Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
EMBO Rep ; 24(6): e56316, 2023 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-37099396

RESUMEN

Spermatozoa have a unique genome organization. Their chromatin is almost completely devoid of histones and is formed instead of protamines, which confer a high level of compaction and preserve paternal genome integrity until fertilization. Histone-to-protamine transition takes place in spermatids and is indispensable for the production of functional sperm. Here, we show that the H3K79-methyltransferase DOT1L controls spermatid chromatin remodeling and subsequent reorganization and compaction of the spermatozoon genome. Using a mouse model in which Dot1l is knocked-out (KO) in postnatal male germ cells, we found that Dot1l-KO sperm chromatin is less compact and has an abnormal content, characterized by the presence of transition proteins, immature protamine 2 forms and a higher level of histones. Proteomic and transcriptomic analyses performed on spermatids reveal that Dot1l-KO modifies the chromatin prior to histone removal and leads to the deregulation of genes involved in flagellum formation and apoptosis during spermatid differentiation. As a consequence of these chromatin and gene expression defects, Dot1l-KO spermatozoa have less compact heads and are less motile, which results in impaired fertility.


Asunto(s)
Cromatina , Histonas , Animales , Masculino , Diferenciación Celular/genética , Cromatina/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Expresión Génica , Histonas/metabolismo , Proteómica , Semen/metabolismo , Espermatogénesis/genética , Espermatozoides/metabolismo , Ratones
2.
Nat Genet ; 54(4): 469-480, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35410378

RESUMEN

DNA methylation plays a critical role in spermatogenesis, as evidenced by the male sterility of DNA methyltransferase (DNMT) mutant mice. Here, we report a division of labor in the establishment of the methylation landscape of male germ cells and its functions in spermatogenesis. Although DNMT3C is essential for preventing retrotransposons from interfering with meiosis, DNMT3A broadly methylates the genome (with the exception of DNMT3C-dependent retrotransposons) and controls spermatogonial stem cell (SSC) plasticity. By reconstructing developmental trajectories through single-cell RNA sequencing and profiling chromatin states, we found that Dnmt3A mutant SSCs can only self-renew and no longer differentiate in association with spurious enhancer activation that enforces an irreversible stem cell gene program. Our findings therefore highlight a key function of DNA methylation in male fertility: the epigenetic programming of SSC commitment to differentiation and lifelong spermatogenesis supply.


Asunto(s)
Metilación de ADN , Espermatogénesis , Espermatogonias , Animales , Metilación de ADN/genética , Metilasas de Modificación del ADN/genética , Masculino , Ratones , Retroelementos , Espermatogénesis/genética , Espermatogonias/metabolismo , Células Madre/metabolismo
3.
Stem Cell Reports ; 17(4): 936-952, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35334216

RESUMEN

Male infertility is responsible for approximately half of all cases of reproductive issues. Spermatogenesis originates in a small pool of spermatogonial stem cells (SSCs), which are of interest for therapy of infertility but remain not well defined in humans. Using multiparametric analysis of the side population (SP) phenotype and the α-6 integrin, THY1, and ß-2 microglobulin cell markers, we identified a population of human primitive undifferentiated spermatogonia with the phenotype ß-2 microglobulin (ß-2M)-SPα-6+THY1+, which is highly enriched in stem cells. By analyzing the expression signatures of this SSC-enriched population along with other germinal progenitors, we established an exhaustive transcriptome of human spermatogenesis. Transcriptome profiling of the human ß-2M-SPα-6+THY1+ population and comparison with the profile of mouse undifferentiated spermatogonia provide insights into the molecular networks and key transcriptional regulators regulating human SSCs, including the basic-helix-loop-helix (bHLH) transcriptional repressor HES1, which we show to be implicated in maintenance of SSCs in vitro.


Asunto(s)
Células Madre Germinales Adultas , Espermatogénesis , Animales , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Espermatogénesis/genética , Espermatogonias/metabolismo , Células Madre/metabolismo , Testículo/metabolismo , Factores de Transcripción/metabolismo
4.
Stem Cell Res ; 60: 102723, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35247845

RESUMEN

In adult testis, the cell mobility is essential for spermatogonia differentiation and is suspected to regulate spermatogonial stem cell fate. Netrin-1 controls cell migration and/or survival according to the cellular context. Its involvement in some self-renewing lineages raises the possibility that Netrin-1 could have a role in spermatogenesis. We show that in addition to Sertoli cells, a fraction of murine undifferentiated spermatogonia express the Netrin-1 receptor UNC5c and that UNC5c contributes to spermatogonia differentiation. Receptor loss in Unc5crcm males leads to the concomitant accumulation of transit-amplifying progenitors and short syncytia of spermatogonia. Without altering cell death rates, the consequences of Unc5c loss worsen with age: the increase in quiescent undifferentiated progenitors associated with a higher spermatogonial stem cell enriched subset leads to the spermatocyte I decline. We demonstrate in vitro that Netrin-1 promotes a guidance effect as it repulses both undifferentiated and differentiating spermatogonia. Finally, we propose that UNC5c triggers undifferentiated spermatogonia adhesion/ migration and that the repulsive activity of Netrin-1 receptors could regulate spermatogonia differentiation, and maintain germ cell homeostasis.


Asunto(s)
Espermatogénesis , Espermatogonias , Animales , Diferenciación Celular/fisiología , Homeostasis , Masculino , Ratones , Receptores de Netrina/metabolismo , Netrina-1/metabolismo , Espermatogénesis/fisiología , Testículo
5.
Hum Mol Genet ; 31(1): 97-110, 2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34368842

RESUMEN

Fanconi anemia (FA) is a rare human genetic disorder characterized by bone marrow failure, predisposition to cancer and developmental defects including hypogonadism. Reproductive defects leading to germ cell aplasia are the most consistent phenotypes seen in FA mouse models. We examined the role of the nuclear FA core complex gene Fancg in the development of primordial germ cells (PGCs), the embryonic precursors of adult gametes, during fetal development. PGC maintenance was severely impaired in Fancg-/- embryos. We observed a defect in the number of PGCs starting at E9.5 and a strong attrition at E11.5 and E13.5. Remarkably, we observed a mosaic pattern reflecting a portion of testicular cords devoid of PGCs in E13.5 fetal gonads. Our in vitro and in vivo data highlight a potential role of Fancg in the proliferation and in the intrinsic cell motility abilities of PGCs. The random migratory process is abnormally activated in Fancg-/- PGCs, altering the migration of cells. Increased cell death and PGC attrition observed in E11.5 Fancg-/- embryos are features consistent with delayed migration of PGCs along the migratory pathway to the genital ridges. Moreover, we show that an inhibitor of RAC1 mitigates the abnormal migratory pattern observed in Fancg-/- PGCs.


Asunto(s)
Anemia de Fanconi , Animales , Movimiento Celular/genética , Anemia de Fanconi/genética , Proteína del Grupo de Complementación G de la Anemia de Fanconi/metabolismo , Células Germinativas/metabolismo , Gónadas/metabolismo , Ratones , Transducción de Señal
7.
Int J Mol Sci ; 20(22)2019 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-31744138

RESUMEN

Ongoing progress in genomic technologies offers exciting tools that can help to resolve transcriptome and genome-wide DNA modifications at single-cell resolution. These methods can be used to characterize individual cells within complex tissue organizations and to highlight various molecular interactions. Here, we will discuss recent advances in the definition of spermatogonial stem cells (SSC) and their progenitors in humans using the single-cell transcriptome sequencing (scRNAseq) approach. Exploration of gene expression patterns allows one to investigate stem cell heterogeneity. It leads to tracing the spermatogenic developmental process and its underlying biology, which is highly influenced by the microenvironment. scRNAseq already represents a new diagnostic tool for the personalized investigation of male infertility. One may hope that a better understanding of SSC biology could facilitate the use of these cells in the context of fertility preservation of prepubertal children, as a key component of regenerative medicine.


Asunto(s)
Medicina Regenerativa , Células Madre/metabolismo , Transcriptoma , Humanos , Infertilidad Masculina/diagnóstico , Infertilidad Masculina/terapia , Masculino , Análisis de la Célula Individual , Espermatogénesis , Espermatogonias/citología , Trasplante de Células Madre , Células Madre/citología
8.
Nat Commun ; 10(1): 3858, 2019 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-31451685

RESUMEN

The Polycomb group of proteins is required for the proper orchestration of gene expression due to its role in maintaining transcriptional silencing. It is composed of several chromatin modifying complexes, including Polycomb Repressive Complex 2 (PRC2), which deposits H3K27me2/3. Here, we report the identification of a cofactor of PRC2, EZHIP (EZH1/2 Inhibitory Protein), expressed predominantly in the gonads. EZHIP limits the enzymatic activity of PRC2 and lessens the interaction between the core complex and its accessory subunits, but does not interfere with PRC2 recruitment to chromatin. Deletion of Ezhip in mice leads to a global increase in H3K27me2/3 deposition both during spermatogenesis and at late stages of oocyte maturation. This does not affect the initial number of follicles but is associated with a reduction of follicles in aging. Our results suggest that mature oocytes Ezhip-/- might not be fully functional and indicate that fertility is strongly impaired in Ezhip-/- females. Altogether, our study uncovers EZHIP as a regulator of chromatin landscape in gametes.


Asunto(s)
Proteínas Oncogénicas/metabolismo , Óvulo/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Espermatozoides/metabolismo , Adulto , Animales , Línea Celular Tumoral , Cromatina/metabolismo , Femenino , Células HEK293 , Histonas/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Mutación , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/aislamiento & purificación , Oogénesis , Ovario/citología , Ovario/patología , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Células Sf9 , Espermatogénesis , Testículo/citología , Testículo/patología
9.
Development ; 146(6)2019 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-30824552

RESUMEN

Neonatal germ cell development provides the foundation of spermatogenesis. However, a systematic understanding of this process is still limited. To resolve cellular and molecular heterogeneity in this process, we profiled single cell transcriptomes of undifferentiated germ cells from neonatal mouse testes and employed unbiased clustering and pseudotime ordering analysis to assign cells to distinct cell states in the developmental continuum. We defined the unique transcriptional programs underlying migratory capacity, resting cellular states and apoptosis regulation in transitional gonocytes. We also identified a subpopulation of primitive spermatogonia marked by CD87 (plasminogen activator, urokinase receptor), which exhibited a higher level of self-renewal gene expression and migration potential. We further revealed a differentiation-primed state within the undifferentiated compartment, in which elevated Oct4 expression correlates with lower expression of self-renewal pathway factors, higher Rarg expression, and enhanced retinoic acid responsiveness. Lastly, a knockdown experiment revealed the role of Oct4 in the regulation of gene expression related to the MAPK pathway and cell adhesion, which may contribute to stem cell differentiation. Our study thus provides novel insights into cellular and molecular regulation during early germ cell development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Análisis de Secuencia de ARN , Espermatogonias/citología , Animales , Animales Recién Nacidos , Apoptosis , Adhesión Celular , Diferenciación Celular , Perfilación de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Microscopía Fluorescente , Factor 3 de Transcripción de Unión a Octámeros/fisiología , Receptores de Ácido Retinoico/fisiología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/fisiología , Espermatogénesis/genética , Transcriptoma , Tretinoina/fisiología , Receptor de Ácido Retinoico gamma
10.
Cell Stem Cell ; 24(1): 1-2, 2019 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-30609395

RESUMEN

Mechanisms regulating maintenance of the stem cell pool in facultative niches of the mammalian testis are poorly understood. In this issue of Cell Stem Cell, Kitadate et al. (2019) propose a minimal model in which stem cells compete for limited resources of fibroblast growth factors, which predicts their density during homeostasis and regenerative conditions.


Asunto(s)
Mitógenos , Células Madre , Animales , Diferenciación Celular , Células Germinativas , Homeostasis , Masculino
11.
Oncotarget ; 8(6): 10050-10063, 2017 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-28052023

RESUMEN

The male germinal lineage, which is defined as unipotent, produces sperm through spermatogenesis. However, embryonic primordial germ cells and postnatal spermatogonial stem cells (SSCs) can change their fate and convert to pluripotency in culture when they are not controlled by the testicular microenvironment. The mechanisms underlying these reprogramming processes are poorly understood. Testicular germ cell tumors, including teratoma, share some molecular characteristics with pluripotent cells, suggesting that cancer could result from an abnormal differentiation of primordial germ cells or from an abnormal conversion of SCCs to pluripotency in the testis. Here, we investigated whether the somatic reprogramming factors Oct3/4, Sox2, Klf4 and c-Myc (OSKM) could play a role in SSCs reprogramming and induce pluripotency using a doxycycline-inducible transgenic Col1a1-4F2A-OSKM mouse model. We showed that, in contrast to somatic cells, SSCs from adult mice are resistant to this reprogramming strategy, even in combination with small molecules, hypoxia, or p53 deficiency, which were previously described to favour the conversion of somatic cells to pluripotency. This finding suggests that adult SSCs have developed specific mechanisms to repress reprogramming by OSKM factors, contributing to circumvent testicular cancer initiation events.


Asunto(s)
Células Madre Germinales Adultas/metabolismo , Técnicas de Reprogramación Celular , Reprogramación Celular , Células Madre Pluripotentes Inducidas/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción SOXB1/metabolismo , Células Madre Germinales Adultas/efectos de los fármacos , Animales , Hipoxia de la Célula , Linaje de la Célula , Células Cultivadas , Reprogramación Celular/efectos de los fármacos , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Proteína-1 Reguladora de Fusión/genética , Proteína-1 Reguladora de Fusión/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genotipo , Células Madre Pluripotentes Inducidas/patología , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre Embrionarias de Ratones/efectos de los fármacos , Factor 3 de Transcripción de Unión a Octámeros/genética , Fenotipo , Proteínas Proto-Oncogénicas c-myc/genética , Factores de Transcripción SOXB1/genética , Transfección , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
13.
Blood ; 124(24): 3613-23, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25261197

RESUMEN

Fanconi anemia (FA) is an inherited chromosomal instability syndrome that is characterized by progressive bone marrow failure. One of the main causes of morbidity and mortality in FA is a bleeding tendency, resulting from low platelet counts. Platelets are the final products of megakaryocyte (MK) maturation. Here, we describe a previously unappreciated role of Fanconi anemia group A protein (Fanca) during the endomitotic process of MK differentiation. Fanca deficiency leads to the accumulation of MKs with low nuclear ploidy and to decreased platelet production. We show, for the first time, that Fanca(-/-) mice are characterized by limited number and proliferative capacity of MK progenitors. Defective megakaryopoiesis of Fanca(-/-) cells is associated with the formation of nucleoplasmic bridges and increased chromosomal instability, indicating that inaccurate endoreplication and karyokinesis occur during MK polyploidization. Sustained DNA damage forces Fanca(-/-) MKs to enter a senescence-like state. Furthermore, inhibition of the Rho-associated kinase, a regulator of cytokinesis, improves the polyploidization of Fanca(-/-) MKs but greatly increases their genomic instability and diminishes their differentiation potential, supporting the notion that accumulation of DNA damage through endomitotic cycles affects MK maturation. Our study indicates that Fanca expression during endomitosis is crucial for normal megakaryopoiesis and platelet production.


Asunto(s)
Proteína del Grupo de Complementación A de la Anemia de Fanconi , Regulación de la Expresión Génica/genética , Megacariocitos/metabolismo , Trombocitopenia , Trombopoyesis/genética , Animales , Senescencia Celular/genética , Inestabilidad Cromosómica/genética , Proteína del Grupo de Complementación A de la Anemia de Fanconi/biosíntesis , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Megacariocitos/patología , Ratones , Ratones Noqueados , Mitosis , Trombocitopenia/genética , Trombocitopenia/metabolismo , Trombocitopenia/patología
14.
Cell Stem Cell ; 11(5): 585-6, 2012 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-23122284

RESUMEN

The feasibility of using spermatogonial stem cells (SSCs) for cell-based infertility treatment has suffered from a lack of evidence in a preclinical nonhuman primate model. In this issue, Hermann et al. (2012) demonstrate that autologous and allogeneic transplantation of SSCs in testes of rhesus macaques produced functional sperm.


Asunto(s)
Espermatogonias/trasplante , Espermatozoides/fisiología , Testículo/trasplante , Animales , Masculino
15.
Hum Mol Genet ; 21(1): 121-35, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21968513

RESUMEN

Fanconi anemia (FA) is a human rare genetic disorder characterized by congenital defects, bone marrow (BM) failure and predisposition to leukemia. The progressive aplastic anemia suggests a defect in the ability of hematopoietic stem cells (HSC) to sustain hematopoieis. We have examined the role of the nuclear FA core complex gene Fancg in the functionality of HSC. In Fancg-/- mice, we observed a decay of long-term HSC and multipotent progenitors that account for the reduction in the LSK compartment containing primitive hematopoietic cells. Fancg-/- lymphoid and myeloid progenitor cells were also affected, and myeloid progenitors show compromised in vitro functionality. HSC from Fancg-/- mice failed to engraft and to reconstitute at short and long term the hematopoiesis in a competitive transplantation assay. Fancg-/- LSK cells showed a loss of quiescence, an impaired migration in vitro in response to the chemokine CXCL12 and a defective homing to the BM after transplantation. Finally, the expression of several key genes involved in self-renewal, quiescence and migration of HSC was dysregulated in Fancg-deficient LSK subset. Collectively, our data reveal that Fancg should play a role in the regulation of physiological functions of HSC.


Asunto(s)
Proteína del Grupo de Complementación G de la Anemia de Fanconi/deficiencia , Anemia de Fanconi/fisiopatología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Animales , Médula Ósea/metabolismo , Movimiento Celular , Quimiocina CXCL12/metabolismo , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación G de la Anemia de Fanconi/genética , Femenino , Hematopoyesis , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
16.
PLoS One ; 5(8): e12134, 2010 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-20711434

RESUMEN

Spermatogonia- stem cells and progenitors of adult spermatogenesis- are killed through a p53-regulated apoptotic process after gamma-irradiation but the death effectors are still poorly characterized. Our data demonstrate that both intrinsic and extrinsic apoptotic pathways are involved, and especially that spermatogonia can be split into two main populations, according to apoptotic effectors. Following irradiation both Dr5 and Puma genes are upregulated in the alpha6-integrin-positive Side Population (SP) fraction, which is highly enriched in spermatogonia. Flow cytometric analysis confirms an increased number of Dr5-expressing SP cells, and Puma-beta isoform accumulates in alpha6-integrin positive cellular extracts, enriched in spermatogonia. Trail-/- or Puma-/- spermatogonia display a reduced sensitivity to radiation-induced apoptosis. The TUNEL kinetics strongly suggest that the extrinsic and intrinsic pathways, via Trail/Dr5 and Puma respectively, could be engaged in distinct subpopulations of spermatogonia. Indeed flow cytometric studies show that Dr5 receptor is constitutively present on more than half of the undifferentiated progenitors (Kit- alpha6+ SP) and half of the differentiated ones (Kit+ alpha6+ SP). In addition after irradiation, Puma is not detected in the Dr5-positive cellular fraction isolated by immunomagnetic purification, while Puma is present in the Dr5-negative cell extracts. In conclusion, adult testicular progenitors are divided into distinct sub-populations by apoptotic effectors, independently of progenitor types (immature Kit-negative versus mature Kit-positive), underscoring differential radiosensitivities characterizing the stem cell/progenitors compartment.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/efectos de la radiación , Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de la radiación , Transducción de Señal/efectos de la radiación , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Testículo/citología , Proteínas Supresoras de Tumor/metabolismo , Células Madre Adultas/metabolismo , Animales , Diferenciación Celular/efectos de la radiación , Rayos gamma , Regulación de la Expresión Génica/efectos de la radiación , Masculino , Ratones , Mitocondrias/metabolismo , Mitocondrias/efectos de la radiación , Espermatogonias/citología , Espermatogonias/metabolismo , Espermatogonias/efectos de la radiación , Proteína p53 Supresora de Tumor/metabolismo
17.
Dev Biol ; 342(1): 74-84, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20346356

RESUMEN

During testis development, proliferation and death of gonocytes are highly regulated to establish a standard population of adult stem spermatogonia that maintain normal spermatogenesis. As Transforming Growth Factor beta (TGFbeta) can regulate proliferation and apoptosis, we investigated its expression and functions during testis development. We show that TGFbeta2 is only expressed in quiescent gonocytes and decreases gonocyte proliferation in vitro. To study the functions of TGFbeta2, we developed conditional mice that invalidate the TGFbeta receptor type II in germ cells. Most of the knock-out animals die during fetal life, but the surviving adults show a reduced pool of spermatogonial stem/progenitor cells and become sterile with time. Using an organ culture system mimicking in vivo development, we show higher proportions of proliferating and apoptotic gonocytes from 13.5 dpc until 1 dpp, suggesting a reduction of germinal quiescence in these animals. Conversely, a 24-hour TGFbeta2-treatment of explanted wild-type testes, isolated every day from 13.5 dpc until 1 dpp, increased the duration of quiescence. These data show that the TGFbeta signaling pathway plays a physiological role during testis development by acting directly as a negative regulator of the fetal and neonatal germ cell proliferation, and indicate that the TGFbeta signaling pathway might regulate the duration of germ cell quiescence and is necessary to maintain adult spermatogenesis.


Asunto(s)
Células Germinativas/metabolismo , Transducción de Señal/genética , Espermatogénesis/genética , Espermatogonias/metabolismo , Factor de Crecimiento Transformador beta/genética , Animales , Apoptosis/genética , Proliferación Celular , Fertilidad/genética , Masculino , Ratones , Ratones Noqueados , Técnicas de Cultivo de Órganos , Proteínas Serina-Treonina Quinasas/fisiología , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Espermatogonias/citología , Células Madre/metabolismo , Testículo/citología , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
18.
Blood ; 115(3): 443-52, 2010 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-19797522

RESUMEN

Few techniques are available to characterize in vivo the early cellular dynamics of long-term reconstitution of hematopoiesis after transplantation of hematopoietic stem cells (HSCs) after lethal irradiation. Using a fiber-optic imaging system, we track the early steps of in vivo recruitment and proliferation of Lin(-)Sca-1(+)c-Kit(+)CD34(-) (LSKCD34(-)) HSCs highly enriched in HSCs and transplanted into lethally irradiated mice. Recruitment of the transplanted LSKCD34(-) hematopoietic cells first occurs in the femoral head and is continuous during 24 hours. Quantification of the fluorescence emitted by the transplanted hematopoietic cells shows that proliferation of LSKCD34(-) hematopoietic cells in the femoral head was potent 3 days after transplantation. Using a development of this fiber-optic imaging system, we show that the transplanted LSKCD34(-) hematopoietic cells are associated with vascularized structures as early as 5 hours after transplantation. This early association is dependent on reactive oxygen species (ROS) partly through the regulation of vascular cell adhesion molecule-1 expression on endothelial cells and is followed by a ROS-dependent proliferation of LSKCD34(-) hematopoietic cells. This new in vivo imaging technique permits the observation of the early steps of hematopoietic reconstitution by HSCs in long bones and shows a new role of ROS in the recruitment of HSCs by bone marrow endothelial cells.


Asunto(s)
Hematopoyesis/efectos de los fármacos , Imagen Molecular/métodos , Especies Reactivas de Oxígeno/farmacología , Animales , Antígenos CD34/metabolismo , Antígenos Ly/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Fémur/citología , Fémur/metabolismo , Fémur/fisiología , Tecnología de Fibra Óptica/métodos , Hematopoyesis/fisiología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/fisiología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Modelos Biológicos , Proteínas Proto-Oncogénicas c-kit/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Estudios de Validación como Asunto , Irradiación Corporal Total
20.
Biol Reprod ; 80(5): 860-73, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19144961

RESUMEN

Neonatal gonocytes are the precursors of both spermatogonial stem cells and spermatogonia; thus, any persistent DNA damage in these cells may lead to heritable mutations. We investigated the response of male mouse neonatal germ cells to ionizing radiation. Both gonocytes and spermatogonia died in large numbers by apoptosis. However, we found that the gonocytes were significantly more sensitive than spermatogonia and somatic cells to radiation-induced double-strand breaks (DSBs), as assayed by the number of gamma-H2AFX foci. In contrast, gonocytes irradiated in G2 phase seemed to repair DSBs faster than spermatogonia. Moreover, when irradiated in S phase, gonocytes arrested their cell cycle at the G1/S phase transition, whereas spermatogonia were mostly blocked in G2/M phase. Despite these differences, both cell types expressed high levels of proteins involved in DSB signaling and repair. Within the first hours after irradiation, the expression of Atr, Mre11a, H2afx, Xrcc6, and Xrcc4 was downregulated in neonatal spermatogonia, whereas, in gonocytes, most gene expression was unaffected. Together, these results suggest that the response of neonatal testis to genotoxic stress is regulated by different mechanisms according to the cell type and the differentiation status.


Asunto(s)
Espermatogonias/efectos de la radiación , Células Madre/efectos de la radiación , Animales , Animales Recién Nacidos , Secuencia de Bases , Ciclo Celular/efectos de la radiación , Diferenciación Celular , Roturas del ADN de Doble Cadena , Cartilla de ADN/genética , Reparación del ADN/genética , Expresión Génica/efectos de la radiación , Histonas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Mutación , Tolerancia a Radiación , Espermatogonias/citología , Espermatogonias/metabolismo , Células Madre/citología , Células Madre/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...