Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Methods Mol Biol ; 2770: 151-170, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38351453

RESUMEN

The regulation of female fertility in mammals depends on critical processes during oocyte development and maturation. Therefore, it is crucial to use specific approaches when studying mammalian female fertility to preserve ovary and oocyte structures effectively. The methods of collecting and culturing ovaries and oocytes play an essential role in the study of mammalian follicle development and oocyte quality. This chapter presents a collection of protocols that focus on various methods for studying mammalian ovaries and oocytes, providing researchers with a variety of approaches to choose from.


Asunto(s)
Folículo Ovárico , Ovario , Animales , Embarazo , Ratones , Femenino , Oocitos/fisiología , Oogénesis , Técnicas Citológicas , Mamíferos
2.
Front Cell Dev Biol ; 11: 1127440, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36910159

RESUMEN

Meiosis is a specialized cell division that generates haploid gametes and is critical for successful sexual reproduction. During the extended meiotic prophase I, homologous chromosomes progressively pair, synapse and desynapse. These chromosomal dynamics are tightly integrated with meiotic recombination (MR), during which programmed DNA double-strand breaks (DSBs) are formed and subsequently repaired. Consequently, parental chromosome arms reciprocally exchange, ultimately ensuring accurate homolog segregation and genetic diversity in the offspring. Surveillance mechanisms carefully monitor the MR and homologous chromosome synapsis during meiotic prophase I to avoid producing aberrant chromosomes and defective gametes. Errors in these critical processes would lead to aneuploidy and/or genetic instability. Studies of mutation in mouse models, coupled with advances in genomic technologies, lead us to more clearly understand how meiosis is controlled and how meiotic errors are linked to mammalian infertility. Here, we review the genetic regulations of these major meiotic events in mice and highlight our current understanding of their surveillance mechanisms. Furthermore, we summarize meiotic prophase genes, the mutations that activate the surveillance system leading to meiotic prophase arrest in mouse models, and their corresponding genetic variants identified in human infertile patients. Finally, we discuss their value for the diagnosis of causes of meiosis-based infertility in humans.

3.
Int J Mol Sci ; 23(17)2022 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-36077210

RESUMEN

Meiosis initiates with the formation of double strand breaks (DSBs) throughout the genome. To avoid genomic instability, these DSBs need to be correctly repaired by homologous recombination. Surveillance mechanisms involving the DNA damage response (DDR) pathway ATM-CHK2-p53 can detect the persistence of unrepaired DBSs and activate the recombination-dependent arrest at the pachytene stage. However, a complete understanding of p53 functions under normal physiological conditions remains lacking. Here, we report a detailed analysis of the p53 role during meiotic prophase in mice spermatocytes. We show that the absence of p53 regulates prophase progression by slowing down the pachytene stage when the recombination-dependent arrest occurs. Furthermore, our results show that p53 is necessary for proper crossover (CO) formation and localization. Our study contributes to a deeper understanding of p53 roles during the meiotic prophase.


Asunto(s)
Roturas del ADN de Doble Cadena , Meiosis , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Masculino , Ratones , Profase , Espermatocitos/metabolismo , Proteína p53 Supresora de Tumor/genética
4.
Curr Biol ; 32(2): 462-469.e6, 2022 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-34847353

RESUMEN

The presence of population-specific phenotypes often reflects local adaptation or barriers to gene flow. The co-occurrence of phenotypic polymorphisms that are restricted within the range of a highly mobile species is more difficult to explain. An example of such polymorphisms is in the common quail Coturnix coturnix, a small migratory bird that moves widely during the breeding season in search of new mating opportunities, following ephemeral habitats,1,2 and whose females may lay successive clutches at different locations while migrating.3 In spite of this vagility, previous studies reported a higher frequency of heavier males with darker throat coloration in the southwest of the distribution (I. Jiménez-Blasco et al., 2015, Int. Union Game Biol., conference). We used population genomics and cytogenetics to explore the basis of this polymorphism and discovered a large inversion in the genome of the common quail. This inversion extends 115 Mbp in length and encompasses more than 7,000 genes (about 12% of the genome), producing two very different forms. Birds with the inversion are larger, have darker throat coloration and rounder wings, are inferred to have poorer flight efficiency, and are geographically restricted despite the high mobility of the species. Stable isotope analyses confirmed that birds carrying the inversion have shorter migratory distances or do not migrate. However, we found no evidence of pre- or post-zygotic isolation, indicating the two forms commonly interbreed and that the polymorphism remains locally restricted because of the effect on behavior. This illustrates a genomic mechanism underlying maintenance of geographically structured polymorphisms despite interbreeding with a lineage with high mobility.


Asunto(s)
Coturnix , Genoma , Animales , Inversión Cromosómica , Coturnix/genética , Femenino , Flujo Génico , Masculino , Estaciones del Año
5.
Rev Esp Patol ; 54(4): 242-249, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34544554

RESUMEN

Adenoid cystic carcinoma of the breast (ACCB) is a rare triple negative tumor (TNT) with an excellent prognosis in most cases. Three different histologic types are recognized: classic ACCB, solid basaloid ACCB (SB-ACCB), and ACCB with high-grade transformation. A majority of these tumors show characteristic molecular and immunohistochemical (IHC) features, with fusion of MYB and NFIB genes and overexpression of MYB, respectively. Basaloid carcinomas of the breast (BCB) are infrequently described. They resemble SB-ACCB and TNT of no special type (TNT-NST). We have studied the clinicopathological features of 17 ACCB and 9 BCB, investigating the expression of MYB by IHC and the rearrangements of MYB by fluorescence in situ hybridization (FISH). MYB was expressed by IHC in 15 ACCB and in 3 BCB. MYB FISH detected rearrangements in 11 ACCB and in 2 BCB. After a mean follow-up of 90 months, with a range of 12-204 months, 2 patients with ACCB with high-grade transformation and 1 patient with BCB developed metastases and died of disease. In summary, most ACCB have a good prognosis, but tumors with adverse histopathological features may metastasize. BCB may overlap with ACCB and TNT-NST, and their prognosis should be further studied.


Asunto(s)
Neoplasias de la Mama , Carcinoma Adenoide Quístico , Mama , Carcinoma Adenoide Quístico/genética , Femenino , Humanos , Hibridación Fluorescente in Situ , Pronóstico
6.
Cell ; 183(6): 1464-1466, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-33306952

RESUMEN

In this issue of Cell, Zuccaro and colleagues show that on-target Cas9-mediated double-strand breaks cause chromosome loss or mis-repair of the disease allele in > 90% of human embryos. End joining repair pathways dominate, causing small insertions or deletions, which raises serious questions about using double-strand breaks for "gene surgery".


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Alelos , Sistemas CRISPR-Cas , Cromosomas , Humanos
7.
PLoS Genet ; 16(11): e1009067, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33206637

RESUMEN

Mammalian oogonia proliferate without completing cytokinesis, forming cysts. Within these, oocytes differentiate and initiate meiosis, promoting double-strand break (DSBs) formation, which are repaired by homologous recombination (HR) causing the pairing and synapsis of the homologs. Errors in these processes activate checkpoint mechanisms, leading to apoptosis. At the end of prophase I, in contrast with what is observed in spermatocytes, oocytes accumulate unrepaired DSBs. Simultaneously to the cyst breakdown, there is a massive oocyte death, which has been proposed to be necessary to enable the individualization of the oocytes to form follicles. Based upon all the above-mentioned information, we hypothesize that the apparently inefficient HR occurring in the oocytes may be a requirement to first eliminate most of the oocytes and enable cyst breakdown and follicle formation. To test this idea, we compared perinatal ovaries from control and mutant mice for the effector kinase of the DNA Damage Response (DDR), CHK2. We found that CHK2 is required to eliminate ~50% of the fetal oocyte population. Nevertheless, the number of oocytes and follicles found in Chk2-mutant ovaries three days after birth was equivalent to that of the controls. These data revealed the existence of another mechanism capable of eliminating oocytes. In vitro inhibition of CHK1 rescued the oocyte number in Chk2-/- mice, implying that CHK1 regulates postnatal oocyte death. Moreover, we found that CHK1 and CHK2 functions are required for the timely breakdown of the cyst and to form follicles. Thus, we uncovered a novel CHK1 function in regulating the oocyte population in mice. Based upon these data, we propose that the CHK1- and CHK2-dependent DDR controls the number of oocytes and is required to properly break down oocyte cysts and form follicles in mammals.


Asunto(s)
Daño del ADN/genética , Oogonios/metabolismo , Folículo Ovárico/metabolismo , Animales , Apoptosis/fisiología , Proteínas de Ciclo Celular/genética , Quinasa de Punto de Control 2/genética , Quinasa de Punto de Control 2/metabolismo , Quistes/metabolismo , Daño del ADN/fisiología , Endodesoxirribonucleasas/genética , Endodesoxirribonucleasas/metabolismo , Femenino , Meiosis/fisiología , Profase Meiótica I/fisiología , Ratones , Ratones Endogámicos C57BL , Oocitos/metabolismo , Oocitos/fisiología , Oogonios/fisiología , Folículo Ovárico/fisiología , Ovario/metabolismo , Progesterona/metabolismo
8.
Chromosoma ; 128(3): 489-500, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31489491

RESUMEN

Mammalian female fertility relies on the proper development of follicles. Right after birth in the mouse, oocytes associate with somatic ovarian cells to form follicles. These follicles grow during the adult lifetime to produce viable gametes. In this study, we analyzed the role of the ATM and rad3-related (ATR) kinase in mouse oogenesis and folliculogenesis using a hypomorphic mutation of the Atr gene (Murga et al. 2009). Female mice homozygotes for this allele have been reported to be sterile. Our data show that female meiotic prophase is not grossly altered when ATR levels are reduced. However, follicle development is substantially compromised, since Atr mutant ovaries present a decrease of growing follicles. Comprehensive analysis of follicular cell death and proliferation suggest that wild-type levels of ATR are required to achieve optimal follicular development. Altogether, these findings suggest that reduced ATR expression causes sterility due to defects in follicular progression rather than in meiotic recombination. We discuss the implications of these findings for the use of ATR inhibitors such as anti-cancer drugs and its possible side-effects on female fertility.


Asunto(s)
Oogénesis , Folículo Ovárico/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Femenino , Meiosis/genética , Ratones , Oogénesis/genética , Ovario/metabolismo , Profase/genética
9.
PLoS Genet ; 15(8): e1008316, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31437213

RESUMEN

The ubiquitin proteasome system regulates meiotic recombination in yeast through its association with the synaptonemal complex, a 'zipper'-like structure that holds homologous chromosome pairs in synapsis during meiotic prophase I. In mammals, the proteasome activator subunit PA200 targets acetylated histones for degradation during somatic DNA double strand break repair and during histone replacement during spermiogenesis. We investigated the role of the testis-specific proteasomal subunit α4s (PSMA8) during spermatogenesis, and found that PSMA8 was localized to and dependent on the central region of the synaptonemal complex. Accordingly, synapsis-deficient mice show delocalization of PSMA8. Moreover, though Psma8-deficient mice are proficient in meiotic homologous recombination, there are alterations in the proteostasis of several key meiotic players that, in addition to the known substrate acetylated histones, have been shown by a proteomic approach to interact with PSMA8, such as SYCP3, SYCP1, CDK1 and TRIP13. These alterations lead to an accumulation of spermatocytes in metaphase I and II which either enter massively into apoptosis or give rise to a low number of aberrant round spermatids that apoptose before histone replacement takes place.


Asunto(s)
Fertilidad/genética , Infertilidad Masculina/genética , Metafase/genética , Complejo de la Endopetidasa Proteasomal/genética , Subunidades de Proteína/genética , Animales , Apoptosis/genética , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Noqueados , Proteínas Nucleares/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Subunidades de Proteína/metabolismo , Espermatocitos/metabolismo , Espermatogénesis/genética , Complejo Sinaptonémico/metabolismo , Testículo/citología , Testículo/metabolismo
10.
Nat Commun ; 9(1): 3961, 2018 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-30305635

RESUMEN

The DNA-damage repair pathway homologous recombination (HR) requires factors that promote the activity of strand-exchange protein RAD51 and its meiosis-specific homolog DMC1. Here we show that the Shu complex SWS1-SWSAP1, a candidate for one such HR regulator, is dispensable for mouse viability but essential for male and female fertility, promoting the assembly of RAD51 and DMC1 on early meiotic HR intermediates. Only a fraction of mutant meiocytes progress to form crossovers, which are crucial for chromosome segregation, demonstrating crossover homeostasis. Remarkably, loss of the DNA damage checkpoint kinase CHK2 rescues fertility in females without rescuing crossover numbers. Concomitant loss of the BRCA2 C terminus aggravates the meiotic defects in Swsap1 mutant spermatocytes, suggesting an overlapping role with the Shu complex during meiotic HR. These results demonstrate an essential role for SWS1-SWSAP1 in meiotic progression and emphasize the complex interplay of factors that ensure recombinase function.


Asunto(s)
Meiosis , Recombinación Genética , Animales , Proteína BRCA2/química , Proteína BRCA2/metabolismo , Secuencia de Bases , Proteínas de Ciclo Celular/metabolismo , Quinasa de Punto de Control 2/genética , Emparejamiento Cromosómico , Intercambio Genético , ADN/metabolismo , Femenino , Infertilidad/patología , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación/genética , Proteínas Nucleares/metabolismo , Proteínas de Unión a Fosfato , Recombinasa Rad51/metabolismo , Recombinación Genética/genética , Espermatozoides/metabolismo
11.
Nat Commun ; 9(1): 2621, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29976923

RESUMEN

Meiotic cells undergo genetic exchange between homologs through programmed DNA double-strand break (DSB) formation, recombination and synapsis. In mice, the DNA damage-regulated phosphatidylinositol-3-kinase-like kinase (PIKK) ATM regulates all of these processes. However, the meiotic functions of the PIKK ATR have remained elusive, because germline-specific depletion of this kinase is challenging. Here we uncover roles for ATR in male mouse prophase I progression. ATR deletion causes chromosome axis fragmentation and germ cell elimination at mid pachynema. This elimination cannot be rescued by deletion of ATM and the third DNA damage-regulated PIKK, PRKDC, consistent with the existence of a PIKK-independent surveillance mechanism in the mammalian germline. ATR is required for synapsis, in a manner genetically dissociable from DSB formation. ATR also regulates loading of recombinases RAD51 and DMC1 to DSBs and recombination focus dynamics on synapsed and asynapsed chromosomes. Our studies reveal ATR as a critical regulator of mouse meiosis.


Asunto(s)
Cromosomas de los Mamíferos/genética , Roturas del ADN de Doble Cadena , Meiosis/genética , Espermatocitos/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Emparejamiento Cromosómico/genética , Cromosomas de los Mamíferos/metabolismo , Hibridación Fluorescente in Situ , Masculino , Profase Meiótica I/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Unión a Fosfato , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo
12.
Nat Commun ; 9(1): 2622, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29977027

RESUMEN

Precise execution of recombination during meiosis is essential for forming chromosomally-balanced gametes. Meiotic recombination initiates with the formation and resection of DNA double-strand breaks (DSBs). Cellular responses to meiotic DSBs are critical for efficient repair and quality control, but molecular features of these remain poorly understood, particularly in mammals. Here we report that the DNA damage response protein kinase ATR is crucial for meiotic recombination and completion of meiotic prophase in mice. Using a hypomorphic Atr mutation and pharmacological inhibition of ATR in vivo and in cultured spermatocytes, we show that ATR, through its effector kinase CHK1, promotes efficient RAD51 and DMC1 assembly at RPA-coated resected DSB sites and establishment of interhomolog connections during meiosis. Furthermore, our findings suggest that ATR promotes local accumulation of recombination markers on unsynapsed axes during meiotic prophase to favor homologous chromosome synapsis. These data reveal that ATR plays multiple roles in mammalian meiotic recombination.


Asunto(s)
Roturas del ADN de Doble Cadena , Recombinación Homóloga , Meiosis/genética , Espermatocitos/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Emparejamiento Cromosómico/genética , Hibridación Fluorescente in Situ , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Unión a Fosfato , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Testículo/citología , Testículo/metabolismo
13.
J Cell Sci ; 131(6)2018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29437857

RESUMEN

In somatic cells, H2afx and Mdc1 are close functional partners in DNA repair and damage response. However, it is not known whether they are also involved in the maintenance of genome integrity in meiosis. By analyzing chromosome dynamics in H2afx-/- spermatocytes, we found that the synapsis of autosomes and X-Y chromosomes was impaired in a fraction of cells. Such defects correlated with an abnormal recombination profile. Conversely, Mdc1 was dispensable for the synapsis of the autosomes and played only a minor role in X-Y synapsis, compared with the action of H2afx This suggested that those genes have non-overlapping functions in chromosome synapsis. However, we observed that both genes play a similar role in the assembly of MLH3 onto chromosomes, a key step in crossover formation. Moreover, we show that H2afx and Mdc1 cooperate in promoting the activation of the recombination-dependent checkpoint, a mechanism that restrains the differentiation of cells with unrepaired DSBs. This occurs by a mechanism that involves P53. Overall, our data show that, in male germ cells, H2afx and Mdc1 promote the maintenance of genome integrity.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Histonas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Espermatocitos/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Ciclo Celular , Emparejamiento Cromosómico , Inestabilidad Genómica , Genómica , Histonas/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas MutL/genética , Proteínas MutL/metabolismo , Recombinación Genética , Cromosomas Sexuales/genética , Cromosomas Sexuales/metabolismo , Espermatocitos/citología
14.
PLoS Genet ; 13(6): e1006845, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28617799

RESUMEN

To protect germ cells from genomic instability, surveillance mechanisms ensure meiosis occurs properly. In mammals, spermatocytes that display recombination defects experience a so-called recombination-dependent arrest at the pachytene stage, which relies on the MRE11 complex-ATM-CHK2 pathway responding to unrepaired DNA double-strand breaks (DSBs). Here, we asked if p53 family members-targets of ATM and CHK2-participate in this arrest. We bred double-mutant mice combining a mutation of a member of the p53 family (p53, TAp63, or p73) with a Trip13 mutation. Trip13 deficiency triggers a recombination-dependent response that arrests spermatocytes in pachynema before they have incorporated the testis-specific histone variant H1t into their chromatin. We find that deficiency for either p53 or TAp63, but not p73, allowed spermatocytes to progress further into meiotic prophase despite the presence of numerous unrepaired DSBs. Even so, the double mutant spermatocytes apoptosed at late pachynema because of sex body deficiency; thus p53 and TAp63 are dispensable for arrest caused by sex body defects. These data affirm that recombination-dependent and sex body-deficient arrests occur via genetically separable mechanisms.


Asunto(s)
Meiosis/genética , Fosfoproteínas/genética , Recombinación Genética , Transactivadores/genética , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis/genética , Puntos de Control del Ciclo Celular , Cromatina/genética , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Histonas/genética , Masculino , Ratones , Fase Paquiteno/genética , Espermatocitos/crecimiento & desarrollo , Espermatocitos/metabolismo , Testículo/crecimiento & desarrollo , Testículo/metabolismo
15.
Nat Commun ; 6: 7676, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-26158450

RESUMEN

CEP63 is a centrosomal protein that facilitates centriole duplication and is regulated by the DNA damage response. Mutations in CEP63 cause Seckel syndrome, a human disease characterized by microcephaly and dwarfism. Here we demonstrate that Cep63-deficient mice recapitulate Seckel syndrome pathology. The attrition of neural progenitor cells involves p53-dependent cell death, and brain size is rescued by the deletion of p53. Cell death is not the result of an aberrant DNA damage response but is triggered by centrosome-based mitotic errors. In addition, Cep63 loss severely impairs meiotic recombination, leading to profound male infertility. Cep63-deficient spermatocytes display numerical and structural centrosome aberrations, chromosome entanglements and defective telomere clustering, suggesting that a reduction in centrosome-mediated chromosome movements underlies recombination failure. Our results provide novel insight into the molecular pathology of microcephaly and establish a role for the centrosome in meiotic recombination.


Asunto(s)
Proteínas de Ciclo Celular/genética , Centrosoma/metabolismo , Enanismo/genética , Recombinación Homóloga/genética , Meiosis/genética , Microcefalia/genética , Espermatocitos/metabolismo , Proteína p53 Supresora de Tumor/genética , Animales , Daño del ADN , Facies , Inmunohistoquímica , Masculino , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Recombinación Genética/genética , Recuento de Espermatozoides , Espermatocitos/patología
16.
PLoS Genet ; 11(3): e1005017, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25768017

RESUMEN

Most mutations that compromise meiotic recombination or synapsis in mouse spermatocytes result in arrest and apoptosis at the pachytene stage of the first meiotic prophase. Two main mechanisms are thought to trigger arrest: one independent of the double-strand breaks (DSBs) that initiate meiotic recombination, and another activated by persistent recombination intermediates. Mechanisms underlying the recombination-dependent arrest response are not well understood, so we sought to identify factors involved by examining mutants deficient for TRIP13, a conserved AAA+ ATPase required for the completion of meiotic DSB repair. We find that spermatocytes with a hypomorphic Trip13 mutation (Trip13mod/mod) arrest with features characteristic of early pachynema in wild type, namely, fully synapsed chromosomes without incorporation of the histone variant H1t into chromatin. These cells then undergo apoptosis, possibly in response to the arrest or in response to a defect in sex body formation. However, TRIP13-deficient cells that additionally lack the DSB-responsive kinase ATM progress further, reaching an H1t-positive stage (i.e., similar to mid/late pachynema in wild type) despite the presence of unrepaired DSBs. TRIP13-deficient spermatocytes also progress to an H1t-positive stage if ATM activity is attenuated by hypomorphic mutations in Mre11 or Nbs1 or by elimination of the ATM-effector kinase CHK2. These mutant backgrounds nonetheless experience an apoptotic block to further spermatogenic progression, most likely caused by failure to form a sex body. DSB numbers are elevated in Mre11 and Nbs1 hypomorphs but not Chk2 mutants, thus delineating genetic requirements for the ATM-dependent negative feedback loop that regulates DSB numbers. The findings demonstrate for the first time that ATM-dependent signaling enforces the normal pachytene response to persistent recombination intermediates. Our work supports the conclusion that recombination defects trigger spermatocyte arrest via pathways than are genetically distinct from sex body failure-promoted apoptosis and confirm that the latter can function even when recombination-dependent arrest is inoperative. Implications of these findings for understanding the complex relationships between spermatocyte arrest and apoptosis are discussed.


Asunto(s)
Adenosina Trifosfatasas/genética , Proteínas de Ciclo Celular/genética , Quinasa de Punto de Control 2/genética , Enzimas Reparadoras del ADN/genética , Proteínas de Unión al ADN/genética , Proteínas Nucleares/genética , Espermatocitos/crecimiento & desarrollo , ATPasas Asociadas con Actividades Celulares Diversas , Animales , Apoptosis/genética , Proteínas de la Ataxia Telangiectasia Mutada/genética , Puntos de Control del Ciclo Celular/genética , Emparejamiento Cromosómico/genética , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Proteína Homóloga de MRE11 , Masculino , Ratones , Mutación , Fase Paquiteno/genética , Recombinación Genética , Transducción de Señal/genética , Espermatocitos/metabolismo
17.
Mol Biol Evol ; 32(2): 510-23, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25414125

RESUMEN

Recombination rates vary in intensity and location at the species, individual, sex and chromosome levels. Despite the fundamental biological importance of this process, the selective forces that operate to shape recombination rate and patterns are unclear. Domestication offers a unique opportunity to study the interplay between recombination and selection. In domesticates, intense selection for particular traits is imposed on small populations over many generations, resulting in organisms that differ, sometimes dramatically, in morphology and physiology from their wild ancestor. Although earlier studies suggested increased recombination rate in domesticates, a formal comparison of recombination rates between domestic mammals and their wild congeners was missing. In order to determine broad-scale recombination rate, we used immunolabeling detection of MLH1 foci as crossover markers in spermatocytes in three pairs of closely related wild and domestic species (dog and wolf, goat and ibex, and sheep and mouflon). In the three pairs, and contrary to previous suggestions, our data show that contemporary recombination rate is higher in the wild species. Subsequently, we inferred recombination breakpoints in sequence data for 16 genomic regions in dogs and wolves, each containing a locus associated with a dog phenotype potentially under selection during domestication. No difference in the number and distribution of recombination breakpoints was found between dogs and wolves. We conclude that our data indicate that strong directional selection did not result in changes in recombination in domestic mammals, and that both upper and lower bounds for crossover rates may be tightly regulated.


Asunto(s)
Variación Genética/genética , Recombinación Genética/genética , Animales , Canidae/genética , Perros , Femenino , Genómica , Cabras/genética , Masculino , Mamíferos , Ovinos/genética , Espermatocitos/metabolismo
18.
Front Genet ; 4: 37, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23532176

RESUMEN

The DNA damage response (DDR) rapidly recognizes DNA lesions and initiates the appropriate cellular programs to maintain genome integrity. This includes the coordination of cell cycle checkpoints, transcription, translation, DNA repair, metabolism, and cell fate decisions, such as apoptosis or senescence (Jackson and Bartek, 2009). DNA double-strand breaks (DSBs) represent one of the most cytotoxic DNA lesions and defects in their metabolism underlie many human hereditary diseases characterized by genomic instability (Stracker and Petrini, 2011; McKinnon, 2012). Patients with hereditary defects in the DDR display defects in development, particularly affecting the central nervous system, the immune system and the germline, as well as aberrant metabolic regulation and cancer predisposition. Central to the DDR to DSBs is the ataxia-telangiectasia mutated (ATM) kinase, a master controller of signal transduction. Understanding how ATM signaling regulates various aspects of the DDR and its roles in vivo is critical for our understanding of human disease, its diagnosis and its treatment. This review will describe the general roles of ATM signaling and highlight some recent advances that have shed light on the diverse roles of ATM and related proteins in human disease.

19.
Nat Cell Biol ; 14(4): 424-30, 2012 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-22388890

RESUMEN

Humans suffer from high rates of fetal aneuploidy, often arising from the absence of meiotic crossover recombination between homologous chromosomes. Meiotic recombination is initiated by double-strand breaks (DSBs) generated by the SPO11 transesterase. In yeast and worms, at least one buffering mechanism, crossover homeostasis, maintains crossover numbers despite variation in DSB numbers. We show here that mammals exhibit progressive homeostatic control of recombination. In wild-type mouse spermatocytes, focus numbers for early recombination proteins (RAD51, DMC1) were highly variable from cell to cell, whereas foci of the crossover marker MLH1 showed little variability. Furthermore, mice with greater or fewer copies of the Spo11 gene--with correspondingly greater or fewer numbers of early recombination foci--exhibited relatively invariant crossover numbers. Homeostatic control is enforced during at least two stages, after the formation of early recombination intermediates and later while these intermediates mature towards crossovers. Thus, variability within the mammalian meiotic program is robustly managed by homeostatic mechanisms to control crossover formation, probably to suppress aneuploidy. Meiotic recombination exemplifies how order can be progressively implemented in a self-organizing system despite natural cell-to-cell disparities in the underlying biochemical processes.


Asunto(s)
Homeostasis , Meiosis/genética , Recombinación Genética/genética , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Endodesoxirribonucleasas/genética , Endodesoxirribonucleasas/metabolismo , Masculino , Ratones , Ratones Endogámicos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Unión a Fosfato , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Espermatocitos/metabolismo
20.
PLoS Genet ; 7(7): e1002157, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21811412

RESUMEN

The mammalian Cdkn2a (Ink4a-Arf) locus encodes two tumor suppressor proteins (p16(Ink4a) and p19(Arf)) that respectively enforce the anti-proliferative functions of the retinoblastoma protein (Rb) and the p53 transcription factor in response to oncogenic stress. Although p19(Arf) is not normally detected in tissues of young adult mice, a notable exception occurs in the male germ line, where Arf is expressed in spermatogonia, but not in meiotic spermatocytes arising from them. Unlike other contexts in which the induction of Arf potently inhibits cell proliferation, expression of p19(Arf) in spermatogonia does not interfere with mitotic cell division. Instead, inactivation of Arf triggers germ cell-autonomous, p53-dependent apoptosis of primary spermatocytes in late meiotic prophase, resulting in reduced sperm production. Arf deficiency also causes premature, elevated, and persistent accumulation of the phosphorylated histone variant H2AX, reduces numbers of chromosome-associated complexes of Rad51 and Dmc1 recombinases during meiotic prophase, and yields incompletely synapsed autosomes during pachynema. Inactivation of Ink4a increases the fraction of spermatogonia in S-phase and restores sperm numbers in Ink4a-Arf doubly deficient mice but does not abrogate γ-H2AX accumulation in spermatocytes or p53-dependent apoptosis resulting from Arf inactivation. Thus, as opposed to its canonical role as a tumor suppressor in inducing p53-dependent senescence or apoptosis, Arf expression in spermatogonia instead initiates a salutary feed-forward program that prevents p53-dependent apoptosis, contributing to the survival of meiotic male germ cells.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Espermatogonias/metabolismo , Espermatozoides/metabolismo , Animales , Apoptosis , Proteínas de Ciclo Celular/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Femenino , Técnica del Anticuerpo Fluorescente , Hormona Folículo Estimulante/sangre , Histonas/metabolismo , Immunoblotting , Hormona Luteinizante/sangre , Masculino , Meiosis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mitosis , Proteínas Nucleares/metabolismo , Proteínas de Unión a Fosfato , Recombinasa Rad51/metabolismo , Espermatogonias/citología , Testículo/citología , Testículo/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA