Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
N Engl J Med ; 376(11): 1038-1046, 2017 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-28296613

RESUMEN

We assessed the feasibility of transplanting a sheet of retinal pigment epithelial (RPE) cells differentiated from induced pluripotent stem cells (iPSCs) in a patient with neovascular age-related macular degeneration. The iPSCs were generated from skin fibroblasts obtained from two patients with advanced neovascular age-related macular degeneration and were differentiated into RPE cells. The RPE cells and the iPSCs from which they were derived were subject to extensive testing. A surgery that included the removal of the neovascular membrane and transplantation of the autologous iPSC-derived RPE cell sheet under the retina was performed in one of the patients. At 1 year after surgery, the transplanted sheet remained intact, best corrected visual acuity had not improved or worsened, and cystoid macular edema was present. (Funded by Highway Program for Realization of Regenerative Medicine and others; University Hospital Medical Information Network Clinical Trials Registry [UMIN-CTR] number, UMIN000011929 .).


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Degeneración Macular/terapia , Epitelio Pigmentado de la Retina/citología , Anciano , Técnicas de Cultivo de Célula , Diferenciación Celular , Estudios de Factibilidad , Femenino , Fibroblastos , Humanos , Masculino , Epitelio Pigmentado de la Retina/trasplante , Trasplante Autólogo
2.
Proc Natl Acad Sci U S A ; 110(51): 20569-74, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24259714

RESUMEN

We examined the gene expression and DNA methylation of 49 human induced pluripotent stem cells (hiPSCs) and 10 human embryonic stem cells and found overlapped variations in gene expression and DNA methylation in the two types of human pluripotent stem cell lines. Comparisons of the in vitro neural differentiation of 40 hiPSCs and 10 human embryonic stem cells showed that seven hiPSC clones retained a significant number of undifferentiated cells even after neural differentiation culture and formed teratoma when transplanted into mouse brains. These differentiation-defective hiPSC clones were marked by higher expression levels of several genes, including those expressed from long terminal repeats of specific human endogenous retroviruses. These data demonstrated a subset of hiPSC lines that have aberrant gene expression and defective potential in neural differentiation, which need to be identified and eliminated before applications in regenerative medicine.


Asunto(s)
Diferenciación Celular , Metilación de ADN , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes/metabolismo , Teratoma/metabolismo , Animales , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Xenoinjertos , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Células Jurkat , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Tejido Nervioso/metabolismo , Tejido Nervioso/patología , Células Madre Pluripotentes/patología , Teratoma/patología
3.
Stem Cells ; 31(3): 458-66, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23193063

RESUMEN

The generation of induced pluripotent stem cells (iPSCs) provides the opportunity to use patient-specific somatic cells, which are a valuable source for disease modeling and drug discovery. To promote research involving these cells, it is important to make iPSCs from easily accessible and less invasive tissues, like blood. We have recently reported the efficient generation of human iPSCs from adult fibroblasts using a combination of plasmids encoding OCT3/4, SOX2, KLF4, L-MYC, LIN28, and shRNA for TP53. We herein report a modified protocol enabling efficient iPSC induction from CD34+ cord blood cells and from peripheral blood isolated from healthy donors using these plasmid vectors. The original plasmid mixture could induce iPSCs; however, the efficiency was low. The addition of EBNA1, an essential factor for episomal amplification of the vectors, by an extra plasmid greatly increased the efficiency of iPSC induction, especially when the induction was performed from αßT cells. This improvement enabled the establishment of blood-derived iPSCs from seven healthy donors ranging in age from their 20s to their 60s. This induction method will be useful for the derivation of patient-specific integration-free iPSCs and would also be applicable to the generation of clinical-grade iPSCs in the future.


Asunto(s)
Células Sanguíneas/citología , Técnicas de Cultivo de Célula/métodos , Sangre Fetal/citología , Células Madre Pluripotentes Inducidas/citología , Adulto , Femenino , Vectores Genéticos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Masculino , Persona de Mediana Edad
4.
Stem Cell Res Ther ; 15(1): 73, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38475911

RESUMEN

BACKGROUND: Cell- or tissue-based regenerative therapy is an attractive approach to treat heart failure. A tissue patch that can safely and effectively repair damaged heart muscle would greatly improve outcomes for patients with heart failure. In this study, we conducted a preclinical proof-of-concept analysis of the efficacy and safety of clinical-grade human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) patches. METHODS: A clinical-grade hiPSC line was established using peripheral blood mononuclear cells from a healthy volunteer that was homozygous for human leukocyte antigens. The hiPSCs were differentiated into cardiomyocytes. The obtained hiPSC-CMs were cultured on temperature-responsive culture dishes for patch fabrication. The cellular characteristics, safety, and efficacy of hiPSCs, hiPSC-CMs, and hiPSC-CM patches were analyzed. RESULTS: The hiPSC-CMs expressed cardiomyocyte-specific genes and proteins, and electrophysiological analyses revealed that hiPSC-CMs exhibit similar properties to human primary myocardial cells. In vitro and in vivo safety studies indicated that tumorigenic cells were absent. Moreover, whole-genome and exome sequencing revealed no genomic mutations. General toxicity tests also showed no adverse events posttransplantation. A porcine model of myocardial infarction demonstrated significantly improved cardiac function and angiogenesis in response to cytokine secretion from hiPSC-CM patches. No lethal arrhythmias were observed. CONCLUSIONS: hiPSC-CM patches are promising for future translational research and may have clinical application potential for the treatment of heart failure.


Asunto(s)
Insuficiencia Cardíaca , Células Madre Pluripotentes Inducidas , Humanos , Animales , Porcinos , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Leucocitos Mononucleares , Miocardio , Insuficiencia Cardíaca/terapia
5.
Plant Physiol ; 156(1): 20-8, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21415278

RESUMEN

Full-length cDNA (FLcDNA) libraries consisting of 172,000 clones were constructed from a two-row malting barley cultivar (Hordeum vulgare 'Haruna Nijo') under normal and stressed conditions. After sequencing the clones from both ends and clustering the sequences, a total of 24,783 complete sequences were produced. By removing duplicates between these and publicly available sequences, 22,651 representative sequences were obtained: 17,773 were novel barley FLcDNAs, and 1,699 were barley specific. Highly conserved genes were found in the barley FLcDNA sequences for 721 of 881 rice (Oryza sativa) trait genes with 50% or greater identity. These FLcDNA resources from our Haruna Nijo cDNA libraries and the full-length sequences of representative clones will improve our understanding of the biological functions of genes in barley, which is the cereal crop with the fourth highest production in the world, and will provide a powerful tool for annotating the barley genome sequences that will become available in the near future.


Asunto(s)
ADN Complementario/genética , Genoma de Planta/genética , Hordeum/genética , Secuencia de Bases , ADN Complementario/química , Biblioteca de Genes , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN de Planta/genética , Análisis de Secuencia de ADN
6.
Stem Cells Transl Med ; 10(11): 1530-1543, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34342383

RESUMEN

Mesenchymal stem cells (MSCs) can show trisomy 7; however, the safety of these cells has not been fully investigated. The purposes of this study were to determine the ratio of patients whose synovial MSCs were transplanted clinically, to intensively investigate MSCs with trisomy 7 from a safety perspective, and to follow up the patients for 5 years after transplantation. Synovial MSCs at passage 0 were transplanted into a knee for degenerative meniscus tears in 10 patients, and the patients were checked at 5 years. The synovial MSCs were evaluated at passages 0 to 15 by G-bands and digital karyotyping, and trisomy 7 was found in 3 of 10 patients. In those three patients, 5% to 10% of the synovial MSCs showed trisomy 7. The mRNA expressions of representative oncogenes and genes on chromosome 7 did not differ between MSCs with and without trisomy 7. Whole-genome sequencing and DNA methylation analysis showed similar results for MSCs with and without trisomy 7. Transplantation of human synovial MSCs with trisomy 7 into eight mouse knees did not result in tumor formation under the skin or in the knees after 8 weeks in any mouse, whereas transplanted HT1080 cells formed tumors. In vitro chondrogenic potentials were similar between MSCs with and without trisomy 7. Five-year follow-ups revealed no serious adverse events in all 10 human patients, including 3 who had received MSCs with trisomy 7. Overall, our findings indicated that synovial MSCs with trisomy 7 were comparable with MSCs without trisomy 7 from a safety perspective.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Animales , Estudios de Seguimiento , Humanos , Articulación de la Rodilla , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/patología , Ratones , Membrana Sinovial , Trasplante Autólogo , Trisomía/genética , Trisomía/patología
8.
Nat Commun ; 11(1): 3369, 2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32632153

RESUMEN

Induced pluripotent stem cell (iPSC)-derived dopaminergic (DA) neurons are an expected source for cell-based therapies for Parkinson's disease (PD). The regulatory criteria for the clinical application of these therapies, however, have not been established. Here we show the results of our pre-clinical study, in which we evaluate the safety and efficacy of dopaminergic progenitors (DAPs) derived from a clinical-grade human iPSC line. We confirm the characteristics of DAPs by in vitro analyses. We also verify that the DAP population include no residual undifferentiated iPSCs or early neural stem cells and have no genetic aberration in cancer-related genes. Furthermore, in vivo studies using immunodeficient mice reveal no tumorigenicity or toxicity of the cells. When the DAPs are transplanted into the striatum of 6-OHDA-lesioned rats, the animals show behavioral improvement. Based on these results, we started a clinical trial to treat PD patients in 2018.


Asunto(s)
Neuronas Dopaminérgicas/citología , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/trasplante , Enfermedad de Parkinson/terapia , Trasplante de Células Madre/métodos , Animales , Diferenciación Celular/genética , Línea Celular , Modelos Animales de Enfermedad , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Macaca fascicularis , Masculino , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Evaluación de Resultado en la Atención de Salud/métodos , Evaluación de Resultado en la Atención de Salud/estadística & datos numéricos , Ratas Desnudas , Trasplante Heterólogo
9.
J Clin Med ; 9(7)2020 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-32668747

RESUMEN

Immune attacks are key issues for cell transplantation. To assess the safety and the immune reactions after iPS cells-derived retinal pigment epithelium (iPS-RPE) transplantation, we transplanted HLA homozygote iPS-RPE cells established at an iPS bank in HLA-matched patients with exudative age-related macular degeneration. In addition, local steroids without immunosuppressive medications were administered. We monitored immune rejections by routine ocular examinations as well as by lymphocytes-graft cells immune reaction (LGIR) tests using graft RPE and the patient's blood cells. In all five of the cases that underwent iPS-RPE transplantation, the presence of graft cells was indicated by clumps or an area of increased pigmentation at 6 months, which became stable with no further abnormal growth in the graft during the 1-year observation period. Adverse events observed included corneal erosion, epiretinal membrane, retinal edema due to epiretinal membrane, elevated intraocular pressure, endophthalmitis, and mild immune rejection in the eye. In the one case exhibiting positive LGIR tests along with a slight fluid recurrence, we administrated local steroid therapy that subsequently resolved the suspected immune attacks. Although the cell delivery strategy must be further optimized, the present results suggest that it is possible to achieve stable survival and safety of iPS-RPE cell transplantation for a year.

10.
Sci Rep ; 6: 30013, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27418197

RESUMEN

Cardiovascular complications are the leading cause of death in autosomal dominant polycystic kidney disease (ADPKD), and intracranial aneurysm (ICA) causing subarachnoid hemorrhage is among the most serious complications. The diagnostic and therapeutic strategies for ICAs in ADPKD have not been fully established. We here generated induced pluripotent stem cells (iPSCs) from seven ADPKD patients, including four with ICAs. The vascular cells differentiated from ADPKD-iPSCs showed altered Ca(2+) entry and gene expression profiles compared with those of iPSCs from non-ADPKD subjects. We found that the expression level of a metalloenzyme gene, matrix metalloproteinase (MMP) 1, was specifically elevated in iPSC-derived endothelia from ADPKD patients with ICAs. Furthermore, we confirmed the correlation between the serum MMP1 levels and the development of ICAs in 354 ADPKD patients, indicating that high serum MMP1 levels may be a novel risk factor. These results suggest that cellular disease models with ADPKD-specific iPSCs can be used to study the disease mechanisms and to identify novel disease-related molecules or risk factors.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Aneurisma Intracraneal/patología , Metaloproteinasa 1 de la Matriz/sangre , Riñón Poliquístico Autosómico Dominante/patología , Hemorragia Subaracnoidea/patología , Anciano , Animales , Biomarcadores/sangre , Diferenciación Celular , Células Cultivadas , Metilación de ADN/genética , Femenino , Humanos , Aneurisma Intracraneal/sangre , Masculino , Metaloproteinasa 1 de la Matriz/biosíntesis , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Riñón Poliquístico Autosómico Dominante/mortalidad , Factores de Riesgo , Canales Catiónicos TRPP/genética
11.
Stem Cell Reports ; 4(1): 143-154, 2015 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-25434822

RESUMEN

Duchenne muscular dystrophy (DMD) is a severe muscle-degenerative disease caused by a mutation in the dystrophin gene. Genetic correction of patient-derived induced pluripotent stem cells (iPSCs) by TALENs or CRISPR-Cas9 holds promise for DMD gene therapy; however, the safety of such nuclease treatment must be determined. Using a unique k-mer database, we systematically identified a unique target region that reduces off-target sites. To restore the dystrophin protein, we performed three correction methods (exon skipping, frameshifting, and exon knockin) in DMD-patient-derived iPSCs, and found that exon knockin was the most effective approach. We further investigated the genomic integrity by karyotyping, copy number variation array, and exome sequencing to identify clones with a minimal mutation load. Finally, we differentiated the corrected iPSCs toward skeletal muscle cells and successfully detected the expression of full-length dystrophin protein. These results provide an important framework for developing iPSC-based gene therapy for genetic disorders using programmable nucleases.


Asunto(s)
Sistemas CRISPR-Cas/genética , Distrofina/genética , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Distrofia Muscular de Duchenne/genética , Secuencia de Bases , Variaciones en el Número de Copia de ADN , Distrofina/metabolismo , Exones , Orden Génico , Marcación de Gen , Sitios Genéticos , Terapia Genética , Humanos , Cariotipo , Datos de Secuencia Molecular , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/terapia , Mutagénesis Insercional , Mutación , Sistemas de Lectura , Eliminación de Secuencia
12.
Stem Cell Reports ; 4(4): 561-8, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25801509

RESUMEN

Spinal muscular atrophy (SMA) is a neuromuscular disorder caused by mutations of the survival of motor neuron 1 (SMN1) gene. In the pathogenesis of SMA, pathological changes of the neuromuscular junction (NMJ) precede the motor neuronal loss. Therefore, it is critical to evaluate the NMJ formed by SMA patients' motor neurons (MNs), and to identify drugs that can restore the normal condition. We generated NMJ-like structures using MNs derived from SMA patient-specific induced pluripotent stem cells (iPSCs), and found that the clustering of the acetylcholine receptor (AChR) is significantly impaired. Valproic acid and antisense oligonucleotide treatment ameliorated the AChR clustering defects, leading to an increase in the level of full-length SMN transcripts. Thus, the current in vitro model of AChR clustering using SMA patient-derived iPSCs is useful to dissect the pathophysiological mechanisms underlying the development of SMA, and to evaluate the efficacy of new therapeutic approaches.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Atrofia Muscular Espinal/metabolismo , Unión Neuromuscular/metabolismo , Fenotipo , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Línea Celular , Técnicas de Cocultivo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Ratones , Morfolinos/farmacología , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Atrofia Muscular Espinal/genética , Unión Neuromuscular/efectos de los fármacos , Unión Neuromuscular/patología , Atrofias Musculares Espinales de la Infancia/genética , Atrofias Musculares Espinales de la Infancia/metabolismo , Ácido Valproico/farmacología
13.
Stem Cells Transl Med ; 4(4): 333-8, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25762002

RESUMEN

Fanconi anemia (FA) is a disorder of genomic instability characterized by progressive bone marrow failure (BMF), developmental abnormalities, and an increased susceptibility to cancer. Although various consequences in hematopoietic stem/progenitor cells have been attributed to FA-BMF, the quest to identify the initial pathological event is still ongoing. To address this issue, we established induced pluripotent stem cells (iPSCs) from fibroblasts of six patients with FA and FANCA mutations. An improved reprogramming method yielded iPSC-like colonies from all patients, and iPSC clones were propagated from two patients. Quantitative evaluation of the differentiation ability demonstrated that the differentiation propensity toward the hematopoietic and endothelial lineages is already defective in early hemoangiogenic progenitors. The expression levels of critical transcription factors were significantly downregulated in these progenitors. These data indicate that the hematopoietic consequences in FA patients originate from the early hematopoietic stage and highlight the potential usefulness of iPSC technology for elucidating the pathogenesis of FA-BMF.


Asunto(s)
Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Anemia de Fanconi/genética , Células Madre Hematopoyéticas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Médula Ósea/patología , Diferenciación Celular/genética , Linaje de la Célula/genética , Reprogramación Celular/genética , Anemia de Fanconi/patología , Proteína del Grupo de Complementación A de la Anemia de Fanconi/metabolismo , Fibroblastos/metabolismo , Terapia Genética , Células Madre Hematopoyéticas/patología , Humanos , Células Madre Pluripotentes Inducidas/patología
14.
PLoS One ; 9(4): e94735, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24718089

RESUMEN

It has been shown that DNA demethylation plays a pivotal role in the generation of induced pluripotent stem (iPS) cells. However, the underlying mechanism of this action is still unclear. Previous reports indicated that activation-induced cytidine deaminase (Aid, also known as Aicda) is involved in DNA demethylation in several developmental processes, as well as cell fusion-mediated reprogramming. Based on these reports, we hypothesized that Aid may be involved in the DNA demethylation that occurs during the generation of iPS cells. In this study, we examined the function of Aid in iPS cell generation using Aid knockout (Aid⁻/⁻) mice expressing a GFP reporter under the control of a pluripotent stem cell marker, Nanog. By introducing Oct3/4, Sox2, Klf4 and c-Myc, Nanog-GFP-positive iPS cells could be generated from the fibroblasts and primary B cells of Aid⁻/⁻ mice. Their induction efficiency was similar to that of wild-type (Aid⁺/⁺) iPS cells. The Aid⁻/⁻ iPS cells showed normal proliferation and gave rise to chimeras, indicating their capacity for self-renewal and pluripotency. A comprehensive DNA methylation analysis showed only a few differences between Aid⁺/⁺ and Aid⁻/⁻ iPS cells. These data suggest that Aid does not have crucial functions in DNA demethylation during iPS cell generation.


Asunto(s)
Citidina Desaminasa/deficiencia , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Citidina Desaminasa/metabolismo , Metilación de ADN/genética , Embrión de Mamíferos/citología , Fibroblastos/citología , Fibroblastos/metabolismo , Factor 4 Similar a Kruppel , Ratones Endogámicos C57BL
15.
PLoS One ; 8(1): e53771, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23382851

RESUMEN

BACKGROUND: For regenerative therapy using induced pluripotent stem cell (iPSC) technology, cell type of origin to be reprogrammed should be chosen based on accessibility and reprogramming efficiency. Some studies report that iPSCs exhibited a preference for differentiation into their original cell lineages, while others did not. Therefore, the type of cell which is most appropriate as a source for iPSCs needs to be clarified. METHODOLOGY/PRINCIPAL FINDINGS: Genetically matched human iPSCs from different origins were generated using bone marrow stromal cells (BMSCs) and dermal fibroblasts (DFs) of the same donor, and global gene expression profile, DNA methylation status, and differentiation properties into the chondrogenic and osteogenic lineage of each clone were analyzed. Although genome-wide profiling of DNA methylation suggested tissue memory in iPSCs, genes expressed differentially in BMSCs and DFs were equally silenced in our bona fide iPSCs. After cell-autonomous and induced differentiation, each iPSC clone exhibited various differentiation properties, which did not correlate with cell-of-origin. CONCLUSIONS/SIGNIFICANCE: The reprogramming process may remove the difference between DFs and BMSCs at least for chondrogenic and osteogenic differentiation. Qualified and genetically matched human iPSC clone sets established in this study are valuable resources for further basic study of clonal differences.


Asunto(s)
Desarrollo Óseo/genética , Cartílago , Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Cartílago/citología , Cartílago/crecimiento & desarrollo , Linaje de la Célula , Evolución Clonal/genética , Metilación de ADN/genética , Fibroblastos/citología , Regulación del Desarrollo de la Expresión Génica , Humanos , Células Madre Mesenquimatosas/citología
16.
Front Plant Sci ; 3: 5, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22645565

RESUMEN

In support of the international effort to obtain a reference sequence of the bread wheat genome and to provide plant communities dealing with large and complex genomes with a versatile, easy-to-use online automated tool for annotation, we have developed the TriAnnot pipeline. Its modular architecture allows for the annotation and masking of transposable elements, the structural, and functional annotation of protein-coding genes with an evidence-based quality indexing, and the identification of conserved non-coding sequences and molecular markers. The TriAnnot pipeline is parallelized on a 712 CPU computing cluster that can run a 1-Gb sequence annotation in less than 5 days. It is accessible through a web interface for small scale analyses or through a server for large scale annotations. The performance of TriAnnot was evaluated in terms of sensitivity, specificity, and general fitness using curated reference sequence sets from rice and wheat. In less than 8 h, TriAnnot was able to predict more than 83% of the 3,748 CDS from rice chromosome 1 with a fitness of 67.4%. On a set of 12 reference Mb-sized contigs from wheat chromosome 3B, TriAnnot predicted and annotated 93.3% of the genes among which 54% were perfectly identified in accordance with the reference annotation. It also allowed the curation of 12 genes based on new biological evidences, increasing the percentage of perfect gene prediction to 63%. TriAnnot systematically showed a higher fitness than other annotation pipelines that are not improved for wheat. As it is easily adaptable to the annotation of other plant genomes, TriAnnot should become a useful resource for the annotation of large and complex genomes in the future.

17.
DNA Res ; 17(5): 271-9, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20668003

RESUMEN

We present an annotation pipeline that accurately predicts exon-intron structures and protein-coding sequences (CDSs) on the basis of full-length cDNAs (FLcDNAs). This annotation pipeline was used to identify genes in 10 plant genomes. In particular, we show that interspecies mapping of FLcDNAs to genomes is of great value in fully utilizing FLcDNA resources whose availability is limited to several species. Because low sequence conservation at 5'- and 3'-ends of FLcDNAs between different species tends to result in truncated CDSs, we developed an improved algorithm to identify complete CDSs by the extension of both ends of truncated CDSs. Interspecies mapping of 71 801 monocot FLcDNAs to the Oryza sativa genome led to the detection of 22 142 protein-coding regions. Moreover, in comparing two mapping programs and three ab initio prediction programs, we found that our pipeline was more capable of identifying complete CDSs. As demonstrated by monocot interspecies mapping, in which nucleotide identity between FLcDNAs and the genome was ∼80%, the resultant inferred CDSs were sufficiently accurate. Finally, we applied both inter- and intraspecies mapping to 10 monocot and dicot genomes and identified genes in 210 551 loci. Interspecies mapping of FLcDNAs is expected to effectively predict genes and CDSs in newly sequenced genomes.


Asunto(s)
Análisis Citogenético , ADN de Plantas/genética , Genes de Plantas , Modelos Genéticos , Anotación de Secuencia Molecular , Plantas/genética , Secuencia de Aminoácidos , Secuencia de Bases , Biología Computacional , Cotiledón/anatomía & histología , ADN Complementario , Bases de Datos Genéticas , Exones , Predicción , Biblioteca de Genes , Variación Genética , Genoma de Planta , Intrones , Sistemas de Lectura Abierta , Oryza/genética , Análisis de Secuencia de ADN , Especificidad de la Especie
18.
Biol Pharm Bull ; 31(9): 1786-9, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18758077

RESUMEN

Juniperonic acid (Delta-5c,11c,14c,17c-20:4, JA) is a polymethylene-interrupted (PMI) fatty acid that occurs in Biota orientalis. In this study, we found that JA has an antiproliferative activity. Swiss 3T3 cells were preloaded with fatty acids before stimulation with bombesin, a mitogenic neuropeptide, and proliferation of the cells was assessed by [(3)H]thymidine incorporation. Preloading of linoleic acid (Delta-9c,12c-18:2) significantly enhanced bombesin-induced proliferation. In contrast, preloading of eicosapentaenoic acid (Delta-5c,8c,11c,14c,17c-20:5, EPA) suppressed proliferation. Likewise, cells preloaded with JA showed a significantly curtailed response to bombesin. The antiproliferative potency of JA was equivalent to that of EPA. Sciadonic acid (Delta-5c,11c,14c-20:3), an omega-6 analogue of JA did not show antiproliferative activity, suggesting the importance of the omega-3 double bond rather than the PMI structure. The EPA-like activity of JA may be involved in the pharmaceutical activity of biota seeds, a psychoactive Chinese traditional medicine.


Asunto(s)
Bombesina/antagonistas & inhibidores , Bombesina/farmacología , Proliferación Celular/efectos de los fármacos , Ácidos Grasos Insaturados/farmacología , Animales , Ácidos Araquidónicos/farmacología , Ácido Eicosapentaenoico/metabolismo , Ácidos Grasos/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , Fosfolípidos/metabolismo , Semillas/química , Células 3T3 Swiss , Thuja
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA