Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 14(4): e1007260, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29621239

RESUMEN

Mechanisms generating diverse cell types from multipotent progenitors are fundamental for normal development. Pigment cells are derived from multipotent neural crest cells and their diversity in teleosts provides an excellent model for studying mechanisms controlling fate specification of distinct cell types. Zebrafish have three types of pigment cells (melanocytes, iridophores and xanthophores) while medaka have four (three shared with zebrafish, plus leucophores), raising questions about how conserved mechanisms of fate specification of each pigment cell type are in these fish. We have previously shown that the Sry-related transcription factor Sox10 is crucial for fate specification of pigment cells in zebrafish, and that Sox5 promotes xanthophores and represses leucophores in a shared xanthophore/leucophore progenitor in medaka. Employing TILLING, TALEN and CRISPR/Cas9 technologies, we generated medaka and zebrafish sox5 and sox10 mutants and conducted comparative analyses of their compound mutant phenotypes. We show that specification of all pigment cells, except leucophores, is dependent on Sox10. Loss of Sox5 in Sox10-defective fish partially rescued the formation of all pigment cells in zebrafish, and melanocytes and iridophores in medaka, suggesting that Sox5 represses Sox10-dependent formation of these pigment cells, similar to their interaction in mammalian melanocyte specification. In contrast, in medaka, loss of Sox10 acts cooperatively with Sox5, enhancing both xanthophore reduction and leucophore increase in sox5 mutants. Misexpression of Sox5 in the xanthophore/leucophore progenitors increased xanthophores and reduced leucophores in medaka. Thus, the mode of Sox5 function in xanthophore specification differs between medaka (promoting) and zebrafish (repressing), which is also the case in adult fish. Our findings reveal surprising diversity in even the mode of the interactions between Sox5 and Sox10 governing specification of pigment cell types in medaka and zebrafish, and suggest that this is related to the evolution of a fourth pigment cell type.


Asunto(s)
Linaje de la Célula , Melanocitos/metabolismo , Oryzias/genética , Pigmentación/genética , Factores de Transcripción SOXD/genética , Factores de Transcripción SOXE/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Alelos , Animales , Regulación del Desarrollo de la Expresión Génica , Melanocitos/citología , Cresta Neural/metabolismo , Factores de Transcripción SOXD/metabolismo , Factores de Transcripción SOXE/metabolismo , Proteínas de Pez Cebra/metabolismo
2.
Biosci Biotechnol Biochem ; 82(7): 1165-1168, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29570037

RESUMEN

Transglutaminase (TG) is an essential enzyme to catalyze cross-linking reactions of epidermal proteins. Recently, we biochemically characterized human skin TG orthologues for medaka (Oryzias latipes), a model fish. By genome editing, gene-modified fishes for the two orthologues were obtained, both of which lack the ordinal enzymes. These fish appeared to exhibit higher susceptibility to osmolality at the period of larvae.


Asunto(s)
Proteínas de Peces/genética , Mutación , Oryzias/fisiología , Concentración Osmolar , Piel/enzimología , Transglutaminasas/genética , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Femenino , Edición Génica , Humanos , Larva/fisiología , Masculino , Oryzias/genética , Oryzias/crecimiento & desarrollo , Transglutaminasas/química
3.
PLoS Genet ; 11(2): e1005009, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25719383

RESUMEN

To increase individual male fitness, males of various species remain near a (potential) mating partner and repel their rivals (mate-guarding). Mate-guarding is assumed to be mediated by two different types of motivation: sexual motivation toward the opposite sex and competitive motivation toward the same sex. The genetic/molecular mechanisms underlying how mate presence affects male competitive motivation in a triadic relationship has remained largely unknown. Here we showed that male medaka fish prominently exhibit mate-guarding behavior. The presence of a female robustly triggers male-male competition for the female in a triadic relationship (2 males and 1 female). The male-male competition resulted in one male occupying a dominant position near the female while interfering with the other male's approach of the female. Paternity testing revealed that the dominant male had a significantly higher mating success rate than the other male in a triadic relationship. We next generated medaka mutants of arginine-vasotocin (avt) and its receptors (V1a1, V1a2) and revealed that two genes, avt and V1a2, are required for normal mate-guarding behavior. In addition, behavioral analysis of courtship behaviors in a dyadic relationship and aggressive behaviors within a male group revealed that avt mutant males displayed decreased sexual motivation but showed normal aggression. In contrast, heterozygote V1a2 mutant males displayed decreased aggression, but normal mate-guarding and courtship behavior. Thus, impaired mate-guarding in avt and V1a2 homozygote mutants may be due to the loss of sexual motivation toward the opposite sex, and not to the loss of competitive motivation toward rival males. The different behavioral phenotypes between avt, V1a2 heterozygote, and V1a2 homozygote mutants suggest that there are redundant systems to activate V1a2 and that endogenous ligands activating the receptor may differ according to the social context.


Asunto(s)
Oryzias/genética , Reproducción/fisiología , Conducta Sexual Animal/fisiología , Vasotocina/genética , Agresión/fisiología , Animales , Copulación/fisiología , Femenino , Masculino , Matrimonio , Motivación/fisiología , Oryzias/fisiología , Vasotocina/metabolismo
4.
PLoS Genet ; 10(4): e1004246, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24699463

RESUMEN

Mechanisms generating diverse cell types from multipotent progenitors are crucial for normal development. Neural crest cells (NCCs) are multipotent stem cells that give rise to numerous cell-types, including pigment cells. Medaka has four types of NCC-derived pigment cells (xanthophores, leucophores, melanophores and iridophores), making medaka pigment cell development an excellent model for studying the mechanisms controlling specification of distinct cell types from a multipotent progenitor. Medaka many leucophores-3 (ml-3) mutant embryos exhibit a unique phenotype characterized by excessive formation of leucophores and absence of xanthophores. We show that ml-3 encodes sox5, which is expressed in premigratory NCCs and differentiating xanthophores. Cell transplantation studies reveal a cell-autonomous role of sox5 in the xanthophore lineage. pax7a is expressed in NCCs and required for both xanthophore and leucophore lineages; we demonstrate that Sox5 functions downstream of Pax7a. We propose a model in which multipotent NCCs first give rise to pax7a-positive partially fate-restricted intermediate progenitors for xanthophores and leucophores; some of these progenitors then express sox5, and as a result of Sox5 action develop into xanthophores. Our results provide the first demonstration that Sox5 can function as a molecular switch driving specification of a specific cell-fate (xanthophore) from a partially-restricted, but still multipotent, progenitor (the shared xanthophore-leucophore progenitor).


Asunto(s)
Cresta Neural/crecimiento & desarrollo , Oryzias/crecimiento & desarrollo , Pigmentación/genética , Factores de Transcripción SOXD/genética , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proteínas de Peces/genética , Regulación del Desarrollo de la Expresión Génica/genética , Melanóforos/fisiología , Cresta Neural/fisiología , Oryzias/fisiología , Factor de Transcripción PAX7/genética , Fenotipo , Pigmentación/fisiología , Células Madre/fisiología
5.
Hum Mol Genet ; 22(12): 2423-34, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23449626

RESUMEN

Parkinson's disease (PD) is a common neurodegenerative disorder characterized by selective dopaminergic cell loss in the substantia nigra, but its pathogenesis remains unclear. The recessively inherited familial PD genes PARK2 and PARK6 have been attributed to mutations in the Parkin and PTEN-induced kinase 1 (PINK1) genes, respectively. Recent reports suggest that PINK1 works upstream of Parkin in the same pathway to regulate mitochondrial dynamics and/or conduct autophagic clearance of damaged mitochondria. This phenomenon is preserved from Drosophila to human cell lines but has not been demonstrated in a vertebrate animal model in vivo. Here, we developed a medaka fish (Oryzias latipes) model that is deficient in Pink1 and Parkin. We found that despite the lack of a conspicuous phenotype in single mutants for Pink1 or Parkin, medaka that are deficient in both genes developed phenotypes similar to that of human PD: late-onset locomotor dysfunction, a decrease in dopamine levels and a selective degeneration of dopaminergic neurons. Further analysis also revealed defects in mitochondrial enzymatic activity as well as cell death. Consistently, PINK1 and Parkin double-deficient MEF showed a further decrease in mitochondrial membrane potential and mitochondrial complex I activity as well as apoptosis compared with single-deficient MEF. Interestingly, these mitochondrial abnormalities in Parkin-deficient MEF were compensated by exogenous PINK1, but not by disease-related mutants. These results suggest that PINK1 and Parkin work in a complementary way to protect dopaminergic neurons by maintaining mitochondrial function in vertebrates.


Asunto(s)
Dopamina/metabolismo , Proteínas de Peces/metabolismo , Neuronas/metabolismo , Oryzias/metabolismo , Enfermedad de Parkinson/metabolismo , Proteínas Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Apoptosis , Modelos Animales de Enfermedad , Drosophila , Proteínas de Peces/genética , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Neuronas/citología , Oryzias/genética , Enfermedad de Parkinson/genética , Fenotipo , Proteínas Quinasas/genética , Ubiquitina-Proteína Ligasas/genética , Vertebrados/genética , Vertebrados/metabolismo
6.
Development ; 139(13): 2283-7, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22627284

RESUMEN

The function of AMH (Anti-Müllerian hormone), a phylogenetically ancient member of the TGFß family of proteins, in lower vertebrates is largely unknown. Previously, we have shown that the gene encoding the type II anti-Müllerian hormone receptor, amhrII, is responsible for excessive germ cell proliferation and male-to-female sex reversal in the medaka hotei mutant. In this study, functional analyses in cultured cells and of other amhrII mutant alleles indicate that lack of AMH signaling causes the hotei phenotype. BrdU incorporation experiments identified the existence of both quiescent and mitotically active germ cells among the self-renewing, type I population of germ cells in the developing gonad. AMH signaling acts in supporting cells to promote the proliferation of mitotically active germ cells but does not trigger quiescent germ cells to proliferate in the developing gonad. Furthermore, we show that the male-to-female sex reversal phenotype in hotei mutants is not a direct consequence of AMH signaling in supporting cells, but is instead mediated by germ cells. Our data demonstrate that interfollicular AMH signaling regulates proliferation at a specific stage of germ cell development, and that this regulation is crucial for the proper manifestation of gonadal sex directed by sex determination genes.


Asunto(s)
Hormona Antimülleriana/fisiología , Proliferación Celular , Células Germinativas/citología , Oryzias/crecimiento & desarrollo , Receptores de Péptidos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Diferenciación Sexual , Animales , Células Cultivadas , Femenino , Células Germinativas/fisiología , Masculino , Mitosis , Mutación , Oryzias/metabolismo , Receptores de Péptidos/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Transducción de Señal
7.
Gen Comp Endocrinol ; 195: 9-20, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24505600

RESUMEN

The first studies that identified leptin and its receptor (LepR) in mammals were based on mutant animals that displayed dramatic changes in body-weight and regulation of energy homeostasis. Subsequent studies have shown that a deficiency of leptin or LepR in homoeothermic mammals results in hyperphagia, obesity, infertility and a number of other abnormalities. The physiological roles of leptin-mediated signaling in ectothermic teleosts are still being explored. Here, we produced medaka with homozygous LepR gene mutation using the targeting induced local lesions in a genome method. This knockout mutant had a point mutation of cysteine for stop codon at the 357th amino acid just before the leptin-binding domain. The evidence for loss of function of leptin-mediated signaling in the mutant is based on a lack of response to feeding in the expression of key appetite-related neuropeptides in the diencephalon. The mutant lepr−/− medaka expressed constant up-regulated levels of mRNA for the orexigenic neuropeptide Ya and agouti-related protein and a suppressed level of anorexigenic proopiomelanocortin 1 in the diencephalon independent of feeding, which suggests that the mutant did not possess functional LepR. Phenotypes of the LepR-mutant medaka were analyzed in order to understand the effects on food intake, growth, and fat accumulation in the tissues. The food intake of the mutant medaka was higher in post-juveniles and adult stages than that of wild-type (WT) fish. The hyperphagia led to a high growth rate at the post-juvenile stage, but did not to significant alterations in final adult body size. There was no additional deposition of fat in the liver and muscle in the post-juvenile and adult mutants, or in the blood plasma in the adult mutant. However, adult LepR mutants possessed large deposits of visceral fat, unlike in the WT fish, in which there were none. Our analysis confirms that LepR in medaka exert a powerful influence on the control on food intake. Further analyses using the mutant will contribute to a better understanding of the role of leptin in fish. This is the first study to produce fish with leptin receptor deficiency.


Asunto(s)
Animales Modificados Genéticamente/crecimiento & desarrollo , Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/fisiología , Técnicas de Inactivación de Genes , Grasa Intraabdominal/efectos de los fármacos , Neuropéptidos/farmacología , Receptores de Leptina/fisiología , Proteína Relacionada con Agouti/metabolismo , Animales , Animales Modificados Genéticamente/metabolismo , Apetito/efectos de los fármacos , Apetito/fisiología , Diencéfalo/efectos de los fármacos , Diencéfalo/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Hiperfagia/genética , Hiperfagia/patología , Leptina/metabolismo , Mutación/genética , Obesidad/metabolismo , Oryzias/genética , Oryzias/crecimiento & desarrollo , Oryzias/metabolismo , Regulación hacia Arriba
8.
PLoS Genet ; 6(1): e1000828, 2010 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-20107609

RESUMEN

Topoisomerase inhibitors such as camptothecin and etoposide are used as anti-cancer drugs and induce double-strand breaks (DSBs) in genomic DNA in cycling cells. These DSBs are often covalently bound with polypeptides at the 3' and 5' ends. Such modifications must be eliminated before DSB repair can take place, but it remains elusive which nucleases are involved in this process. Previous studies show that CtIP plays a critical role in the generation of 3' single-strand overhang at "clean" DSBs, thus initiating homologous recombination (HR)-dependent DSB repair. To analyze the function of CtIP in detail, we conditionally disrupted the CtIP gene in the chicken DT40 cell line. We found that CtIP is essential for cellular proliferation as well as for the formation of 3' single-strand overhang, similar to what is observed in DT40 cells deficient in the Mre11/Rad50/Nbs1 complex. We also generated DT40 cell line harboring CtIP with an alanine substitution at residue Ser332, which is required for interaction with BRCA1. Although the resulting CtIP(S332A/-/-) cells exhibited accumulation of RPA and Rad51 upon DNA damage, and were proficient in HR, they showed a marked hypersensitivity to camptothecin and etoposide in comparison with CtIP(+/-/-) cells. Finally, CtIP(S332A/-/-)BRCA1(-/-) and CtIP(+/-/-)BRCA1(-/-) showed similar sensitivities to these reagents. Taken together, our data indicate that, in addition to its function in HR, CtIP plays a role in cellular tolerance to topoisomerase inhibitors. We propose that the BRCA1-CtIP complex plays a role in the nuclease-mediated elimination of oligonucleotides covalently bound to polypeptides from DSBs, thereby facilitating subsequent DSB repair.


Asunto(s)
Proteína BRCA1/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN , Proteínas Nucleares/metabolismo , Animales , Proteína BRCA1/genética , Ciclo Celular , Línea Celular Tumoral , Pollos , ADN/genética , Proteínas Nucleares/genética , Unión Proteica , Recombinación Genética
9.
PLoS Genet ; 6(10): e1001170, 2010 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-20975950

RESUMEN

Zn²(+)-finger proteins comprise one of the largest protein superfamilies with diverse biological functions. The ATM substrate Chk2-interacting Zn²(+)-finger protein (ASCIZ; also known as ATMIN and ZNF822) was originally linked to functions in the DNA base damage response and has also been proposed to be an essential cofactor of the ATM kinase. Here we show that absence of ASCIZ leads to p53-independent late-embryonic lethality in mice. Asciz-deficient primary fibroblasts exhibit increased sensitivity to DNA base damaging agents MMS and H2O2, but Asciz deletion knock-down does not affect ATM levels and activation in mouse, chicken, or human cells. Unexpectedly, Asciz-deficient embryos also exhibit severe respiratory tract defects with complete pulmonary agenesis and severe tracheal atresia. Nkx2.1-expressing respiratory precursors are still specified in the absence of ASCIZ, but fail to segregate properly within the ventral foregut, and as a consequence lung buds never form and separation of the trachea from the oesophagus stalls early. Comparison of phenotypes suggests that ASCIZ functions between Wnt2-2b/ß-catenin and FGF10/FGF-receptor 2b signaling pathways in the mesodermal/endodermal crosstalk regulating early respiratory development. We also find that ASCIZ can activate expression of reporter genes via its SQ/TQ-cluster domain in vitro, suggesting that it may exert its developmental functions as a transcription factor. Altogether, the data indicate that, in addition to its role in the DNA base damage response, ASCIZ has separate developmental functions as an essential regulator of respiratory organogenesis.


Asunto(s)
Proteínas Portadoras/fisiología , Reparación del ADN/fisiología , Pulmón/embriología , Proteínas Nucleares/fisiología , Organogénesis/fisiología , Animales , Western Blotting , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Senescencia Celular , Daño del ADN , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Genotipo , Humanos , Peróxido de Hidrógeno/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oxidantes/farmacología , Factores de Tiempo , Tráquea/embriología , Factores de Transcripción , Rayos Ultravioleta
10.
Dev Biol ; 359(1): 82-94, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21925159

RESUMEN

Myostatin (MSTN) functions as a negative regulator of skeletal muscle mass. In mammals, MSTN-deficient animals result in an increase of skeletal muscle mass with both hyperplasia and hypertrophy. A MSTN gene is highly conserved within the fish species, allowing speculation that MSTN-deficient fish could exhibit a double-muscled phenotype. Some strategies for blocking or knocking down MSTN in adult fish have been already performed; however, these fish show either only hyperplastic or hypertrophic growth in muscle fiber. Therefore, the role of MSTN in fish myogenesis during post-hatch growth remains unclear. To address this question, we have made MSTN-deficient medaka (mstnC315Y) by using the targeting induced local lesions in a genome method. mstnC315Y can reproduce and have the same survival period as WT medaka. Growth rates of WT and mstnC315Y were measured at juvenile (1-2wk post-hatching), post-juvenile (3-7wk post-hatching) and adult (8-16wk post-hatching) stages. In addition, effects of MSTN on skeletal muscle differentiation were investigated at histological and molecular levels at each developmental stage. As a result, mstnC315Y show a significant increase in body weight from the post-juvenile to adult stage. Hyper-morphogenesis of skeletal muscle in mstnC315Y was accomplished due to hyperplastic growth from post-juvenile to early adult stage, followed by hypertrophic growth in the adult stage. Myf-5 and MyoD were up-regulated in mstnC315Y at the hyperplastic growth phase, while myogenin was highly expressed in mstnC315Y at the hypertrophic growth phase. These indicated that MSTN in medaka plays a dual role for muscle fiber development. In conclusion, MSTN in medaka regulates the number and size of muscle fiber in a temporally-controlled manner during posthatch growth.


Asunto(s)
Músculo Esquelético/crecimiento & desarrollo , Miostatina/genética , Oryzias/crecimiento & desarrollo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Western Blotting , Clonación Molecular , Cartilla de ADN , Electroforesis en Gel de Poliacrilamida , Hiperplasia , Hipertrofia , Inmunohistoquímica , Datos de Secuencia Molecular , Músculo Esquelético/metabolismo , Oryzias/genética , Fenotipo , Reacción en Cadena de la Polimerasa , Homología de Secuencia de Aminoácido
11.
Biochem Biophys Res Commun ; 423(4): 627-31, 2012 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-22659737

RESUMEN

Tumor suppressor p53 negatively regulates self-renewal of neural stem cells in the adult murine brain. Here, we report that the p53 null mutation in medaka fish (Oryzias latipes) suppressed neurogenesis in the telencephalon, independent of cell death. By using 5-bromo-29-deoxyuridine (BrdU) immunohistochemistry, we identified 18 proliferation zones in the brains of young medaka fish; in situ hybridization showed that p53 was expressed selectively in at least 12 proliferation zones. We also compared the number of BrdU-positive cells present in the whole telencephalon of wild-type (WT) and p53 mutant fish. Immediately after BrdU exposure, the number of BrdU-positive cells did not differ significantly between them. One week after BrdU-exposure, the BrdU-positive cells migrated from the proliferation zone, which was accompanied by an increased number in the WT brain. In contrast, no significant increase was observed in the p53 mutant brain. Terminal deoxynucleotidyl transferase (dUTP) nick end-labeling revealed that there was no significant difference in the number of apoptotic cells in the telencephalon of p53 mutant and WT medaka, suggesting that the decreased number of BrdU-positive cells in the mutant may be due to the suppression of proliferation rather than the enhancement of neural cell death. These results suggest that p53 positively regulates neurogenesis via cell proliferation.


Asunto(s)
Proliferación Celular , Neurogénesis/genética , Oryzias/crecimiento & desarrollo , Telencéfalo/crecimiento & desarrollo , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis , Bromodesoxiuridina/química , Mutación , Oryzias/genética , Supresión Genética , Telencéfalo/citología
12.
Dev Growth Differ ; 54(5): 588-604, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22694322

RESUMEN

Roberts syndrome and SC phocomelia (RBS/SC) are genetic autosomal recessive syndromes caused by establishment of cohesion 1 homolog 2 ( ESCO 2) mutation. RBS/SC appear to have a variety of clinical features, even with the same mutation of the ESCO2 gene. Here, we established and genetically characterized a medaka model of RBS/SC by reverse genetics. The RBS/SC model was screened from a mutant medaka library produced by the Targeting Induced Local Lesions in Genomes method. The medaka mutant carrying the homozygous mutation at R80S in the conserved region of ESCO2 exhibited clinical variety (i.e. developmental arrest with craniofacial and chromosomal abnormalities and embryonic lethality) as characterized in RBS/SC. Moreover, widespread apoptosis and downregulation of some gene expression, including notch1a, were detected in the R80S mutant. The R80S mutant is the animal model for RBS/SC and a valuable resource that provides the opportunity to extend knowledge of ESCO2. Downregulation of some gene expression in the R80S mutant is an important clue explaining non-correlation between genotype and phenotype in RBS/SC.


Asunto(s)
Acetiltransferasas/genética , Anomalías Craneofaciales/genética , Modelos Animales de Enfermedad , Ectromelia/genética , Hipertelorismo/genética , Oryzias , Acetiltransferasas/metabolismo , Animales , Apoptosis/genética , Clonación Molecular , Anomalías Craneofaciales/metabolismo , Ectromelia/metabolismo , Genotipo , Hipertelorismo/metabolismo , Oryzias/genética , Oryzias/metabolismo , Fenotipo , Polimorfismo de Nucleótido Simple , Receptor Notch1/biosíntesis , Genética Inversa
13.
PLoS Genet ; 5(1): e1000356, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19180185

RESUMEN

Homologous recombination (HR) is initiated by DNA double-strand breaks (DSB). However, it remains unclear whether single-strand lesions also initiate HR in genomic DNA. Chicken B lymphocytes diversify their Immunoglobulin (Ig) V genes through HR (Ig gene conversion) and non-templated hypermutation. Both types of Ig V diversification are initiated by AID-dependent abasic-site formation. Abasic sites stall replication, resulting in the formation of single-stranded gaps. These gaps can be filled by error-prone DNA polymerases, resulting in hypermutation. However, it is unclear whether these single-strand gaps can also initiate Ig gene conversion without being first converted to DSBs. The Mre11-Rad50-Nbs1 (MRN) complex, which produces 3' single-strand overhangs, promotes the initiation of DSB-induced HR in yeast. We show that a DT40 line expressing only a truncated form of Nbs1 (Nbs1(p70)) exhibits defective HR-dependent DSB repair, and a significant reduction in the rate--though not the fidelity--of Ig gene conversion. Interestingly, this defective gene conversion was restored to wild type levels by overproduction of Escherichia coli SbcB, a 3' to 5' single-strand-specific exonuclease, without affecting DSB repair. Conversely, overexpression of chicken Exo1 increased the efficiency of DSB-induced gene-targeting more than 10-fold, with no effect on Ig gene conversion. These results suggest that Ig gene conversion may be initiated by single-strand gaps rather than by DSBs, and, like SbcB, the MRN complex in DT40 may convert AID-induced lesions into single-strand gaps suitable for triggering HR. In summary, Ig gene conversion and hypermutation may share a common substrate-single-stranded gaps. Genetic analysis of the two types of Ig V diversification in DT40 provides a unique opportunity to gain insight into the molecular mechanisms underlying the filling of gaps that arise as a consequence of replication blocks at abasic sites, by HR and error-prone polymerases.


Asunto(s)
Linfocitos B/metabolismo , Roturas del ADN de Cadena Simple , Región Variable de Inmunoglobulina/genética , Proteínas Nucleares/metabolismo , Recombinación Genética , Animales , Línea Celular Tumoral , Pollos , Reparación del ADN , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Conversión Génica , Región Variable de Inmunoglobulina/metabolismo , Proteínas Nucleares/genética
14.
Cell Struct Funct ; 36(2): 247-59, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22067999

RESUMEN

The accumulation of unfolded proteins in the endoplasmic reticulum (ER) activates the unfolded protein response (UPR). The ER stress signal is sensed and transmitted by a transmembrane protein(s) in the ER. The number of these transducers has increased with evolution, one in yeast, three in worm and fly, and five in mammals. Here, we examined medaka fish, Oryzias latipes, as a vertebrate model organism, and found that the medaka genome encodes five UPR transducers. Analysis of a medaka embryonic cell line revealed that the mammalian UPR signaling mechanisms are very well conserved. Thus, XBP1 mRNA, which encodes the transcription factor XBP1 downstream of the IRE1 pathway, was spliced in response to ER stress, resulting in production of the active form of XBP1. Translation was generally attenuated in response to ER stress, which paradoxically induced the translation of ATF4, the transcription factor downstream of the PERK pathway. ATF6 was constitutively synthesized as a transmembrane protein and activated by ER stress-induced proteolysis. Results obtained with the overexpression of active ATF6α, ATF6ß, and XBP1 strongly suggested that ATF6α plays a major role in upregulating the major ER chaperone BiP, contrary to the case in non-vertebrates, in which the IRE1 pathway is essential to the induction of BiP. Physiological ER stress occurring during embryonic development was visualized using transgenic medaka carrying the enhanced green fluorescent protein gene under the control of the BiP promoter. Thus, analysis of the vertebrate UPR using medaka will help provide a more comprehensive understanding of the biology and physiology of the UPR.


Asunto(s)
Oryzias/metabolismo , Transducción de Señal , Respuesta de Proteína Desplegada/fisiología , Animales , Retículo Endoplásmico/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Modelos Animales , Desplegamiento Proteico , ARN Mensajero/genética , ARN Mensajero/metabolismo
15.
J Biol Chem ; 284(39): 26360-7, 2009 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-19661064

RESUMEN

Bloom syndrome caused by inactivation of the Bloom DNA helicase (Blm) is characterized by increases in the level of sister chromatid exchange, homologous recombination (HR) associated with cross-over. It is therefore believed that Blm works as an anti-recombinase. Meanwhile, in Drosophila, DmBlm is required specifically to promote the synthesis-dependent strand anneal (SDSA), a type of HR not associating with cross-over. However, conservation of Blm function in SDSA through higher eukaryotes has been a matter of debate. Here, we demonstrate the function of Blm in SDSA type HR in chicken DT40 B lymphocyte line, where Ig gene conversion diversifies the immunoglobulin V gene through intragenic HR between diverged homologous segments. This reaction is initiated by the activation-induced cytidine deaminase enzyme-mediated uracil formation at the V gene, which in turn converts into abasic site, presumably leading to a single strand gap. Ig gene conversion frequency was drastically reduced in BLM(-/-) cells. In addition, BLM(-/-) cells used limited donor segments harboring higher identity compared with other segments in Ig gene conversion event, suggesting that Blm can promote HR between diverged sequences. To further understand the role of Blm in HR between diverged homologous sequences, we measured the frequency of gene targeting induced by an I-SceI-endonuclease-mediated double-strand break. BLM(-/-) cells showed a severer defect in the gene targeting frequency as the number of heterologous sequences increased at the double-strand break site. Conversely, the overexpression of Blm, even an ATPase-defective mutant, strongly stimulated gene targeting. In summary, Blm promotes HR between diverged sequences through a novel ATPase-independent mechanism.


Asunto(s)
Mutación , RecQ Helicasas/metabolismo , Recombinación Genética/genética , Adenosina Trifosfatasas/metabolismo , Animales , Secuencia de Bases , Ciclo Celular , Línea Celular , Pollos , ADN/genética , ADN/metabolismo , Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Rayos gamma , Humanos , Inmunoglobulinas/genética , RecQ Helicasas/genética , Intercambio de Cromátides Hermanas/genética
16.
J Neurochem ; 115(5): 1150-60, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21219329

RESUMEN

Perturbations in protein folding and degradation are key pathological mechanisms in neurodegenerative diseases, including Parkinson's disease (PD). Recent evidence suggests that mishandling of proteins may play an important role in the pathogenesis of PD. We have utilized medaka fish to monitor the effects of injecting neurotoxins into the CSF space. In this study, ammonium chloride, tunicamycin, and lactacystin were tested for their ability to disturb lysosomal proteolysis, N-glycosylation in the endoplasmic reticulum, and proteasomal degradation, respectively. All of the substances tested induced selective loss of dopaminergic neurons, movement disorders and inclusion bodies. Among them, the features of the inclusion bodies that developed after ammonium chloride injection mimicked those of PD: co-localization of ubiquitin and phosphorylated α-synuclein, as well as the presence of LC3 protein in the inclusion bodies. Our study demonstrated that medaka fish are useful for examining the effects of environmental toxins and lysosome inhibition, and lysosome inhibitors may be factors in the development of PD.


Asunto(s)
Dopamina/fisiología , Neuronas/efectos de los fármacos , Neurotoxinas/toxicidad , Oryzias/fisiología , Trastornos Parkinsonianos/inducido químicamente , Fenotipo , Sustancia Negra/efectos de los fármacos , Acetilcisteína/análogos & derivados , Acetilcisteína/toxicidad , Cloruro de Amonio/toxicidad , Animales , Modelos Animales de Enfermedad , Proteínas del Tejido Nervioso/metabolismo , Neuronas/patología , Oryzias/anatomía & histología , Trastornos Parkinsonianos/patología , Trastornos Parkinsonianos/fisiopatología , Sustancia Negra/patología , Sustancia Negra/fisiopatología , Tunicamicina/toxicidad
17.
J Neurochem ; 115(1): 178-87, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20649841

RESUMEN

Recent findings suggest that a defect in the ubiquitin-proteasome system plays an important role in the pathogenesis of Parkinson's disease (PD). A previous report (McNaught et al. 2004) demonstrated that rats systemically injected with proteasome inhibitors exhibited PD-like clinical symptoms and pathology. However, because these findings have not been consistently replicated, this model is not commonly used to study PD. We used medaka fish to test the effect of systemic administration of proteasome inhibitors because of the high level of accessibility of the cerebrospinal fluid in fish. We injected lactacystin or epoxomicin into the CSF of medaka. With proteasome inhibition in the medaka brain, selective dopaminergic and noradrenergic cell loss was observed. Furthermore, treated fish exhibited reduced spontaneous movement. Treatment with proteasome inhibitors also induced the formation of inclusion bodies resembling Lewy bodies, which are characteristic of PD. Treatment with 6-OHDA also induced dopaminergic cell loss but did not produce inclusion bodies. These findings in medaka are consistent with previous results reporting that non-selective proteasome inhibition replicates the cardinal features of PD: locomotor dysfunction, selective dopaminergic cell loss, and inclusion body formation.


Asunto(s)
Química Encefálica/efectos de los fármacos , Química Encefálica/fisiología , Oryzias/fisiología , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/patología , Inhibidores de Proteasoma , Acetilcisteína/análogos & derivados , Acetilcisteína/líquido cefalorraquídeo , Acetilcisteína/farmacología , Animales , Conducta Animal/efectos de los fármacos , Western Blotting , Inhibidores de Cisteína Proteinasa/líquido cefalorraquídeo , Inhibidores de Cisteína Proteinasa/farmacología , Dopamina/fisiología , Discinesia Inducida por Medicamentos/patología , Discinesia Inducida por Medicamentos/fisiopatología , Inmunohistoquímica , Cuerpos de Inclusión/patología , Microscopía Electrónica de Transmisión , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Norepinefrina/fisiología , Oxidopamina/administración & dosificación , Oxidopamina/líquido cefalorraquídeo , Oxidopamina/farmacología , Enfermedad de Parkinson Secundaria/psicología , Natación/fisiología , Simpaticolíticos/administración & dosificación , Simpaticolíticos/líquido cefalorraquídeo , Simpaticolíticos/farmacología , Ubiquitinación/efectos de los fármacos
18.
Mol Cell Biol ; 27(7): 2562-71, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17242200

RESUMEN

The Saccharomyces cerevisiae RAD18 gene is essential for postreplication repair but is not required for homologous recombination (HR), which is the major double-strand break (DSB) repair pathway in yeast. Accordingly, yeast rad18 mutants are tolerant of camptothecin (CPT), a topoisomerase I inhibitor, which induces DSBs by blocking replication. Surprisingly, mammalian cells and chicken DT40 cells deficient in Rad18 display reduced HR-dependent repair and are hypersensitive to CPT. Deletion of nonhomologous end joining (NHEJ), a major DSB repair pathway in vertebrates, in rad18-deficient DT40 cells completely restored HR-mediated DSB repair, suggesting that vertebrate Rad18 regulates the balance between NHEJ and HR. We previously reported that loss of NHEJ normalized the CPT sensitivity of cells deficient in poly(ADP-ribose) polymerase 1 (PARP1). Concomitant deletion of Rad18 and PARP1 synergistically increased CPT sensitivity, and additional inactivation of NHEJ normalized this hypersensitivity, indicating their parallel actions. In conclusion, higher-eukaryotic cells separately employ PARP1 and Rad18 to suppress the toxic effects of NHEJ during the HR reaction at stalled replication forks.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Proteínas de Unión al ADN/fisiología , Poli(ADP-Ribosa) Polimerasas/fisiología , Recombinación Genética , Animales , Antígenos Nucleares/fisiología , Camptotecina/farmacología , Línea Celular , Pollos , Proteínas de Unión al ADN/genética , Humanos , Autoantígeno Ku , Poli(ADP-Ribosa) Polimerasas/genética , Inhibidores de Topoisomerasa I , Ubiquitina-Proteína Ligasas
19.
Biochem Biophys Res Commun ; 371(2): 225-9, 2008 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-18433721

RESUMEN

ASCIZ (ATMIN) was recently identified as a novel DNA damage response protein. Here we report that ASCIZ-deficient chicken DT40 B lymphocyte lines displayed markedly increased Ig gene conversion rates, whereas overexpression of human ASCIZ reduced Ig gene conversion below wild-type levels. However, neither the efficiency of double-strand break repair nor hypermutation was affected by ASCIZ levels, indicating that ASCIZ does not directly control homologous recombination or formation of abasic sites. Loss of ASCIZ led to mild sensitivity to the base damaging agent methylmethane sulfonate (MMS), yet remarkably, suppressed the dramatic MMS hypersensitivity of polbeta-deficient cells. These data suggest that ASCIZ may affect the choice between competing base repair pathways in a manner that reduces the amount of substrates available for Ig gene conversion.


Asunto(s)
Alquilantes/farmacología , Proteínas Portadoras/metabolismo , Daño del ADN/genética , Reparación del ADN/genética , Conversión Génica , Genes de Inmunoglobulinas/genética , Animales , Proteínas Portadoras/genética , Línea Celular , ADN Polimerasa beta/genética , Resistencia a Medicamentos/genética , Humanos , Metilmetanosulfonato/farmacología , Ratones , Ratones Transgénicos , Mutágenos/farmacología , Proteínas Nucleares , Supresión Genética , Factores de Transcripción
20.
Mol Cell Biol ; 25(16): 6948-55, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16055708

RESUMEN

Homologous recombination (HR) requires nuclease activities at multiple steps, but the contribution of individual nucleases to the processing of double-strand DNA ends at different stages of HR has not been clearly defined. We used chicken DT40 cells to investigate the role of flap endonuclease 1 (Fen-1) in HR. FEN-1-deficient cells exhibited a significant decrease in the efficiency of immunoglobulin gene conversion while being proficient in recombination between sister chromatids, suggesting that Fen-1 may play a role in HR between sequences of considerable divergence. To clarify whether sequence divergence at DNA ends is truly the reason for the observed HR defect in FEN-1(-/-) cells we inserted a unique I-SceI restriction site in the genome and tested various donor and recipient HR substrates. We found that the efficiency of HR-mediated DNA repair was indeed greatly diminished when divergent sequences were present at the DNA break site. We conclude that Fen-1 eliminates heterologous sequences at DNA damage site and facilitates DNA repair by HR.


Asunto(s)
Daño del ADN , Endonucleasas de ADN Solapado/fisiología , Recombinación Genética , Animales , Secuencia de Bases , Ciclo Celular , Pollos , ADN/química , Reparación del ADN , ADN Complementario/metabolismo , Desoxirribonucleasas de Localización Especificada Tipo II/farmacología , Endonucleasas de ADN Solapado/metabolismo , Citometría de Flujo , Rayos gamma , Cinética , Modelos Genéticos , Datos de Secuencia Molecular , Mutación , Plásmidos/metabolismo , Proteínas de Saccharomyces cerevisiae , Homología de Secuencia de Ácido Nucleico , Intercambio de Cromátides Hermanas , Factores de Tiempo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA