Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros













Base de dados
Intervalo de ano de publicação
1.
Biol Trace Elem Res ; 2023 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-37932617

RESUMO

Oral intake of purified selenoneine and seafoods has been reported to result in selenoneine accumulation in erythrocytes in mice and human. In addition, Se-methylselenoneine was suggested to be produced as a metabolite of selenoneine in the urine and whole blood of humans. In order to confirm the molecular mechanism of production of Se-methylselenoneine, a stable isotope (Se-76) labeled selenoneine was biosynthesized using genetically modified fission yeast and administered to mice. The Se-76-labeled Se-methylselenoneine was detected in urine but Se-78 and Se-80-labeled Se-methylselenoneine arising from natural isotopes of Se was hardly detected. These results suggest that Se-methylselenoneine was a metabolite and the excreted form of selenoneine. The methylation of selenoneine in mice administered selenoneine continuously was evaluated by the analyses of organs using an online liquid chromatograph system with an inductively coupled plasma mass spectrometer (LC-ICP-MS). These experiments indicate that selenoneine is methylated in the liver and (or) kidneys.

2.
Brain ; 145(3): 1139-1150, 2022 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-35355059

RESUMO

Cerebellar ataxia, neuropathy, vestibular areflexia syndrome (CANVAS) is a late-onset, slow-progressing multisystem neurodegenerative disorder. Biallelic AAGGG repeat expansion in RFC1 has been identified as causative of this disease, and repeat conformation heterogeneity (ACAGG repeat) was also recently implied. To molecularly characterize this disease in Japanese patients with adult-onset ataxia, we accumulated and screened 212 candidate families by an integrated approach consisting of flanking PCR, repeat-primed PCR, Southern blotting and long-read sequencing using Sequel II, GridION or PromethION. We identified 16 patients from 11 families, of whom seven had ACAGG expansions [(ACAGG)exp/(ACAGG)exp] (ACAGG homozygotes), two had ACAGG and AAGGG expansions [(ACAGG)exp/(AAGGG)exp] (ACAGG/AAGGG compound heterozygotes) and seven had AAGGG expansions [(AAGGG)exp/(AAGGG)exp] (AAGGG homozygotes). The overall detection rate was 5.2% (11/212 families including one family having two expansion genotypes). Long-read sequencers revealed the entire sequence of both AAGGG and ACAGG repeat expansions at the nucleotide level of resolution. Clinical assessment and neuropathology results suggested that patients with ACAGG expansions have similar clinical features to previously reported patients with homozygous AAGGG expansions, although motor neuron involvement was more notable in patients with ACAGG expansions (even if one allele was involved). Furthermore, a later age of onset and slower clinical progression were implied in patients with ACAGG/AAGGG compound heterozygous expansions compared with either ACAGG or AAGGG homozygotes in our very limited cohort. Our study clearly shows the occurrence of repeat conformation heterogeneity, with possible different impacts on the affected nervous systems. The difference in disease onset and progression between compound heterozygotes and homozygotes might also be suspected but with very limited certainty due to the small sample number of cases in our study. Studies of additional patients are needed to confirm this.


Assuntos
Vestibulopatia Bilateral , Ataxia Cerebelar , Doenças do Sistema Nervoso Periférico , Doenças Vestibulares , Neuronite Vestibular , Adulto , Ataxia , Vestibulopatia Bilateral/diagnóstico , Vestibulopatia Bilateral/genética , Ataxia Cerebelar/diagnóstico , Ataxia Cerebelar/genética , Humanos , Reflexo Anormal , Proteína de Replicação C/genética , Síndrome , Doenças Vestibulares/genética
3.
Am J Hum Genet ; 106(4): 549-558, 2020 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-32169168

RESUMO

De novo variants (DNVs) cause many genetic diseases. When DNVs are examined in the whole coding regions of genes in next-generation sequencing analyses, pathogenic DNVs often cluster in a specific region. One such region is the last exon and the last 50 bp of the penultimate exon, where truncating DNVs cause escape from nonsense-mediated mRNA decay [NMD(-) region]. Such variants can have dominant-negative or gain-of-function effects. Here, we first developed a resource of rates of truncating DNVs in NMD(-) regions under the null model of DNVs. Utilizing this resource, we performed enrichment analysis of truncating DNVs in NMD(-) regions in 346 developmental and epileptic encephalopathy (DEE) trios. We observed statistically significant enrichment of truncating DNVs in semaphorin 6B (SEMA6B) (p value: 2.8 × 10-8; exome-wide threshold: 2.5 × 10-6). The initial analysis of the 346 individuals and additional screening of 1,406 and 4,293 independent individuals affected by DEE and developmental disorders collectively identified four truncating DNVs in the SEMA6B NMD(-) region in five individuals who came from unrelated families (p value: 1.9 × 10-13) and consistently showed progressive myoclonic epilepsy. RNA analysis of lymphoblastoid cells established from an affected individual showed that the mutant allele escaped NMD, indicating stable production of the truncated protein. Importantly, heterozygous truncating variants in the NMD(+) region of SEMA6B are observed in general populations, and SEMA6B is most likely loss-of-function tolerant. Zebrafish expressing truncating variants in the NMD(-) region of SEMA6B orthologs displayed defective development of brain neurons and enhanced pentylenetetrazole-induced seizure behavior. In summary, we show that truncating DNVs in the final exon of SEMA6B cause progressive myoclonic epilepsy.


Assuntos
Exoma/genética , Éxons/genética , Predisposição Genética para Doença/genética , Variação Genética/genética , Epilepsias Mioclônicas Progressivas/genética , Semaforinas/genética , Adolescente , Adulto , Alelos , Animais , Feminino , Heterozigoto , Humanos , Masculino , Degradação do RNAm Mediada por Códon sem Sentido/genética , Convulsões/genética , Adulto Jovem , Peixe-Zebra/genética
4.
Dev Comp Immunol ; 80: 15-23, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-27965016

RESUMO

We have monoclonal antibodies (mAbs) against CD4-1 (6D1) and CD8α (2C3) in ginbuna crucian carp Carassius auratus langsdorfii. In our previous studies we showed that 2C3 mAb positive cells are the primary cell type showing specific cytotoxicity against allogeneic targets, suggesting that CD8α+ lymphocytes in ginbuna are equivalent to cytotoxic T lymphocytes (CTLs) in mammals. We further demonstrated the helper T cell function of 6D1 mAb positive cells by studying mixed leukocyte culture (MLC) and hapten/carrier effects. Here, we report that our mAbs cross-react with zebrafish lymphocytes. First, mAbs 6D1 and 2C3 recognized 7-11% of zebrafish lymphocytes that were ZAP-70 positive and had the typical morphology of lymphocytes. Second, to verify the cell types reacting with the 6D1 and 2C3 mAbs we examined the expression profiles of zebrafish lymphocyte surface markers in FACS-sorted lymphocytes from kidney. cd4-1 (cd8a) and tcrac but not iglc transcripts were detected in 6D1(2C3)+ lymphocytes, whereas cd4-1 (cd8a) transcripts were not found in 6D1 (2C3)- lymphocytes. Third, we further confirmed that 6D1 reacted with zebrafish CD4-1 but not CD4-2, and 2C3 recognized zebrafish CD8α expressed on HEK293T cells. Collectively, these findings suggest that 6D1+ and 2C3+ lymphocytes in zebrafish are equivalent to CD4+ and CD8α+ T lymphocytes in mammals, respectively. Furthermore, we found the cross-reactivity of our 6D1 and 2C3 mAbs with other cyprinid species including goldfish, common carp and grass carp.


Assuntos
Anticorpos Monoclonais/metabolismo , Antígenos CD4/imunologia , Antígenos CD8/imunologia , Carpas/imunologia , Reações Cruzadas , Proteínas de Peixes/imunologia , Linfócitos/imunologia , Peixe-Zebra/imunologia , Animais , Anticorpos Monoclonais/imunologia , Antígenos CD4/genética , Antígenos CD8/genética , Cyprinidae , Citotoxicidade Imunológica , Proteínas de Peixes/genética , Células HEK293 , Humanos , Mamíferos , Transcriptoma
5.
Allergol Int ; 65(3): 272-9, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26875665

RESUMO

BACKGROUND: Parvalbumin and collagen have been identified as cross-reactive allergens for fish allergies. Although doctors realize that various fish elicit allergies, the targets of food allergen labeling laws were only mackerels and salmons in Japan and mackerels in South Korea. This study aimed to reveal the causative species for fish allergy via questionnaires and blood tests. METHODS: Questionnaire research was conducted in Japan via the internet concerning allergies for fish-allergic patients or their family members. Next, IgE reactivities and cross-reactivities of 26 fish species were analyzed using sera obtained from 16 Japanese patients who were allergic to fish parvalbumin or collagen by enzyme-linked immunosorbent assay (ELISA) and inhibition ELISA. RESULTS: Questionnaire research revealed that 88% patients cannot eat mackerel and salmon in addition to other fish. In addition, 85% respondents were not satisfied with the current food allergen labeling law. In ELISA analyses, we clarified that pooled serum obtained from patients with fish parvalbumin-specific allergies exhibited IgE reactivity to the extracts of most fish species, and pooled serum obtained from patients with fish collagen-specific allergies displayed IgE reactivity to the extracts of all types of fish. Inhibition ELISA experiments revealed cross-reactivities of parvalbumin or collagen to extracts from all fish tested. CONCLUSIONS: Most patients with fish allergies displayed allergic symptoms following the intake of various fish species. In addition, fish parvalbumin and collagen were causative factors of fish allergy and were highly cross-reactive fish panallergens. Therefore, current laws should be revised in Japan and South Korea.


Assuntos
Alérgenos/imunologia , Reações Cruzadas/imunologia , Peixes/imunologia , Hipersensibilidade Alimentar/diagnóstico , Hipersensibilidade Alimentar/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Criança , Pré-Escolar , Colágeno/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Peixes/classificação , Hipersensibilidade Alimentar/epidemiologia , Humanos , Imunoglobulina E/sangue , Imunoglobulina E/imunologia , Lactente , Recém-Nascido , Japão/epidemiologia , Masculino , Pessoa de Meia-Idade , Parvalbuminas/imunologia , Inquéritos e Questionários , Adulto Jovem
6.
Am J Hum Genet ; 97(4): 555-66, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26411495

RESUMO

The nuclear pore complex (NPC) is a huge protein complex embedded in the nuclear envelope. It has central functions in nucleocytoplasmic transport, nuclear framework, and gene regulation. Nucleoporin 107 kDa (NUP107) is a component of the NPC central scaffold and is an essential protein in all eukaryotic cells. Here, we report on biallelic NUP107 mutations in nine affected individuals who are from five unrelated families and show early-onset steroid-resistant nephrotic syndrome (SRNS). These individuals have pathologically focal segmental glomerulosclerosis, a condition that leads to end-stage renal disease with high frequency. NUP107 is ubiquitously expressed, including in glomerular podocytes. Three of four NUP107 mutations detected in the affected individuals hamper NUP107 binding to NUP133 (nucleoporin 133 kDa) and NUP107 incorporation into NPCs in vitro. Zebrafish with nup107 knockdown generated by morpholino oligonucleotides displayed hypoplastic glomerulus structures and abnormal podocyte foot processes, thereby mimicking the pathological changes seen in the kidneys of the SRNS individuals with NUP107 mutations. Considering the unique properties of the podocyte (highly differentiated foot-process architecture and slit membrane and the inability to regenerate), we propose a "podocyte-injury model" as the pathomechanism for SRNS due to biallelic NUP107 mutations.


Assuntos
Idade de Início , Mutação/genética , Síndrome Nefrótica/congênito , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Alelos , Animais , Células Cultivadas , Criança , Pré-Escolar , Citoplasma/metabolismo , Feminino , Haplótipos , Humanos , Immunoblotting , Imunoprecipitação , Lactente , Rim/metabolismo , Rim/patologia , Masculino , Microscopia de Fluorescência , Síndrome Nefrótica/etiologia , Síndrome Nefrótica/patologia , Poro Nuclear , Complexo de Proteínas Formadoras de Poros Nucleares/antagonistas & inibidores , Oligorribonucleotídeos Antissenso/farmacologia , Linhagem , Podócitos/metabolismo , Podócitos/patologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Peixe-Zebra/crescimento & desenvolvimento , Proteínas de Peixe-Zebra/antagonistas & inibidores
7.
Nat Commun ; 5: 4011, 2014 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-24886874

RESUMO

Coffin-Siris syndrome (CSS) is a congenital disorder characterized by growth deficiency, intellectual disability, microcephaly, characteristic facial features and hypoplastic nails of the fifth fingers and/or toes. We previously identified mutations in five genes encoding subunits of the BAF complex, in 55% of CSS patients. Here we perform whole-exome sequencing in additional CSS patients, identifying de novo SOX11 mutations in two patients with a mild CSS phenotype. sox11a/b knockdown in zebrafish causes brain abnormalities, potentially explaining the brain phenotype of CSS. SOX11 is the downstream transcriptional factor of the PAX6-BAF complex, highlighting the importance of the BAF complex and SOX11 transcriptional network in brain development.


Assuntos
Anormalidades Múltiplas/genética , Face/anormalidades , Deformidades Congênitas da Mão/genética , Deficiência Intelectual/genética , Micrognatismo/genética , Pescoço/anormalidades , Fatores de Transcrição SOX/genética , Fatores de Transcrição SOXC/genética , Proteínas de Peixe-Zebra/genética , Adolescente , Animais , Pré-Escolar , Estudos de Coortes , Feminino , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Mutação , Peixe-Zebra
8.
Am J Hum Genet ; 93(1): 6-18, 2013 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-23746549

RESUMO

Nemaline myopathy (NEM) is a common congenital myopathy. At the very severe end of the NEM clinical spectrum are genetically unresolved cases of autosomal-recessive fetal akinesia sequence. We studied a multinational cohort of 143 severe-NEM-affected families lacking genetic diagnosis. We performed whole-exome sequencing of six families and targeted gene sequencing of additional families. We identified 19 mutations in KLHL40 (kelch-like family member 40) in 28 apparently unrelated NEM kindreds of various ethnicities. Accounting for up to 28% of the tested individuals in the Japanese cohort, KLHL40 mutations were found to be the most common cause of this severe form of NEM. Clinical features of affected individuals were severe and distinctive and included fetal akinesia or hypokinesia and contractures, fractures, respiratory failure, and swallowing difficulties at birth. Molecular modeling suggested that the missense substitutions would destabilize the protein. Protein studies showed that KLHL40 is a striated-muscle-specific protein that is absent in KLHL40-associated NEM skeletal muscle. In zebrafish, klhl40a and klhl40b expression is largely confined to the myotome and skeletal muscle, and knockdown of these isoforms results in disruption of muscle structure and loss of movement. We identified KLHL40 mutations as a frequent cause of severe autosomal-recessive NEM and showed that it plays a key role in muscle development and function. Screening of KLHL40 should be a priority in individuals who are affected by autosomal-recessive NEM and who present with prenatal symptoms and/or contractures and in all Japanese individuals with severe NEM.


Assuntos
Proteínas Musculares/metabolismo , Músculo Esquelético/patologia , Mutação de Sentido Incorreto , Miopatias da Nemalina/genética , Substituição de Aminoácidos , Animais , Povo Asiático/genética , Estudos de Coortes , Mutação da Fase de Leitura , Genes Recessivos , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Proteínas Musculares/genética , Miopatias da Nemalina/etnologia , Miopatias da Nemalina/patologia , Linhagem , Polimorfismo de Nucleotídeo Único , Índice de Gravidade de Doença , Peixe-Zebra/genética
9.
Mar Biotechnol (NY) ; 15(5): 559-70, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23709046

RESUMO

The selenium (Se)-containing antioxidant selenoneine (2-selenyl-N α,N α,N α-trimethyl-L-histidine) has recently been discovered to be the predominant form of organic Se in tuna blood. Although dietary intake of fish Se has been suggested to reduce methylmercury (MeHg) toxicity, the molecular mechanism of MeHg detoxification by Se has not yet been determined. Here, we report evidence that selenoneine accelerates the excretion and demethylation of MeHg, mediated by a selenoneine-specific transporter, organic cations/carnitine transporter-1 (OCTN1). Selenoneine was incorporated into human embryonic kidney HEK293 cells transiently overexpressing OCTN1 and zebrafish blood cells by OCTN1. The K m for selenoneine uptake was 13.0 µM in OCTN1-overexpressing HEK293 cells and 9.5 µM in zebrafish blood cells, indicating high affinity of OCTN1 for selenoneine in human and zebrafish cells. When such OCTN1-expressing cells and embryos were exposed to MeHg-cysteine (MeHgCys), MeHg accumulation was decreased and the excretion and demethylation of MeHg were enhanced by selenoneine. In addition, exosomal secretion vesicles were detected in the culture water of embryos that had been microinjected with MeHgCys, suggesting that these may be responsible for MeHg excretion and demethylation. In contrast, OCTN1-deficient embryos accumulated MeHg, and MeHg excretion and demethylation were decreased. Furthermore, Hg accumulation was decreased in OCTN1-overexpressing HEK293 cells, but not in mock vector-transfected cells, indicating that selenoneine and OCTN1 can regulate MeHg detoxification in human cells. Thus, the selenoneine-mediated OCTN1 system regulates secretory lysosomal vesicle formation and MeHg demethylation.


Assuntos
Histidina/análogos & derivados , Inativação Metabólica/fisiologia , Compostos de Metilmercúrio/farmacocinética , Compostos Organosselênicos/farmacologia , Peixe-Zebra/fisiologia , Animais , Elementos Antissenso (Genética) , Western Blotting , Fluorescência , Células HEK293 , Histidina/farmacologia , Humanos , Marcação In Situ das Extremidades Cortadas , Larva/efeitos dos fármacos , Lisossomos/metabolismo , Compostos de Metilmercúrio/toxicidade , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Simportadores , Ultracentrifugação , Peixe-Zebra/metabolismo
10.
J Biol Chem ; 287(27): 23047-56, 2012 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-22549779

RESUMO

Cell division cycle 48 (CDC48), a ubiquitin-dependent molecular chaperone, is thought to mediate a variety of degradative and regulatory processes and maintain cellular homoeostasis. To investigate the protective function of CDC48 against accumulated ubiquitinated proteins during neurodevelopment, we developed an in vivo bioassay technique that detects expression and accumulation of fluorescent proteins with a polyubiquitination signal at the N terminus. When we introduced CDC48 antisense morpholino oligonucleotides into zebrafish embryos, the morphant embryos were lethal and showed defects in neuronal outgrowth and neurodegeneration, and polyubiquitinated fluorescent proteins accumulated in the inner plexiform and ganglion cell layers, as well as the diencephalon and mesencephalon, indicating that the degradation of polyubiquitinated proteins by the ubiquitin-proteasome system was blocked. These abnormal phenotypes in the morphant were rescued by CDC48 or human valosin-containing protein overexpression. Therefore, the protective function of CDC48 is essential for neurodevelopment.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Ciclo Celular/metabolismo , Degeneração Neural/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Adenosina Trifosfatases/química , Adenosina Trifosfatases/genética , Animais , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Divisão Celular/fisiologia , Diencéfalo/anormalidades , Diencéfalo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Mesencéfalo/anormalidades , Mesencéfalo/metabolismo , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Degeneração Neural/fisiopatologia , Fenótipo , Estrutura Terciária de Proteína , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/enzimologia , Medula Espinal/anormalidades , Medula Espinal/metabolismo , Proteína com Valosina , Peixe-Zebra/genética
11.
Mar Biotechnol (NY) ; 14(4): 491-501, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22290406

RESUMO

Autophagy is well established as a starvation-induced process in yeast and mammalian cells and tissues. To elucidate the cellular mechanisms induced by starvation in fish, we characterized the induction of autophagy in cultured zebrafish cells under starvation conditions. As an autophagic marker protein, the microtubule-associated protein 1-light chain 3B protein (MAP1-LC3B) was cloned from the fish cells, and its expression and localization were characterized. In zebrafish embryonic (ZE) cells, posttranslational modifications produced two distinct forms of MAP1-LC3B, i.e., a cytosolic form and a membrane-bound form (types I and II, respectively). Immunofluorescence microscopy revealed fluorescently labeled autophagosomes in cells stably transfected with a green fluorescent protein (GFP)­MAP1-LC3B fusion protein and showed that this protein accumulated in punctate dots in a time-dependent manner in response to amino acid starvation. Starvation also induced the degradation of long-lived proteins. Treatment with 3-methyladenine and wortmannin, two class-III inhibitors of phosphoinositide 3-kinase (PI3K), repressed autophagy under starvation conditions, indicating that the PI3K class-III pathway regulates starvation-induced autophagy in fish.


Assuntos
Aminoácidos/deficiência , Autofagia , Inanição/fisiopatologia , Peixe-Zebra/metabolismo , Animais
12.
ACS Chem Biol ; 6(9): 900-4, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21732624

RESUMO

The reduction of plasma low-density lipoprotein levels by HMG-CoA reductase inhibitors, or statins, has had a revolutionary impact in medicine, but muscle-related side effects remain a dose-limiting toxicity in many patients. We describe a chemical epistasis approach that can be useful in refining the mechanism of statin muscle toxicity, as well as in screening for agents that suppress muscle toxicity while preserving the ability of statins to increase the expression of the low-density lipoprotein receptor. Using this approach, we identified one compound that attenuates the muscle side effects in both cellular and animal models of statin toxicity, likely by influencing Rab prenylation. Our proof-of-concept screen lays the foundation for truly high-throughput screens that could help lead to the development of clinically useful adjuvants that can one day be co-administered with statins.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Doenças Musculares/induzido quimicamente , Doenças Musculares/prevenção & controle , Animais , Carbazóis/farmacologia , Linhagem Celular , Humanos , Estrutura Molecular , Peso Molecular , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/patologia , Doenças Musculares/patologia , Peixe-Zebra
13.
PLoS One ; 6(3): e17688, 2011 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-21479207

RESUMO

BACKGROUND: Mutations that disrupt the conversion of prelamin A to mature lamin A cause the rare genetic disorder Hutchinson-Gilford progeria syndrome and a group of laminopathies. Our understanding of how A-type lamins function in vivo during early vertebrate development through aging remains limited, and would benefit from a suitable experimental model. The zebrafish has proven to be a tractable model organism for studying both development and aging at the molecular genetic level. Zebrafish show an array of senescence symptoms resembling those in humans, which can be targeted to specific aging pathways conserved in vertebrates. However, no zebrafish models bearing human premature senescence currently exist. PRINCIPAL FINDINGS: We describe the induction of embryonic senescence and laminopathies in zebrafish harboring disturbed expressions of the lamin A gene (LMNA). Impairments in these fish arise in the skin, muscle and adipose tissue, and sometimes in the cartilage. Reduced function of lamin A/C by translational blocking of the LMNA gene induced apoptosis, cell-cycle arrest, and craniofacial abnormalities/cartilage defects. By contrast, induced cryptic splicing of LMNA, which generates the deletion of 8 amino acid residues lamin A (zlamin A-Δ8), showed embryonic senescence and S-phase accumulation/arrest. Interestingly, the abnormal muscle and lipodystrophic phenotypes were common in both cases. Hence, both decrease-of-function of lamin A/C and gain-of-function of aberrant lamin A protein induced laminopathies that are associated with mesenchymal cell lineages during zebrafish early development. Visualization of individual cells expressing zebrafish progerin (zProgerin/zlamin A-Δ37) fused to green fluorescent protein further revealed misshapen nuclear membrane. A farnesyltransferase inhibitor reduced these nuclear abnormalities and significantly prevented embryonic senescence and muscle fiber damage induced by zProgerin. Importantly, the adult Progerin fish survived and remained fertile with relatively mild phenotypes only, but had shortened lifespan with obvious distortion of body shape. CONCLUSION: We generated new zebrafish models for a human premature aging disorder, and further demonstrated the utility for studying laminopathies. Premature aging could also be modeled in zebrafish embryos. This genetic model may thus provide a new platform for future drug screening as well as genetic analyses aimed at identifying modifier genes that influence not only progeria and laminopathies but also other age-associated human diseases common in vertebrates.


Assuntos
Envelhecimento/patologia , Embrião não Mamífero/patologia , Lamina Tipo A/genética , Progéria/complicações , Progéria/patologia , Peixe-Zebra/metabolismo , Envelhecimento/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Apoptose/efeitos dos fármacos , Cartilagem/anormalidades , Cartilagem/efeitos dos fármacos , Cartilagem/patologia , Modelos Animais de Doenças , Embrião não Mamífero/anormalidades , Embrião não Mamífero/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Proteínas de Fluorescência Verde/metabolismo , Humanos , Lamina Tipo A/química , Lipodistrofia/complicações , Lipodistrofia/patologia , Longevidade/efeitos dos fármacos , Dados de Sequência Molecular , Músculos/anormalidades , Músculos/efeitos dos fármacos , Músculos/patologia , Proteínas Mutantes/metabolismo , Proteínas Nucleares/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Precursores de Proteínas/metabolismo , Transgenes/genética , Peixe-Zebra/genética
14.
FEBS J ; 278(4): 673-85, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21205201

RESUMO

A cell line derived from the tailfin of the marine teleost yellowtail fish Seriola quinqueradiata was established to examine cellular temperature regulation in an ectothermic animal. Three cytosolic members of the HSP70 family, heat-shock cognate proteins HSC70-1, HSC70-2 and heat-shock protein HSP70, were isolated from cultured yellowtail cells as stress-responsive biomarkers. Expression of hsp70 was heat-inducible, in contrast to the hsc70-1 gene product, which was expressed constitutively. In addition, expression of hsc70-2 was only induced under severe heat-shock conditions. Subcellular fractionation and immunocytochemistry showed localization of HSC70/HSP70 in the lysosomes, indicating that chaperone-mediated autophagy is induced by heat shock. Thus, chaperone-mediated autophagy is assisted by HSC70/HSP70, and heat-inducible expression of the genes encoding these proteins may be responsible for survival and adaptation under heat-shock conditions in fish cells.


Assuntos
Autofagia , Regulação da Expressão Gênica , Proteínas de Choque Térmico HSC70/genética , Proteínas de Choque Térmico HSP70/genética , Perciformes/genética , Sequência de Aminoácidos , Animais , Células Cultivadas , Clonagem Molecular , Proteínas de Choque Térmico HSC70/química , Proteínas de Choque Térmico HSP70/química , Humanos , Dados de Sequência Molecular , Filogenia , Transporte Proteico , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Temperatura
15.
FASEB J ; 23(9): 2844-54, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19406843

RESUMO

Statins are widely used to treat hypercholesterolemia but can lead to a number of side effects in muscle, including rhabdomyolysis. Our recent findings implicated the induction of atrogin-1, a gene required for the development of muscle atrophy, in statin-induced muscle damage. Since statins inhibit many biochemical reactions besides cholesterol synthesis, we sought to define the statin-inhibited pathways responsible for atrogin-1 expression and muscle damage. We report here that lovastatin-induced atrogin-1 expression and muscle damage in cultured mouse myotubes and zebrafish can be prevented in the presence of geranylgeranol but not farnesol. Further, inhibitors of the transfer of geranylgeranyl isoprene units to protein targets cause statin muscle damage and atrogin-1 induction in cultured cells and in fish. These findings support the concept that dysfunction of small GTP-binding proteins lead to statin-induced muscle damage since these molecules require modification by geranylgeranyl moieties for their cellular localization and activity. Collectively, our animal and in vitro findings shed light on the molecular mechanism of statin-induced myopathy and suggest that atrogin-1 may be regulated by novel signaling pathways.


Assuntos
Proteínas F-Box/genética , Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Fibras Musculares Esqueléticas/patologia , Proteínas Musculares/genética , Atrofia Muscular/induzido quimicamente , Prenilação/genética , Proteínas Ligases SKP Culina F-Box/genética , Proteínas de Peixe-Zebra/genética , Animais , Células Cultivadas , Proteínas de Ligação ao GTP , Lovastatina/efeitos adversos , Camundongos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Atrofia Muscular/etiologia , Ativação Transcricional , Peixe-Zebra
16.
PLoS One ; 3(10): e3364, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18846223

RESUMO

Although it is clear that telomerase expression is crucial for the maintenance of telomere homeostasis, there is increasing evidence that the TERT protein can have physiological roles that are independent of this central function. To further examine the role of telomerase during vertebrate development, the zebrafish telomerase reverse transcriptase (zTERT) was functionally characterized. Upon zTERT knockdown, zebrafish embryos show reduced telomerase activity and are viable, but develop pancytopenia resulting from aberrant hematopoiesis. The blood cell counts in TERT-depleted zebrafish embryos are markedly decreased and hematopoietic cell differentiation is impaired, whereas other somatic lineages remain morphologically unaffected. Although both primitive and definitive hematopoiesis is disrupted by zTERT knockdown, the telomere lengths are not significantly altered throughout early development. Induced p53 deficiency, as well as overexpression of the anti-apoptotic proteins Bcl-2 and E1B-19K, significantly relieves the decreased blood cells numbers caused by zTERT knockdown, but not the impaired blood cell differentiation. Surprisingly, only the reverse transcriptase motifs of zTERT are crucial, but the telomerase RNA-binding domain of zTERT is not required, for rescuing complete hematopoiesis. This is therefore the first demonstration of a non-canonical catalytic activity of TERT, which is different from "authentic" telomerase activity, is required for during vertebrate hematopoiesis. On the other hand, zTERT deficiency induced a defect in hematopoiesis through a potent and specific effect on the gene expression of key regulators in the absence of telomere dysfunction. These results suggest that TERT non-canonically functions in hematopoietic cell differentiation and survival in vertebrates, independently of its role in telomere homeostasis. The data also provide insights into a non-canonical pathway by which TERT functions to modulate specification of hematopoietic stem/progenitor cells during vertebrate development. (276 words).


Assuntos
Hematopoese/fisiologia , Telomerase/metabolismo , Peixe-Zebra/fisiologia , Animais , Apoptose/fisiologia , Diferenciação Celular/fisiologia , Sobrevivência Celular/fisiologia , Embrião não Mamífero/anatomia & histologia , Embrião não Mamífero/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Homeostase , Humanos , Hibridização In Situ , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Telomerase/genética , Telômero/metabolismo , Telômero/ultraestrutura , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Peixe-Zebra/anatomia & histologia , Peixe-Zebra/embriologia
17.
PLoS Genet ; 4(8): e1000152, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18704191

RESUMO

There is an interesting overlap of function in a wide range of organisms between genes that modulate the stress responses and those that regulate aging phenotypes and, in some cases, lifespan. We have therefore screened mutagenized zebrafish embryos for the altered expression of a stress biomarker, senescence-associated beta-galactosidase (SA-beta-gal) in our current study. We validated the use of embryonic SA-beta-gal production as a screening tool by analyzing a collection of retrovirus-insertional mutants. From a pool of 306 such mutants, we identified 11 candidates that showed higher embryonic SA-beta-gal activity, two of which were selected for further study. One of these mutants is null for a homologue of Drosophila spinster, a gene known to regulate lifespan in flies, whereas the other harbors a mutation in a homologue of the human telomeric repeat binding factor 2 (terf2) gene, which plays roles in telomere protection and telomere-length regulation. Although the homozygous spinster and terf2 mutants are embryonic lethal, heterozygous adult fish are viable and show an accelerated appearance of aging symptoms including lipofuscin accumulation, which is another biomarker, and shorter lifespan. We next used the same SA-beta-gal assay to screen chemically mutagenized zebrafish, each of which was heterozygous for lesions in multiple genes, under the sensitizing conditions of oxidative stress. We obtained eight additional mutants from this screen that, when bred to homozygosity, showed enhanced SA-beta-gal activity even in the absence of stress, and further displayed embryonic neural and muscular degenerative phenotypes. Adult fish that are heterozygous for these mutations also showed the premature expression of aging biomarkers and the accelerated onset of aging phenotypes. Our current strategy of mutant screening for a senescence-associated biomarker in zebrafish embryos may thus prove to be a useful new tool for the genetic dissection of vertebrate stress response and senescence mechanisms.


Assuntos
Envelhecimento/metabolismo , Mutagênese Insercional , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , beta-Galactosidase/metabolismo , Envelhecimento/genética , Animais , Biomarcadores/análise , Expressão Gênica , Humanos , Longevidade , Estresse Oxidativo , Proteína 2 de Ligação a Repetições Teloméricas/genética , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/genética , beta-Galactosidase/genética
18.
J Biol Chem ; 283(44): 29971-82, 2008 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-18678863

RESUMO

Neutral sphingomyelinases (SMases) are involved in the induction of ceramide-mediated proapoptotic signaling under heat stress conditions. Although ceramide is an important mediator of apoptosis, the neutral SMase that is activated under heat stress has not been identified. In this study, we cloned an Mg(2+)-dependent neutral SMase from a zebrafish embryonic cell cDNA library using an Escherichia coli expression-cloning vector. Screening of the clones using an SMase activity assay with C(6)-7-nitro-2-1,3-benzoxadiazol-4-yl-sphingomyelin as the substrate resulted in the isolation of one neutral SMase cDNA clone. This cDNA encoded a polypeptide of 420 amino acids (putative molecular weight: 46,900) containing two predicted transmembrane domains in its C-terminal region. The cloned neutral SMase 1 acted as a mediator of stress-induced apoptosis. Bacterially expressed recombinant neutral SMase 1 hydrolyzed [choline-methyl-(14)C]sphingomyelin optimally at pH 7.5 in the presence of an Mg(2+) ion. In zebrafish embryonic cells, the endogenous SMase enzyme was localized in the microsomal fraction. In FLAG-tagged SMase-overexpressing cells, neutral SMase 1 colocalized with a Golgi marker in a cytochemical analysis. Inactivation of the enzyme by an antisense phosphorothioate oligonucleotide repressed the induction of ceramide generation, caspase-3 activation, and apoptotic cell death by heat stress. Thus, neutral SMase 1 participates in an inducible ceramide-mediating, proapoptotic signaling pathway that operates in heat-induced apoptosis in zebrafish embryonic cells.


Assuntos
Apoptose , Ceramidas/metabolismo , Regulação Enzimológica da Expressão Gênica , Magnésio/química , Esfingomielina Fosfodiesterase/fisiologia , Sequência de Aminoácidos , Animais , Clonagem Molecular , Complexo de Golgi/metabolismo , Temperatura Alta , Concentração de Íons de Hidrogênio , Hidrólise , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Esfingomielina Fosfodiesterase/metabolismo , Peixe-Zebra
19.
Cell ; 133(5): 864-77, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18510930

RESUMO

Evasion of DNA damage-induced cell death, via mutation of the p53 tumor suppressor or overexpression of prosurvival Bcl-2 family proteins, is a key step toward malignant transformation and therapeutic resistance. We report that depletion or acute inhibition of checkpoint kinase 1 (Chk1) is sufficient to restore gamma-radiation-induced apoptosis in p53 mutant zebrafish embryos. Surprisingly, caspase-3 is not activated prior to DNA fragmentation, in contrast to classical intrinsic or extrinsic apoptosis. Rather, an alternative apoptotic program is engaged that cell autonomously requires atm (ataxia telangiectasia mutated), atr (ATM and Rad3-related) and caspase-2, and is not affected by p53 loss or overexpression of bcl-2/xl. Similarly, Chk1 inhibitor-treated human tumor cells hyperactivate ATM, ATR, and caspase-2 after gamma-radiation and trigger a caspase-2-dependent apoptotic program that bypasses p53 deficiency and excess Bcl-2. The evolutionarily conserved "Chk1-suppressed" pathway defines a novel apoptotic process, whose responsiveness to Chk1 inhibitors and insensitivity to p53 and BCL2 alterations have important implications for cancer therapy.


Assuntos
Apoptose , Caspase 2/metabolismo , Dano ao DNA , Proteínas Quinases/metabolismo , Transdução de Sinais , Peixe-Zebra/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Caspase 3/metabolismo , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Embrião não Mamífero/efeitos da radiação , Inibidores Enzimáticos/farmacologia , Raios gama , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
20.
J Clin Invest ; 117(12): 3940-51, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17992259

RESUMO

Statins inhibit HMG-CoA reductase, a key enzyme in cholesterol synthesis, and are widely used to treat hypercholesterolemia. These drugs can lead to a number of side effects in muscle, including muscle fiber breakdown; however, the mechanisms of muscle injury by statins are poorly understood. We report that lovastatin induced the expression of atrogin-1, a key gene involved in skeletal muscle atrophy, in humans with statin myopathy, in zebrafish embryos, and in vitro in murine skeletal muscle cells. In cultured mouse myotubes, atrogin-1 induction following lovastatin treatment was accompanied by distinct morphological changes, largely absent in atrogin-1 null cells. In zebrafish embryos, lovastatin promoted muscle fiber damage, an effect that was closely mimicked by knockdown of zebrafish HMG-CoA reductase. Moreover, atrogin-1 knockdown in zebrafish embryos prevented lovastatin-induced muscle injury. Finally, overexpression of PGC-1alpha, a transcriptional coactivator that induces mitochondrial biogenesis and protects against the development of muscle atrophy, dramatically prevented lovastatin-induced muscle damage and abrogated atrogin-1 induction both in fish and in cultured mouse myotubes. Collectively, our human, animal, and in vitro findings shed light on the molecular mechanism of statin-induced myopathy and suggest that atrogin-1 may be a critical mediator of the muscle damage induced by statins.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Lovastatina/efeitos adversos , Proteínas Musculares/metabolismo , Transtornos Musculares Atróficos/enzimologia , Proteínas Ligases SKP Culina F-Box/metabolismo , Transativadores/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Colesterol/metabolismo , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Hidroximetilglutaril-CoA Redutases/genética , Hidroximetilglutaril-CoA Redutases/metabolismo , Camundongos , Fibras Musculares Esqueléticas/enzimologia , Fibras Musculares Esqueléticas/patologia , Proteínas Musculares/genética , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Transtornos Musculares Atróficos/induzido quimicamente , Transtornos Musculares Atróficos/genética , Transtornos Musculares Atróficos/patologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Proteínas Ligases SKP Culina F-Box/genética , Transativadores/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA