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










Base de datos
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.
Artículo en Inglés | MEDLINE | ID: mdl-29567411

RESUMEN

Lamin is an intermediate protein underlying the nuclear envelope and it plays a key role in maintaining the integrity of the nucleus. A defect in the processing of its precursor by a metalloprotease, ZMPSTE24, results in the accumulation of farnesylated prelamin in the nucleus and causes various diseases, including Hutchinson-Gilford progeria syndrome (HGPS). However, the role of lamin processing is unclear in fish species. Here, we generated zmpste24-deficient medaka and evaluated their phenotype. Unlike humans and mice, homozygous mutants did not show growth defects or lifespan shortening, despite lamin precursor accumulation. Gonadosomatic indices, blood glucose levels, and regenerative capacity of fins were similar in 1-year-old mutants and their wild-type (WT) siblings. Histological examination showed that the muscles, subcutaneous fat tissues, and gonads were normal in the mutants at the age of 1 year. However, the mutants showed hypersensitivity to X-ray irradiation, although p53target genes, p21 and mdm2, were induced 6 h after irradiation. Immunostaining of primary cultured cells from caudal fins and visualization of nuclei using H2B-GFP fusion proteins revealed an abnormal nuclear shape in the mutants both in vitro and in vivo. The telomere lengths were significantly shorter in the mutants compared to WT. Taken together, these results suggest that zmpste24-deficient medaka phenocopied HGPS only partially and that abnormal nuclear morphology and lifespan shortening are two independent events in vertebrates.


Asunto(s)
Núcleo Celular/patología , Modelos Animales de Enfermedad , Proteínas de Peces/deficiencia , Proteínas de la Membrana/deficiencia , Metaloendopeptidasas/deficiencia , Oryzias/genética , Progeria/patología , Aletas de Animales/enzimología , Aletas de Animales/patología , Aletas de Animales/efectos de la radiación , Animales , Animales Modificados Genéticamente , Núcleo Celular/enzimología , Núcleo Celular/efectos de la radiación , Forma del Núcleo Celular/efectos de la radiación , Células Cultivadas , Codón sin Sentido , Femenino , Proteínas de Peces/química , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Técnicas de Inactivación de Genes , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Heterocigoto , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Oryzias/metabolismo , Progeria/enzimología , Progeria/genética , Tolerancia a Radiación , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Análisis de Supervivencia , Acortamiento del Telómero/efectos de la radiación
4.
Mol Cell Endocrinol ; 460: 104-122, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-28711606

RESUMEN

Although estrogens have been generally considered to play a critical role in ovarian differentiation in non-mammalian vertebrates, the specific functions of estrogens during ovarian differentiation remain unclear. We isolated two mutants with premature stops in the ovarian aromatase (cyp19a1) gene from an N-ethyl-N-nitrosourea-based gene-driven mutagenesis library of the medaka, Oryzias latipes. In XX mutants, gonads first differentiated into normal ovaries containing many ovarian follicles that failed to accumulate yolk. Subsequently, ovarian tissues underwent extensive degeneration, followed by the appearance of testicular tissues on the dorsal side of ovaries. In the newly formed testicular tissue, strong expression of gsdf was detected in sox9a2-positive somatic cells surrounding germline stem cells suggesting that gsdf plays an important role in testicular differentiation during estrogen-depleted female-to-male sex reversal. We conclude that endogenous estrogens synthesized after fertilization are not essential for early ovarian differentiation but are critical for the maintenance of adult ovaries.


Asunto(s)
Mutación con Pérdida de Función/genética , Oryzias/genética , Ovario/patología , Procesos de Determinación del Sexo , Maduración Sexual , Secuencia de Aminoácidos , Animales , Aromatasa/química , Aromatasa/genética , Secuencia de Bases , Linaje de la Célula , Regulación hacia Abajo/genética , Estrógenos/biosíntesis , Femenino , Perfilación de la Expresión Génica , Masculino , Folículo Ovárico/patología , Procesos de Determinación del Sexo/genética , Testículo/patología , Regulación hacia Arriba/genética , Vitelogeninas/metabolismo
5.
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
6.
Cell Rep ; 8(4): 1184-97, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25131198

RESUMEN

The thymus is a lymphoid organ unique to vertebrates, and it provides a unique microenvironment that facilitates the differentiation of immature hematopoietic precursors into mature T cells. We subjected the evolutionary trajectory of the thymic microenvironment to experimental analysis. A hypothetical primordial form of the thymus was established in mice by replacing FOXN1, the vertebrate-specific master regulator of thymic epithelial cell function, with its metazoan ancestor, FOXN4, thereby resetting the regulatory and coding changes that have occurred since the divergence of these two paralogs. FOXN4 exhibited substantial thymopoietic activity. Unexpectedly, histological changes and a functional imbalance between the lymphopoietic cytokine IL7 and the T cell specification factor DLL4 within the reconstructed thymus resulted in coincident but spatially segregated T and B cell development. Our results identify an evolutionary mechanism underlying the conversion of a general lymphopoietic organ to a site of exclusive T cell generation.


Asunto(s)
Proteínas del Ojo/genética , Factores de Transcripción Forkhead/genética , Timo/metabolismo , Animales , Linfocitos B/fisiología , Células Cultivadas , Células Epiteliales/metabolismo , Proteínas del Ojo/metabolismo , Factores de Transcripción Forkhead/metabolismo , Expresión Génica , Ingeniería Genética , Hematopoyesis Extramedular , Tejido Linfoide , Linfopoyesis , Ratones , Ratones Transgénicos , Oryzias , Filogenia , Linfocitos T/fisiología , Timo/citología , Pez Cebra
7.
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
8.
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
9.
Science ; 343(6166): 91-4, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24385628

RESUMEN

Social familiarity affects mating preference among various vertebrates. Here, we show that visual contact of a potential mating partner before mating (visual familiarization) enhances female preference for the familiarized male, but not for an unfamiliarized male, in medaka fish. Terminal-nerve gonadotropin-releasing hormone 3 (TN-GnRH3) neurons, an extrahypothalamic neuromodulatory system, function as a gate for activating mating preferences based on familiarity. Basal levels of TN-GnRH3 neuronal activity suppress female receptivity for any male (default mode). Visual familiarization facilitates TN-GnRH3 neuron activity (preference mode), which correlates with female preference for the familiarized male. GnRH3 peptides, which are synthesized specifically in TN-GnRH3 neurons, are required for the mode-switching via self-facilitation. Our study demonstrates the central neural mechanisms underlying the regulation of medaka female mating preference based on visual social familiarity.


Asunto(s)
Hormona Liberadora de Gonadotropina/fisiología , Preferencia en el Apareamiento Animal , Neuronas/fisiología , Oryzias/fisiología , Ácido Pirrolidona Carboxílico/análogos & derivados , Reconocimiento en Psicología , Percepción Visual , Animales , Femenino , Masculino , Mutación , Oryzias/genética , Factores Sexuales
10.
Cancer Res ; 73(14): 4362-71, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23576554

RESUMEN

DNA double-strand breaks (DSB) occur frequently during replication in sister chromatids and are dramatically increased when cells are exposed to chemotherapeutic agents including camptothecin. Such DSBs are efficiently repaired specifically by homologous recombination (HR) with the intact sister chromatid. HR, therefore, plays pivotal roles in cellular proliferation and cellular tolerance to camptothecin. Mammalian cells carry several structure-specific endonucleases, such as Xpf-Ercc1 and Mus81-Eme1, in which Xpf and Mus81 are the essential subunits for enzymatic activity. Here, we show the functional overlap between Xpf and Mus81 by conditionally inactivating Xpf in the chicken DT40 cell line, which has no Mus81 ortholog. Although mammalian cells deficient in either Xpf or Mus81 are viable, Xpf inactivation in DT40 cells was lethal, resulting in a marked increase in the number of spontaneous chromosome breaks. Similarly, inactivation of both Xpf and Mus81 in human HeLa cells and murine embryonic stem cells caused numerous spontaneous chromosome breaks. Furthermore, the phenotype of Xpf-deficient DT40 cells was reversed by ectopic expression of human Mus81-Eme1 or human Xpf-Ercc1 heterodimers. These observations indicate the functional overlap of Xpf-Ercc1 and Mus81-Eme1 in the maintenance of genomic DNA. Both Mus81-Eme1 and Xpf-Ercc1 contribute to the completion of HR, as evidenced by the data that the expression of Mus81-Eme1 or Xpf-Ercc1 diminished the number of camptothecin-induced chromosome breaks in Xpf-deficient DT40 cells, and to preventing early steps in HR by deleting XRCC3 suppressed the nonviability of Xpf-deficient DT40 cells. In summary, Xpf and Mus81 have a substantially overlapping function in completion of HR.


Asunto(s)
Reparación del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Endonucleasas/genética , Endonucleasas/metabolismo , Recombinación Homóloga , Animales , Muerte Celular/genética , Línea Celular Tumoral , Pollos , Aberraciones Cromosómicas , Roturas del ADN de Doble Cadena , Células HeLa , Humanos , Ratones
11.
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
12.
Mol Biol Cell ; 24(9): 1387-95, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23447699

RESUMEN

ATF6α and ATF6ß are membrane-bound transcription factors activated by regulated intramembrane proteolysis in response to endoplasmic reticulum (ER) stress to induce various ER quality control proteins. ATF6α- and ATF6ß single-knockout mice develop normally, but ATF6α/ß double knockout causes embryonic lethality, the reason for which is unknown. Here we show in medaka fish that ATF6α is primarily responsible for transcriptional induction of the major ER chaperone BiP and that ATF6α/ß double knockout, but not ATF6α- or ATF6ß single knockout, causes embryonic lethality, as in mice. Analyses of ER stress reporters reveal that ER stress occurs physiologically during medaka early embryonic development, particularly in the brain, otic vesicle, and notochord, resulting in ATF6α- and ATF6ß-mediated induction of BiP, and that knockdown of the α1 chain of type VIII collagen reduces such ER stress. The absence of transcriptional induction of several ER chaperones in ATF6α/ß double knockout causes more profound ER stress and impaired notochord development, which is partially rescued by overexpression of BiP. Thus ATF6α/ß-mediated adjustment of chaperone levels to increased demands in the ER is essential for development of the notochord, which synthesizes and secretes large amounts of extracellular matrix proteins to serve as the body axis before formation of the vertebra.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas de Peces/metabolismo , Proteínas de Choque Térmico/metabolismo , Notocorda/embriología , Oryzias/embriología , Factor de Transcripción Activador 6/genética , Secuencia de Aminoácidos , Animales , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico , Femenino , Proteínas de Peces/genética , Técnicas de Inactivación de Genes , Genes Letales , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/genética , Masculino , Datos de Secuencia Molecular , Notocorda/metabolismo , Oryzias/metabolismo , Mutación Puntual , Empalme del ARN , Activación Transcripcional
13.
FEBS Lett ; 587(9): 1316-25, 2013 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-23499937

RESUMEN

Kufor-Rakeb syndrome (KRS) was originally described as an autosomal recessive form of early-onset parkinsonism with pyramidal degeneration and dementia. ATP13A2 was identified as the causative gene in KRS. ATP13A2 encodes the ATP13A2 protein, which is a lysosomal type5 P-type ATPase, and ATP13A2 mutations are linked to autosomal recessive familial parkinsonism. Here, we report that normal ATP13A2 localizes in the lysosome, whereas disease-associated variants remain in the endoplasmic reticulum. Cathepsin D activity was decreased in ATP13A2-knockdown cells that displayed lysosome-like bodies characterized by fingerprint-like structures. Furthermore, an atp13a2 mutation in medaka fish resulted in dopaminergic neuronal death, decreased cathepsin D activity, and fingerprint-like structures in the brain. Based on these results, lysosome abnormality is very likely to be the primary cause of KRS/PARK9.


Asunto(s)
Catepsina D/metabolismo , Neuronas Dopaminérgicas/citología , Cuerpos de Inclusión/metabolismo , ATPasas de Translocación de Protón/deficiencia , ATPasas de Translocación de Protón/genética , Animales , Línea Celular Tumoral , Neuronas Dopaminérgicas/enzimología , Neuronas Dopaminérgicas/patología , Retículo Endoplásmico/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Lisosomas/metabolismo , Mutación , Oryzias/genética , Transporte de Proteínas
14.
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
15.
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
16.
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
17.
PLoS One ; 7(1): e29982, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22253846

RESUMEN

The sex determining gene is divergent among different animal species. However, sox9 is up-regulated in the male gonads in a number of species in which it is the essential regulator of testis determination. It is therefore often discussed that the sex determining gene-sox9 axis functions in several vertebrates. In our current study, we show that sox9b in the medaka (Oryzias latipes) is one of the orthologues of mammalian Sox9 at syntenic and expression levels. Medaka sox9b affects the organization of extracellular matrices, which represents a conserved role of sox9, but does not directly regulate testis determination. We made this determination via gene expression and phenotype analyses of medaka with different copy numbers of sox9b. Sox9b is involved in promoting cellular associations and is indispensible for the proper proliferation and survival of germ cells in both female and male medaka gonads. Medaka mutants that lack sox9b function exhibit a seemingly paradoxical phenotype of sex reversal to male. This is explained by a reduction in the germ cell number associated with aberrant extracellular matrices. Together with its identified roles in other vertebrate gonads, a testis-determining role for Sox9 in mammals is likely to have been neofunctionalized and appended to its conserved role in germ cell maintenance.


Asunto(s)
Secuencia Conservada , Células Germinativas/citología , Células Germinativas/metabolismo , Oryzias/metabolismo , Factor de Transcripción SOX9/química , Factor de Transcripción SOX9/metabolismo , Alelos , Animales , Apoptosis/genética , Secuencia de Bases , Recuento de Células , Proliferación Celular , Supervivencia Celular , Matriz Extracelular/metabolismo , Femenino , Regulación de la Expresión Génica , Heterocigoto , Masculino , Datos de Secuencia Molecular , Mutación/genética , Oryzias/genética , Fenotipo , Factor de Transcripción SOX9/genética , Homología de Secuencia de Aminoácido , Procesos de Determinación del Sexo/genética , Diferenciación Sexual/genética , Sintenía/genética , Testículo/citología , Testículo/metabolismo
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...