Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cell ; 174(3): 636-648.e18, 2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-30017246

RESUMEN

The ex vivo generation of platelets from human-induced pluripotent cells (hiPSCs) is expected to compensate donor-dependent transfusion systems. However, manufacturing the clinically required number of platelets remains unachieved due to the low platelet release from hiPSC-derived megakaryocytes (hiPSC-MKs). Here, we report turbulence as a physical regulator in thrombopoiesis in vivo and its application to turbulence-controllable bioreactors. The identification of turbulent energy as a determinant parameter allowed scale-up to 8 L for the generation of 100 billion-order platelets from hiPSC-MKs, which satisfies clinical requirements. Turbulent flow promoted the release from megakaryocytes of IGFBP2, MIF, and Nardilysin to facilitate platelet shedding. hiPSC-platelets showed properties of bona fide human platelets, including circulation and hemostasis capacities upon transfusion in two animal models. This study provides a concept in which a coordinated physico-chemical mechanism promotes platelet biogenesis and an innovative strategy for ex vivo platelet manufacturing.


Asunto(s)
Plaquetas/metabolismo , Técnicas de Cultivo de Célula/métodos , Trombopoyesis/fisiología , Reactores Biológicos , Técnicas de Cultivo de Célula/instrumentación , Humanos , Hidrodinámica , Células Madre Pluripotentes Inducidas/metabolismo , Megacariocitos/metabolismo , Megacariocitos/fisiología
2.
Nucleic Acids Res ; 47(D1): D1218-D1224, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30295851

RESUMEN

Rapid progress is being made in mass spectrometry (MS)-based proteomics, yielding an increasing number of larger datasets with higher quality and higher throughput. To integrate proteomics datasets generated from various projects and institutions, we launched a project named jPOST (Japan ProteOme STandard Repository/Database, https://jpostdb.org/) in 2015. Its proteomics data repository, jPOSTrepo, began operations in 2016 and has accepted more than 10 TB of MS-based proteomics datasets in the past two years. In addition, we have developed a new proteomics database named jPOSTdb in which the published raw datasets in jPOSTrepo are reanalyzed using standardized protocol. jPOSTdb provides viewers showing the frequency of detected post-translational modifications, the co-occurrence of phosphorylation sites on a peptide and peptide sharing among proteoforms. jPOSTdb also provides basic statistical analysis tools to compare proteomics datasets.


Asunto(s)
Biología Computacional/métodos , Bases de Datos de Proteínas , Proteoma/metabolismo , Proteómica/métodos , Manejo de Datos/métodos , Humanos , Almacenamiento y Recuperación de la Información/métodos , Internet , Japón , Espectrometría de Masas/métodos , Procesamiento Proteico-Postraduccional , Interfaz Usuario-Computador
3.
Proc Natl Acad Sci U S A ; 114(2): 340-345, 2017 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-28003464

RESUMEN

Novel APOBEC1 target 1 (Nat1) (also known as "p97," "Dap5," and "Eif4g2") is a ubiquitously expressed cytoplasmic protein that is homologous to the C-terminal two thirds of eukaryotic translation initiation factor 4G (Eif4g1). We previously showed that Nat1-null mouse embryonic stem cells (mES cells) are resistant to differentiation. In the current study, we found that NAT1 and eIF4G1 share many binding proteins, such as the eukaryotic translation initiation factors eIF3 and eIF4A and ribosomal proteins. However, NAT1 did not bind to eIF4E or poly(A)-binding proteins, which are critical for cap-dependent translation initiation. In contrast, compared with eIF4G1, NAT1 preferentially interacted with eIF2, fragile X mental retardation proteins (FMR), and related proteins and especially with members of the proline-rich and coiled-coil-containing protein 2 (PRRC2) family. We also found that Nat1-null mES cells possess a transcriptional profile similar, although not identical, to the ground state, which is established in wild-type mES cells when treated with inhibitors of the ERK and glycogen synthase kinase 3 (GSK3) signaling pathways. In Nat1-null mES cells, the ERK pathway is suppressed even without inhibitors. Ribosome profiling revealed that translation of mitogen-activated protein kinase kinase kinase 3 (Map3k3) and son of sevenless homolog 1 (Sos1) is suppressed in the absence of Nat1 Forced expression of Map3k3 induced differentiation of Nat1-null mES cells. These data collectively show that Nat1 is involved in the translation of proteins that are required for cell differentiation.


Asunto(s)
Arilamina N-Acetiltransferasa/metabolismo , Diferenciación Celular/fisiología , Isoenzimas/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Embrionarias de Ratones/fisiología , Biosíntesis de Proteínas/fisiología , Animales , Línea Celular , Células Cultivadas , Factor 4G Eucariótico de Iniciación/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , MAP Quinasa Quinasa Quinasa 3/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Unión Proteica/fisiología , Ribosomas/metabolismo , Proteína SOS1/metabolismo , Transducción de Señal/fisiología , Transcripción Genética/fisiología
4.
J Proteome Res ; 18(6): 2535-2544, 2019 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-31039306

RESUMEN

Rapid progress in mass spectrometry (MS) has made comprehensive analyses of the proteome possible, but accurate quantification remains challenging. Isobaric tags for relative and absolute quantification (iTRAQ) is widely used as a tool to quantify proteins expressed in different cell types and various cellular conditions. The quantification precision of iTRAQ is quite high, but the accuracy dramatically decreases in the presence of interference peptides that are coeluted and coisolated with the target peptide. Here, we developed "removal of interference mixture MS/MS spectra (RiMS)" to improve the quantification accuracy of isobaric tag approaches. The presence of spectrum interference is judged by examining the overlap in the elution time of all scanned precursor ions. Removal of this interference decreased protein identification (11% loss) but improved quantification accuracy. Further, RiMS does not require any specialized equipment, such as MS3 instruments or an additional ion separation mode. Finally, we demonstrated that RiMS can be used to quantitatively compare human-induced pluripotent stem cells and human dermal fibroblasts, as it revealed differential protein expressions that reflect the biological characteristics of the cells.


Asunto(s)
Péptidos/genética , Proteoma/genética , Proteómica/métodos , Espectrometría de Masas en Tándem/métodos , Fibroblastos/química , Fibroblastos/metabolismo , Regulación de la Expresión Génica/genética , Humanos , Péptidos/química , Péptidos/aislamiento & purificación , Piel/química , Piel/metabolismo , Coloración y Etiquetado
5.
Genes Cells ; 22(10): 918-928, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28776863

RESUMEN

The molecular mechanisms of cell reprogramming and differentiation involve various signaling factors. Small molecule compounds have been identified to artificially influence these factors through interacting cellular proteins. Although such small molecule compounds are useful to enhance reprogramming and differentiation and to show the mechanisms that underlie these events, the screening usually requires a large number of compounds to identify only a very small number of hits (e.g., one hit among several tens of thousands of compounds). Here, we show a proof of concept that xenospecific gene products can affect the efficiency of cell reprogramming to pluripotency. Thirty genes specific for the bacterium Wolbachia pipientis were forcibly expressed individually along with reprogramming factors (Oct4, Sox2, Klf4 and c-Myc) that can generate induced pluripotent stem cells in mammalian cells, and eight were found to affect the reprogramming efficiency either positively or negatively (hit rate 26.7%). Mechanistic analysis suggested one of these proteins interacted with cytoskeleton to promote reprogramming. Our results raise the possibility that xenospecific gene products provide an alternative way to study the regulatory mechanism of cell identity.


Asunto(s)
Reprogramación Celular/genética , Genes Bacterianos , Células-Madre Neurales/citología , Células Madre Pluripotentes/citología , Animales , Línea Celular , Citoesqueleto/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Células-Madre Neurales/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Wolbachia/genética
6.
Cell Stem Cell ; 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38996472

RESUMEN

Recent advances have made modeling human small intestines in vitro possible, but it remains a challenge to recapitulate fully their structural and functional characteristics. We suspected interstitial flow within the intestine, powered by circulating blood plasma during embryonic organogenesis, to be a vital factor. We aimed to construct an in vivo-like multilayered small intestinal tissue by incorporating interstitial flow into the system and, in turn, developed the micro-small intestine system by differentiating definitive endoderm and mesoderm cells from human pluripotent stem cells simultaneously on a microfluidic device capable of replicating interstitial flow. This approach enhanced cell maturation and led to the development of a three-dimensional small intestine-like tissue with villi-like epithelium and an aligned mesenchymal layer. Our micro-small intestine system not only overcomes the limitations of conventional intestine models but also offers a unique opportunity to gain insights into the detailed mechanisms underlying intestinal tissue development.

7.
J Proteome Res ; 12(1): 214-21, 2013 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-23210603

RESUMEN

Proteome analyses of human induced pluripotent stem cells (iPSC) were carried out on a liquid chromatography-tandem mass spectrometry system using meter-scale monolithic silica-C18 capillary columns without prefractionation. Tryptic peptides from five different iPSC lysates and three different fibroblast lysates (4 µg each) were directly injected onto a 200 cm long, 100 µm i.d. monolithic silica-C18 column and an 8-h gradient was applied at 500 nL/min at less than 20 MPa. We identified 98,977 nonredundant tryptic peptides from 9510 proteins (corresponding to 8712 genes), including low-abundance protein groups (such as 329 protein kinases) from triplicate measurements within 10 days. The obtained proteome profiles of the eight cell lysates were categorized into two groups, iPSC and fibroblast, by hierarchical cluster analysis. Further quantitative analysis based on an exponentially modified protein abundance index approach combined with UniProt keyword enrichment analysis revealed that the iPSC group contains more "transcription regulation"-related proteins, while the fibroblast group contained more "transport"-related proteins. Our results indicate that this simplified one-shot proteomics approach with long monolithic columns is advantageous for rapid, deep, sensitive, and reproducible proteome analysis.


Asunto(s)
Cromatografía Liquida , Células Madre Pluripotentes Inducidas/metabolismo , Proteómica , Espectrometría de Masas en Tándem , Línea Celular , Expresión Génica , Humanos , Péptidos/genética , Péptidos/aislamiento & purificación , Péptidos/metabolismo , Dióxido de Silicio/química , Tripsina/genética , Tripsina/aislamiento & purificación , Tripsina/metabolismo
8.
Cell Rep Methods ; 2(2): 100155, 2022 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-35474962

RESUMEN

Xeno-free culture systems have expanded the clinical and industrial application of human pluripotent stem cells (PSCs). However, reproducibility issues, often arising from variability during passaging steps, remain. Here, we describe an improved method for the subculture of human PSCs. The revised method significantly enhances the viability of human PSCs by lowering DNA damage and apoptosis, resulting in more efficient and reproducible downstream applications such as gene editing and directed differentiation. Furthermore, the method does not alter PSC characteristics after long-term culture and attenuates the growth advantage of abnormal subpopulations. This robust passaging method minimizes experimental error and reduces the rate of PSCs failing quality control of human PSC research and application.


Asunto(s)
Células Madre Pluripotentes , Humanos , Reproducibilidad de los Resultados , Diferenciación Celular/genética
9.
iScience ; 25(7): 104516, 2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35754732

RESUMEN

Cellular global translation is often measured using ribosome profiling or quantitative mass spectrometry, but these methods do not provide direct information at the level of elongating nascent polypeptide chains (NPCs) and associated co-translational events. Here, we describe pSNAP, a method for proteome-wide profiling of NPCs by affinity enrichment of puromycin- and stable isotope-labeled polypeptides. pSNAP does not require ribosome purification and/or chemical labeling, and captures bona fide NPCs that characteristically exhibit protein N-terminus-biased positions. We applied pSNAP to evaluate the effect of silmitasertib, a potential molecular therapy for cancer, and revealed acute translational repression through casein kinase II and mTOR pathways. We also characterized modifications on NPCs and demonstrated that the combination of different types of modifications, such as acetylation and phosphorylation in the N-terminal region of histone H1.5, can modulate interactions with ribosome-associated factors. Thus, pSNAP provides a framework for dissecting co-translational regulations on a proteome-wide scale.

10.
iScience ; 25(5): 104289, 2022 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-35573189

RESUMEN

The effects of transcription factors on the maintenance and differentiation of human-induced or embryonic pluripotent stem cells (iPSCs/ESCs) have been well studied. However, the importance of posttranscriptional regulatory mechanisms, which cause the quantitative dissociation of mRNA and protein expression, has not been explored in detail. Here, by combining transcriptome and proteome profiling, we identified 228 posttranscriptionally regulated genes with strict upregulation of the protein level in iPSCs/ESCs. Among them, we found 84 genes were vital for the survival of iPSCs and HDFs, including 20 genes that were specifically necessary for iPSC survival. These 20 proteins were upregulated only in iPSCs/ESCs and not in differentiated cells derived from the three germ layers. Although there are still unknown mechanisms that downregulate protein levels in HDFs, these results reveal that posttranscriptionally regulated genes have a crucial role in iPSC survival.

11.
Sci Rep ; 11(1): 24254, 2021 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-34930932

RESUMEN

Human induced pluripotent stem cells (hiPSCs) can differentiate into cells of the three germ layers and are promising cell sources for regenerative medicine therapies. However, current protocols generate hiPSCs with low efficiency, and the generated iPSCs have variable differentiation capacity among different clones. Our previous study reported that MYC proteins (c-MYC and MYCL) are essential for reprogramming and germline transmission but that MYCL can generate hiPSC colonies more efficiently than c-MYC. The molecular underpinnings for the different reprogramming efficiencies between c-MYC and MYCL, however, are unknown. In this study, we found that MYC Box 0 (MB0) and MB2, two functional domains conserved in the MYC protein family, contribute to the phenotypic differences and promote hiPSC generation in MYCL-induced reprogramming. Proteome analyses suggested that in MYCL-induced reprogramming, cell adhesion-related cytoskeletal proteins are regulated by the MB0 domain, while the MB2 domain regulates RNA processes. These findings provide a molecular explanation for why MYCL has higher reprogramming efficiency than c-MYC.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Proteínas Proto-Oncogénicas c-myc/fisiología , Adhesión Celular , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/genética , Células Cultivadas , Reprogramación Celular , Citometría de Flujo , Eliminación de Gen , Humanos , Mutación , Fenotipo , Dominios Proteicos , Proteoma , Proteómica , Proteínas Recombinantes/química
12.
Nat Commun ; 12(1): 5012, 2021 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-34408142

RESUMEN

Tendon self-renewal is a rare occurrence because of the poor vascularization of this tissue; therefore, reconstructive surgery using autologous tendon is often performed in severe injury cases. However, the post-surgery re-injury rate is relatively high, and the collection of autologous tendons leads to muscle weakness, resulting in prolonged rehabilitation. Here, we introduce an induced pluripotent stem cell (iPSC)-based technology to develop a therapeutic option for tendon injury. First, we derived tenocytes from human iPSCs by recapitulating the normal progression of step-wise narrowing fate decisions in vertebrate embryos. We used single-cell RNA sequencing to analyze the developmental trajectory of iPSC-derived tenocytes. We demonstrated that iPSC-tenocyte grafting contributed to motor function recovery after Achilles tendon injury in rats via engraftment and paracrine effects. The biomechanical strength of regenerated tendons was comparable to that of healthy tendons. We suggest that iPSC-tenocytes will provide a therapeutic option for tendon injury.


Asunto(s)
Tendón Calcáneo/lesiones , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/trasplante , Traumatismos de los Tendones/terapia , Tenocitos/citología , Tenocitos/trasplante , Tendón Calcáneo/citología , Tendón Calcáneo/fisiopatología , Animales , Autorrenovación de las Células , Tratamiento Basado en Trasplante de Células y Tejidos , Células Cultivadas , Humanos , Masculino , Ratas , Ratas Endogámicas F344 , Recuperación de la Función , Traumatismos de los Tendones/fisiopatología
13.
Anal Chem ; 82(7): 2616-20, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20222674

RESUMEN

We successfully identified the proteome expressed in Escherichia coli (E. coli) cells on a microarray scale using one-dimensional capillary liquid chromatography-tandem mass spectrometry (LC-MS/MS) with a 350 cm long, 100 microm i. d., monolithic silica-C(18) capillary column. E. coli tryptic digest (4 microg) was injected onto the column, and a 41 h gradient was applied with a flow rate of 500 nL/min at less than 20 MPa. In total, 22,196 nonredundant tryptic peptides from 2602 proteins, including 830 membrane proteins, were identified from the E. coli cells (triplicate analysis), in which an equivalent number of genes was detected by transcriptome analysis. Approximately a 5-fold larger peak response on average was obtained in this system, compared with that obtained by conventional capillary LC-MS/MS analysis with a 15 cm long, 3 microm diameter C(18) silica particle-packed column. The higher response suggests that the influence of ionization suppression was drastically reduced by the high-efficiency separation on the long monolithic silica column coupled with the shallow gradient. Because this high-resolution system does not require any additional separation prior to LC-MS/MS, this "one-shot" proteomics approach can simplify the workflow of shotgun proteomics and minimize the sample amount, as well as reduce the total analysis time, despite the use of prolonged shallow gradient elution.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Proteínas de Escherichia coli/análisis , Escherichia coli/metabolismo , Espectrometría de Masas en Tándem/métodos , Proteínas de Escherichia coli/aislamiento & purificación , Proteoma , Tripsina/metabolismo , Flujo de Trabajo
14.
Cell Chem Biol ; 27(12): 1561-1572.e7, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33125912

RESUMEN

The proliferation of human pancreatic progenitor cells (PPCs) is critical for developing cell therapies for diabetes. Here, using transcriptome analysis combined with small interfering RNA (siRNA) screening, we revealed that WNT7B is a downstream growth factor of AT7867, a compound known to promote the proliferation of PPCs generated from human pluripotent stem cells. Feeder cell lines stably expressing mouse Wnt7a or Wnt7b, but not other Wnts, enhanced PPC proliferation in the absence of AT7867. Importantly, Wnt7a/b ligands did not activate the canonical Wnt pathway, and PPC proliferation depended on the non-canonical Wnt/PKC pathway. A comparison of the phosphoproteome in response to AT7867 or a newly synthesized AT7867 derivative uncovered the function of YY1 as a transcriptional regulator of WNT7B. Overall, our data highlight unknown roles of non-canonical WNT7B/PKC signaling and YY1 in human PPC proliferation and will contribute to the stable supply of a cell source for pancreatic disease modeling and therapeutic applications.


Asunto(s)
Páncreas/citología , Células Madre Pluripotentes/citología , Transducción de Señal , Proteínas Wnt/metabolismo , Factor de Transcripción YY1/metabolismo , Animales , Línea Celular , Proliferación Celular , Células Nutrientes/citología , Humanos , Ratones , Proteína Quinasa C/metabolismo
15.
Cell Rep ; 31(9): 107715, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32492424

RESUMEN

Previous studies have suggested that the loss of the translation initiation factor eIF4G1 homolog NAT1 induces excessive self-renewability of naive pluripotent stem cells (PSCs); yet the role of NAT1 in the self-renewal and differentiation of primed PSCs is still unclear. Here, we generate a conditional knockout of NAT1 in primed PSCs and use the cells for the functional analyses of NAT1. Our results show that NAT1 is required for the self-renewal and neural differentiation of primed PSCs. In contrast, NAT1 deficiency in naive pluripotency attenuates the differentiation to all cell types. We also find that NAT1 is involved in efficient protein expression of an RNA uridyltransferase, TUT7. TUT7 is involved in the neural differentiation of primed PSCs via the regulation of human endogenous retrovirus accumulation. These data demonstrate the essential roles of NAT1 and TUT7 in the precise transition of stem cell fate.


Asunto(s)
Diferenciación Celular , Retrovirus Endógenos/metabolismo , Neuronas/citología , Células Madre Pluripotentes/citología , ARN Viral/metabolismo , Animales , Arilamina N-Acetiltransferasa/deficiencia , Arilamina N-Acetiltransferasa/genética , Arilamina N-Acetiltransferasa/metabolismo , Línea Celular , Linaje de la Célula , Autorrenovación de las Células , Retrovirus Endógenos/genética , Edición Génica , Humanos , Isoenzimas/deficiencia , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Neuronas/metabolismo , Iniciación de la Cadena Peptídica Traduccional , Células Madre Pluripotentes/metabolismo , Interferencia de ARN , ARN Nucleotidiltransferasas/genética , ARN Nucleotidiltransferasas/metabolismo , ARN Interferente Pequeño/metabolismo , ARN Viral/antagonistas & inhibidores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
16.
Nat Commun ; 11(1): 1334, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-32170079

RESUMEN

Prolonged expression of the CRISPR-Cas9 nuclease and gRNA from viral vectors may cause off-target mutagenesis and immunogenicity. Thus, a transient delivery system is needed for therapeutic genome editing applications. Here, we develop an extracellular nanovesicle-based ribonucleoprotein delivery system named NanoMEDIC by utilizing two distinct homing mechanisms. Chemical induced dimerization recruits Cas9 protein into extracellular nanovesicles, and then a viral RNA packaging signal and two self-cleaving riboswitches tether and release sgRNA into nanovesicles. We demonstrate efficient genome editing in various hard-to-transfect cell types, including human induced pluripotent stem (iPS) cells, neurons, and myoblasts. NanoMEDIC also achieves over 90% exon skipping efficiencies in skeletal muscle cells derived from Duchenne muscular dystrophy (DMD) patient iPS cells. Finally, single intramuscular injection of NanoMEDIC induces permanent genomic exon skipping in a luciferase reporter mouse and in mdx mice, indicating its utility for in vivo genome editing therapy of DMD and beyond.


Asunto(s)
Proteína 9 Asociada a CRISPR/genética , Sistemas CRISPR-Cas , Exones/genética , Vesículas Extracelulares/metabolismo , Nanopartículas/química , ARN Guía de Kinetoplastida/metabolismo , Secuencia de Bases , Supervivencia Celular , Dimerización , Edición Génica , Vectores Genéticos/metabolismo , Células HEK293 , Proteasa del VIH/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ligandos , Luciferasas/metabolismo , Empalme del ARN/genética , ARN Catalítico/metabolismo , Ribonucleoproteínas/metabolismo , Donantes de Tejidos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo
17.
Infect Immun ; 77(11): 5130-8, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19737900

RESUMEN

Porphyromonas gingivalis forms communities with antecedent oral biofilm constituent streptococci. P. gingivalis major fimbriae bind to glyceraldehyde-3-phosphate dehydrogenase (GAPDH) present on the streptococcal surface, and this interaction plays an important role in P. gingivalis colonization. This study identified the binding domain of Streptococcus oralis GAPDH for P. gingivalis fimbriae. S. oralis recombinant GAPDH (rGAPDH) was digested with lysyl endopeptidase. Cleaved fragments of rGAPDH were applied to a reverse-phase high-pressure liquid chromatograph equipped with a C18 column. Each peak was collected; the binding activity toward P. gingivalis recombinant fimbrillin (rFimA) was analyzed with a biomolecular interaction analysis system. The fragment displaying the strongest binding activity was further digested with various proteinases, after which the binding activity of each fragment was measured. The amino acid sequence of each fragment was determined by direct sequencing, mass spectrometric analysis, and amino acid analysis. Amino acid residues 166 to 183 of S. oralis GAPDH exhibited the strongest binding activity toward rFimA; confocal laser scanning microscopy revealed that the synthetic peptide corresponding to amino acid residues 166 to 183 of S. oralis GAPDH (pep166-183, DNFGVVEGLMTTIHAYTG) inhibits S. oralis-P. gingivalis biofilm formation in a dose-dependent manner. Moreover, pep166-183 inhibited interbacterial biofilm formation by several oral streptococci and P. gingivalis strains with different types of FimA. These results indicate that the binding domain of S. oralis GAPDH for P. gingivalis fimbriae exists within the region encompassing amino acid residues 166 to 183 of GAPDH and that pep166-183 may be a potent inhibitor of P. gingivalis colonization in the oral cavity.


Asunto(s)
Proteínas Bacterianas/metabolismo , Biopelículas , Proteínas Fimbrias/metabolismo , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Porphyromonas gingivalis/fisiología , Streptococcus oralis/fisiología , Secuencia de Aminoácidos , Cromatografía Líquida de Alta Presión , Proteínas Fimbrias/química , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/química , Microscopía Confocal , Datos de Secuencia Molecular , Unión Proteica
18.
Stem Cell Reports ; 8(3): 634-647, 2017 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-28238797

RESUMEN

The blood-brain barrier (BBB) is composed of four cell populations, brain endothelial cells (BECs), pericytes, neurons, and astrocytes. Its role is to precisely regulate the microenvironment of the brain through selective substance crossing. Here we generated an in vitro model of the BBB by differentiating human induced pluripotent stem cells (hiPSCs) into all four populations. When the four hiPSC-derived populations were co-cultured, endothelial cells (ECs) were endowed with features consistent with BECs, including a high expression of nutrient transporters (CAT3, MFSD2A) and efflux transporters (ABCA1, BCRP, PGP, MRP5), and strong barrier function based on tight junctions. Neuron-derived Dll1, which activates Notch signaling in ECs, was essential for the BEC specification. We performed in vitro BBB permeability tests and assessed ten clinical drugs by nanoLC-MS/MS, finding a good correlation with the BBB permeability reported in previous cases. This technology should be useful for research on human BBB physiology, pathology, and drug development.


Asunto(s)
Astrocitos/metabolismo , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Células Madre Pluripotentes Inducidas/citología , Neuronas/metabolismo , Pericitos/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Astrocitos/citología , Biomarcadores , Permeabilidad Capilar , Diferenciación Celular , Línea Celular , Células Endoteliales/citología , Humanos , Neuronas/citología , Pericitos/citología
19.
Cell Chem Biol ; 24(6): 685-694.e4, 2017 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-28529132

RESUMEN

The incomplete differentiation of human induced pluripotent stem cells (iPSCs) poses a serious safety risk owing to their potential tumorigenicity, hindering their clinical application. Here, we explored the potential of phospho-D-peptides as novel iPSC-eliminating agents. Alkaline phosphatases overexpressed on iPSCs dephosphorylate phospho-D-peptides into hydrophobic peptides that aggregate and induce cell death. We isolated a peptide candidate, D-3, that selectively and rapidly induced toxicity in iPSCs within 1 hr but had little influence on various non-iPSCs, including primary hepatocytes and iPSC-derived cardiomyocytes. Two hours of D-3 treatment efficiently eliminated iPSCs from both single cultures and co-cultures spiked with increasing ratios of iPSCs. In addition, D-3 prevented residual iPSC-induced teratoma formation in a mouse tumorigenicity assay. These results suggest the enormous potential of D-3 as a low-cost and effective anti-iPSC agent for both laboratory use and for the safe clinical application of iPSC-derived cells in regenerative medicine.


Asunto(s)
Fosfatasa Alcalina/metabolismo , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Fosfopéptidos/química , Fosfopéptidos/farmacología , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células HeLa , Humanos , Células Madre Pluripotentes Inducidas/citología , Fosfopéptidos/síntesis química , Seguridad
20.
BMC Syst Biol ; 9: 75, 2015 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-26542228

RESUMEN

BACKGROUND: Cellular function and diversity are orchestrated by complex interactions of fundamental biomolecules including DNA, RNA and proteins. Technological advances in genomics, epigenomics, transcriptomics and proteomics have enabled massively parallel and unbiased measurements. Such high-throughput technologies have been extensively used to carry out broad, unbiased studies, particularly in the context of human diseases. Nevertheless, a unified analysis of the genome, epigenome, transcriptome and proteome of a single human cell type to obtain a coherent view of the complex interplay between various biomolecules has not yet been undertaken. Here, we report the first multi-omic analysis of human primary naïve CD4+ T cells isolated from a single individual. RESULTS: Integrating multi-omics datasets allowed us to investigate genome-wide methylation and its effect on mRNA/protein expression patterns, extent of RNA editing under normal physiological conditions and allele specific expression in naïve CD4+ T cells. In addition, we carried out a multi-omic comparative analysis of naïve with primary resting memory CD4+ T cells to identify molecular changes underlying T cell differentiation. This analysis provided mechanistic insights into how several molecules involved in T cell receptor signaling are regulated at the DNA, RNA and protein levels. Phosphoproteomics revealed downstream signaling events that regulate these two cellular states. Availability of multi-omics data from an identical genetic background also allowed us to employ novel proteogenomics approaches to identify individual-specific variants and putative novel protein coding regions in the human genome. CONCLUSIONS: We utilized multiple high-throughput technologies to derive a comprehensive profile of two primary human cell types, naïve CD4+ T cells and memory CD4+ T cells, from a single donor. Through vertical as well as horizontal integration of whole genome sequencing, methylation arrays, RNA-Seq, miRNA-Seq, proteomics, and phosphoproteomics, we derived an integrated and comparative map of these two closely related immune cells and identified potential molecular effectors of immune cell differentiation following antigen encounter.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Inmunidad Innata/fisiología , Modelos Biológicos , Metilación de ADN , Epigenómica , Perfilación de la Expresión Génica , Variación Genética , Genoma Humano , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunidad Innata/genética , Fosforilación , Proteómica , Edición de ARN/efectos de los fármacos , ARN Mensajero/metabolismo , Transducción de Señal/genética , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA