoa. This process involves a phase of proliferative expansion, meiosis, and cytodifferentiation. Mouse models have been widely used to study spermatogenesis and have revealed many genes and molecular mechanisms that are crucial in this process. Although meiosis is generally considered as the most crucial phase of spermatogenesis, mouse models have shown that pre-meiotic and post-meiotic phases are equally important. Using knowledge generated from mouse models and in vitro studies, the current review provides an overview of the molecular control of rodent spermatogenesis. Finally, we briefly relate this knowledge to fertility problems in humans and discuss implications for future research. This article is part of a Special Issue entitled: Molecular Genetics of Human Reproductive Failure.


Assuntos
Roedores/fisiologia , Espermatogênese/genética , Animais , Masculino
19.
Nat Methods ; 7(6): 473-8, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20453865

RESUMO

The ubiquitin-proteasome system (UPS) orchestrates many cellular and tissue-specific processes by degrading damaged and key regulatory proteins. To enable investigation of UPS activity in different cell types in a living animal, we developed a photoconvertible fluorescent UPS reporter system for live imaging and quantification of protein degradation in Caenorhabditis elegans. Our reporter consists of the photoconvertible fluorescent protein Dendra2 targeted for proteasomal degradation by fusion to the UbG76V mutant form of ubiquitin. In contrast to previous reporters, this system permits quantification of UPS activity independently of protein synthesis. Our reporter revealed that UPS-mediated protein degradation varies in a cell type-specific and age-dependent manner in C. elegans.


Assuntos
Caenorhabditis elegans/metabolismo , Proteínas Luminescentes/metabolismo , Complexo de Endopeptidases do Proteassoma/fisiologia , Ubiquitina/metabolismo , Envelhecimento/metabolismo , Animais , Proteínas de Caenorhabditis elegans/fisiologia , Dopamina/fisiologia , Microscopia de Fluorescência , Interferência de RNA , Receptores Citoplasmáticos e Nucleares/fisiologia , Ácido gama-Aminobutírico/fisiologia
20.
Curr Top Dev Biol ; 151: 345-369, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36681476

RESUMO

Successful in vitro spermatogenesis would generate functional haploid spermatids, and thus, form the basis for novel approaches to treat patients with impaired spermatogenesis or develop alternative strategies for male fertility preservation. Several culture strategies, including cell cultures using various stem cells and ex vivo cultures of testicular tissue, have been investigated to recapitulate spermatogenesis in vitro. Although some studies have described complete meiosis and subsequent generation of functional spermatids, key meiotic events, such as chromosome synapsis and homologous recombination required for successful meiosis and faithful in vitro-derived gametes, are often not reported. To guarantee the generation of in vitro-formed spermatids without persistent DNA double-strand breaks (DSBs) and chromosomal aberrations, criteria to evaluate whether all meiotic events are completely executed in vitro need to be established. In vivo, these meiotic events are strictly monitored by meiotic checkpoints that eliminate aberrant spermatocytes. To establish criteria to evaluate in vitro meiosis, we review the meiotic events and checkpoints that have been investigated by previous in vitro spermatogenesis studies. We found that, although major meiotic events such as initiation of DSBs and recombination, complete chromosome synapsis, and XY-body formation can be achieved in vitro, crossover formation, chiasmata frequency, and checkpoint mechanisms have been mostly ignored. In addition, complete spermiogenesis, during which round spermatids differentiate into elongated spermatids, has not been achieved in vitro by various cell culture strategies. Finally, we discuss the implications of meiotic checkpoints for in vitro spermatogenesis protocols and future clinical use.


Assuntos
Espermátides , Espermatogênese , Humanos , Masculino , Espermatogênese/genética , Espermatócitos , Meiose , Cromossomos Sexuais
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa